Next Article in Journal
Combination Treatment Targeting mTOR and MAPK Pathways Has Synergistic Activity in Multiple Myeloma
Next Article in Special Issue
Cyclooxygenase and Lipoxygenase Gene Expression in the Inflammogenesis of Colorectal Cancer: Correlated Expression of EGFR, JAK STAT and Src Genes, and a Natural Antisense Transcript, RP11-C67.2.2
Previous Article in Journal
Hashimoto’s Thyroiditis: A Protective Factor against Recurrence in BRAF-Wild Type Differentiated Thyroid Carcinoma
Previous Article in Special Issue
Potential Role of Non-Steroidal Anti-Inflammatory Drugs in Colorectal Cancer Chemoprevention for Inflammatory Bowel Disease: An Umbrella Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cooperation between Prostaglandin E2 and Epidermal Growth Factor Receptor in Cancer Progression: A Dual Target for Cancer Therapy

Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(8), 2374; https://doi.org/10.3390/cancers15082374
Submission received: 14 March 2023 / Revised: 15 April 2023 / Accepted: 17 April 2023 / Published: 19 April 2023

Abstract

:

Simple Summary

Inflammation is the biological response of the body to damaging and toxic stimuli, and is a positive event that evolves with the resolution of critical events (acute inflammation). However, when the process becomes chronic it acquires pathological characteristics, and is associated with detrimental diseases such as cancer. It is recognized that prostaglandin E2 (PGE2) is one key lipid mediator involved in chronic inflammation, and is directly implicated in tumor development by regulating cancer cell growth and migration, apoptosis, epithelial–mesenchymal transition, metastasis, angiogenesis, and immune escape. The enzymes’ expression in PGE2 synthesis positively correlates with tumor progression and aggressiveness. This review describes the interplay between the PGE2 cascade and epidermal growth factor receptor that fuel cancer progression, and new therapeutic strategies that target these signaling pathways, to outline the importance of the modulation of the inflammatory process in cancer fighting.

Abstract

It is recognized that prostaglandin E2 (PGE2) is one key lipid mediator involved in chronic inflammation, and it is directly implicated in tumor development by regulating cancer cell growth and migration, apoptosis, epithelial–mesenchymal transition, angiogenesis, and immune escape. In addition, the expression of the enzymes involved in PGE2 synthesis, cyclooxygenase 2 (COX-2) and microsomal prostaglandin E synthase 1 (mPGES1), positively correlates with tumor progression and aggressiveness, clearly indicating the crucial role of the entire pathway in cancer. Moreover, several lines of evidence suggest that the COX2/mPGES1/PGE2 inflammatory axis is involved in the modulation of epidermal growth factor receptor (EGFR) signaling to reinforce the oncogenic drive of EGFR activation. Similarly, EGFR activation promotes the induction of COX2/mPGES1 expression and PGE2 production. In this review, we describe the interplay between COX2/mPGES1/PGE2 and EGFR in cancer, and new therapeutic strategies that target this signaling pathway, to outline the importance of the modulation of the inflammatory process in cancer fighting.

1. Introduction

The inflammatory process is a complex biological response of the body to damaging and toxic stimuli, and is considered a positive event that evolves with the resolution of critical events (acute inflammation) [1]. However, when the process becomes chronic, it acquires pathological characteristics, and is associated with detrimental diseases grouped into the class of chronic inflammatory diseases. In addition, chronic inflammation is considered a critical factor able to promote cancer aggressiveness [2,3,4,5].
Cancer is one of the major causes of death in industrialized countries, and despite advancements in diagnosis and therapeutic approaches, remains a problem with great social and economic impact. Malignant transformation of normal cells may occur as a consequence of genetic and epigenetic alterations of cancer-related genes (oncogenes or oncosuppressor genes), and is associated with the disruption of key processes that are involved in the control of normal cell growth and tissue homeostasis. Other than genetic alterations of normal cells, the microenvironment surrounding transformed cells represents the drive of tumor development and progression [6,7]. Starting from Virchow in 1863, it became evident that inflammation, and mainly chronic inflammation, is associated with the majority of tumors, and supports their progression through the promotion of growth, migration, invasion, apoptotic escape, angiogenesis, and metabolic reprogramming of cancer cells [8]. It is accepted that microbial and viral infections, autoimmune diseases, and inflammatory conditions of different origin are triggers of chronic inflammation associated with cancer development. For example, Helicobacter pylori and the hepatitis C virus are associated with gastric cancer and hepatocellular carcinoma, respectively, inflammatory bowel disease is associated with colon cancer, and prostatitis may promote prostate cancer [3,5,8,9]. All these conditions are included in the definition of “extrinsic inflammation”, which includes all the events associated with an inflammatory milieu.
Moreover, the activation of pathways that promote the production of inflammatory mediators and the recruitment of inflammatory cells is also observed in tumors that are not directly related to surrounding inflammatory conditions [3,9]. It has been demonstrated that different genetic alterations of proto-oncogene (oncogene activation) may result in both cancer and inflammation. Abundant evidence has clearly demonstrated that the expression of the inflammation-related pathways is driven by the activation of different classes of oncogenes [2,3,8,9,10]. In this context, the definition of “intrinsic inflammation” describes the inflammatory process observed in cancer that is related to intrinsic characteristics of tumor cells and associated with the expression and induction of specific inflammatory pathways.
To further outline the fundamental role of inflammation in driving tumor progression, in 2011, Hanahan and Weinberg, who had previously published a famous manuscript that summarized six main common cancer hallmarks [11], expanded their considerations and inserted the inflammation process as an enabling characteristic of tumors [12], outlining how inflammation contributes to the appearance of multiple hallmark capabilities of cancer cells. The inflammatory milieu can promote cancer cell growth, migration, tumor angiogenesis, and metastasis by supplying bioactive molecules as growth factors, proangiogenic factors, extracellular-matrix-modifying enzymes, and proinflammatory mediators (such as cytokines, chemokines, and eicosanoids) [13,14,15,16]. The presence of inflammatory conditions in the tumor microenvironment drives tumor progression by activating different processes, such as the induction of genomic instability, altering gene expression as a consequence of epigenetic events, enhancing the proliferation and resistance to apoptosis of cancer cells, promoting cell motility, and inducing tumor angiogenesis and tissue remodeling, with the consequent promotion of tumor cell invasion and metastasis [3,9].
Among all the proinflammatory mediators, prostaglandins, mainly prostaglandin E2 (PGE2), appear to be a fundamental piece of the genesis and progression of cancer [17,18,19,20,21]. To corroborate the protumoral role of PGE2, several epidemiological studies have highlighted that the treatment with non-steroidal anti-inflammatory agents (NSAIDs), such as cyclooxygenase (COX) inhibitors, reduce the risk of developing certain cancers (i.e., colon and breast cancer) and the mortality related to these diseases [21].
Several molecular mechanisms have been proposed to justify the protumoral activity of PGE2. In vitro and in vivo studies demonstrate that PGE2 may induce epigenetic modifications that contribute to the growth and metastasis formation of breast and gastric cancer [22,23], promote miRNA modifications that contribute to modulate cancer cell growth and migration and tumor angiogenesis [24,25,26,27], and increases cancer cell growth, migration, and resistance to apoptosis through the modulation of several molecular pathways, such as JAK/STAT [28], PI3K/AKT [29], and RAS/Raf/MAPK signaling [30]. Moreover, PGE2 is involved in cancer immunomodulation, as previously reviewed [21].
In addition, a plethora of studies have reported the strong interplay between prostaglandins and receptors other than their own involved in tumor progression, outlining and suggesting the strong potential of inflammation in driving tumor aggressiveness [31,32,33,34].
In this review, we summarize the protumoral activity of PGE2 inflammatory pathways, mainly focusing on the interplay with the epidermal growth factor receptor (EGFR), and we show the recent advancement in the pharmacological modulation of these signaling pathways in cancer management.

2. Prostaglandin E2

PGE2 is now considered a promoter of tumor progression by boosting the appearance of several cancer hallmarks [21]. This notion has been established by experimental and epidemiological studies that reported increased expression of cyclooxygenase isoforms (COX-1 and COX-2) and microsomal prostaglandin E synthase (mPGES-1), as well as enhanced levels of PGE2, in several tumor types (colon, breast, prostate, and lung tumors) [17,18,19,21,35].
The biosynthesis of prostaglandins, i.e., the COX pathway, is an essential component of inflammatory responses, and may be commonly activated by several proinflammatory mediators, such as lipopolysaccharides (LPS) [36], cytokines [37], and xenobiotics [38,39]. PGs biosynthesis starts from arachidonic acid (AA), which is released by the action of various phospholipases, and is initially converted to an unstable prostaglandin G2 (PGG2), which is further reduced by the peroxidase activity of the same enzyme to a stable prostaglandin H2 (PGH2) (Figure 1). The enzymes that catalyze these steps are COX-1 and COX-2. COX-1 is constitutively expressed in many cell types, and is responsible for the generation of low levels of prostaglandins involved in body homeostasis. In contrast, COX-2 is not expressed in physiological conditions, but is induced by different stimuli, such as growth factors and cytokines [19,40,41].
Furthermore, the production of all PGs, such as prostaglandin D2 (PGD2), PGE2, prostaglandin F2α (PGF2α), prostacyclin (PGI2), and thromboxane A2 (TXA2), occurs by the action of different specific terminal synthases [42]. PGE2 is synthesized through the action of three different prostaglandin E2 synthases (PGESs). The PGES family includes cytosolic prostaglandin E synthase (cPGES) [43], microsomal PGES-1 (mPGES-1) [44], and microsomal PGES-2 (mPGES-2) [45], and each of them plays a different role in the synthesis of PGE2 (Figure 1). In cancer tissues, both COX-2 and mPGES-1 are overexpressed [46].
The biological actions of PGE2 are mediated by specific signaling pathways activated via various G-protein-coupled receptors, which include EP1, EP2, EP3, and EP4 receptors (Figure 2). The distribution and relative abundance of these receptors vary between different animal species and tissues [47]. Their signaling mechanism involves the G-protein-mediated activation of downstream targets via second messengers, such as cyclic AMP (cAMP), Ca2+, and inositol phosphates. More in detail, the activation of the EP1 receptor induces Ca2+ mobilization, whereas EP2 and EP4 receptor signaling is mediated by Gs protein and induces adenylyl cyclase (AC) activation and the increase in cAMP levels. EP3, which exists in multiple splicing isoforms, is coupled with Gi proteins, and inhibits AC, leading to a decrease in cAMP levels. Several in vitro and in vivo studies indicate that the specific activation of one of these receptors by PGE2 may promote cancer progression [48,49].
A high volume of data indicate that PGE2 promotes tumor progression, since both COX2 or mPGES1 upregulation, and/or high levels of PGE2, are linked to the development of several types of cancers [21,42,46,50,51].
In human cancer specimens, it has been observed that COX-2 and/or mPGES-1 are constitutively expressed in non-small cell lung cancer [52,53], colorectal cancer [54,55], breast cancer [56,57,58], prostate cancer [59,60], melanoma [61,62], and hepatocellular carcinoma [63,64], and contribute to tumor aggressiveness. Similar results have been reported in animal models, where overexpression of COX-2 and mPGES1 induces tumor formation, while its inhibition suppresses tumorigenesis or tumor progression [21]. For example, mPGES-1 is overexpressed in gastrointestinal-hamartoma-induced mice [65], increases the number of aberrant crypt foci at an early stage and increases tumor size in mice colorectal cancer [66], and promotes intestinal tumorigenesis in LPS-induced transgenic mice [67]; meanwhile, mPGES-1 deletion suppresses intestinal tumorigenesis in Apc(Min/+) mice [68]. Increased PGE2 synthesis was also reported in models of rat prostate cancers, and was linked to a worse prognosis [69]. The contribution of this signaling has been reported also in the immunogenic response to cancer, as extensively discussed in a previous review [21].
During cancer onset and progression, in addition to the dysregulation of the key enzymes involved in the biosynthesis of PGE2, it has been described that there is an aberrant expression and activation of EP receptors [47,50,70] (Figure 2). Extensive analysis of the role of EP receptors has been reported for colorectal cancer. In this type of tumor, the EP4 expression levels are upregulated during carcinogenesis, as reported in in vitro and in vivo models [71,72,73]. Watanabe et al. [74] demonstrated that the EP1 receptor is involved in the early stages of colon carcinogenesis, while EP3 receptor contributes to the later stages. However, Shoji et al. [75] showed that the expression levels of EP3 receptor are low in AOM-induced tumors, and that its deficiency increased the incidence and multiplicity of the tumor. Furthermore, it has been reported that the EP2 receptor plays a key role in tumorigenesis in the small intestine [76]. The loss of the EP2 receptor in ApcΔ716 compound mutant mice caused a reduction in the size and number of intestinal tumors. Furthermore, PGE2-EP2 receptor signaling promotes colon cancer cell migration, and amplifies the actions of COX-2 by increasing cAMP levels [77]. Considering the above-reported data, PGE2 is important in the development and progression of colorectal cancer. Still, there are several pieces of evidence that PGE2 may affect not only cancer cells, but also cells in the tumor microenvironment [21,78]. Moreover, in colorectal cancer, PGE2 is also involved in tumor immune evasion by suppressing the activity of immune cells (CD8+ T cells) and macrophages. PGE2 induces the expression of proteins involved in the suppression of the immune system, such as programmed death ligand 1 (PD-1), through the activation of the EP4 receptor, and by reducing the number of CD8+ T cells [79].
Several recent studies have focused on the potential role of EP receptor signaling in other types of cancers. For example, EP1, EP2, and EP4 receptor expression is increased while EP3 receptor expression is reduced in prostate cancer tissues, and EP1 receptor expression seems to be positively associated with tumor grade and TNM stage [80,81].
Moreover, recent studies suggest that the effects of PGE2 on tumor progression are also related to the effects of this prostaglandin on the tumor microenvironment, and actively trigger tumor immune evasion, influencing patient survival [21,82,83,84]. Furthermore, in vivo experiments on different tumor-based animal models indicate that the EP4 receptor may be involved in immune cell activity modulation during tumor growth [58,85,86,87,88,89]. However, the mechanisms by which the pharmacological modulation of EP4 signaling may be useful in cancer immunotherapy have not yet been clearly demonstrated.
In addition to the above-reported data, PGE2 may also elicit its protumoral effects through the activation of non-canonical pathways, in which the PGE2-mediated effects are linked to the activation of receptors different from its own. In the past few years, the interplay between PGE2 and several tyrosine kinase receptors has been extensively studied, with particular attention given to EGFR. Firstly, in 2002, it was demonstrated that PGE2 promotes EGFR phosphorylation and triggers the ERK2 signaling pathway in normal gastric epithelial and colon cancer cell lines. Inhibition of EGFR with selective inhibitors blocks PGE2-induced ERK2 activation, c-fos mRNA expression, and cell proliferation, indicating the importance of this signaling pathway in cell biology [33]. In 2003, Buchanan and co-workers demonstrated that PGE2 induces the migration and invasion of colorectal cancer cells through rapid transactivation and phosphorylation of EGFR [90]. They demonstrated that, in cellular models, PGE2-induced EGFR phosphorylation occurs through the activation of an intracellular pathway (Src-mediated activation) rather than through the release of an extracellular epidermal growth factor-like ligand. They also reported that EGFR transactivation was present in malignant human colorectal samples [90]. In the same year, Pai et al. showed that PGE2-increased colon cancer cell invasiveness is consequent to the activation of the EGFR-c-Met-R-β-catenin-uPAR signaling pathway [32], confirming the existence of PGE2 non-canonical pathways. Similarly, in squamous cell carcinoma, PGE2 promotes tumor cell growth and invasion by the activation of the EP2 receptor, which in turn promotes EGFR transactivation via protein kinase A (PKA) and cSrc activation [91]. In the following years, a high volume of scientific evidence suggests that TKR activation may represent an important oncogenic signal induced by PGE2. In fact, EGFR transactivation induced by PGE2 has been observed in many tumor types, such as endometrial adenocarcinomas [92], gastric cancer [93], and prostate cancer [94]. In addition, EGFR transactivation has been reported for EP1 [95], EP2 [92], and EP4 [96], increasing the complexity of EP receptor signaling pathways.
Moreover, PGE2-induced EGFR activation is not limited to intracellular phosphorylation of the receptor, but may also be due to the extracellular mobilization of EGFR ligands [96,97,98]. Oshima and co-workers demonstrated that, in mouse models of gastric cancer, the expression levels of EGFR ligands (i.e., epiregulin, amphiregulin, heparin-binding EGF-like growth factor, and betacellulin), and metalloproteinases are increased in a PGE2-pathway-dependent manner. Indeed, metalloproteases (MMPs) can activate EGFR by the ectodomain shedding of EGFR ligands, dependent on the EP4 receptor [96]. Similarly, it has been demonstrated that PGE2, through EP3 receptor activation, promotes EGFR phosphorylation and its nuclear translocation through the induction of EGFR ligands in lung cancer cell models [97]. A schematic representation of PGE2/EGFR crosstalk is reported in Figure 3.
In summary, a vast amount of experimental data confirm that PGE2 promotes EGFR activation, albeit via different mechanisms. Depending on experimental conditions and cellular models, there are two main mechanisms by which PGE2 may affect EGFR activity: (1) mobilization of intracellular pathways (such as Src or PKA) that promote direct intracellular tyrosine phosphorylation of EGFR; (2) PGE2-induced activation of intracellular signaling that activates metalloproteinases, which in turn induce EGFR activation after EGFR ligand shedding from the plasma membrane (Figure 3).
In addition, it is important to outline that the link between PGE2 and EGFR was also observed in cellular and in vivo models of cancers where endogenous PGE2 production is abrogated by COX2 or mPGES1 inhibition. Pharmacological inhibition or genetic suppression of mPGES1 inhibits EGFR phosphorylation both in human epidermoid carcinoma cells and in in vivo animal models of cancer, with a consequent reduction in tumor growth and inhibition of angiogenesis [99].
The promotion of the activity of oncogenes, such as EGFR, is very important in cancer cells, where TKR phosphorylation induces the activation of several signaling pathways, such as the MAPK, PI3K/Akt, STAT, and PLC signaling pathways, that lead to cell growth, differentiation, migration, and survival.

3. Epidermal Growth Factor Receptor, Cancer, and Intrinsic Inflammation

The ErbB family contains four proteins structurally and functionally related to the first discovered member EGFR (ErbB1), and involved in the pathogenesis and progression of numerous tumor types [100].
The ErbB proteins are normally expressed in different cell types, such as epithelial, mesenchymal, and neuronal cells, where they exert physiological roles. All four ErbB family members are tyrosine kinase receptors. They consist of an extracellular domain for ligand binding, a hydrophobic transmembrane region, and an intracellular region for signal transduction, with a conserved tyrosine kinase domain [100,101]. After EGFR activation by a specific ligand, multiple adaptors and signaling molecules are docked to the phosphorylated site and generate diverse intracellular responses. For example, the PI3K-Akt and RAS-ERK pathways [100,101].
EGFR overexpression or aberrant activation has been found in several tumors and drives tumor progression, inducing cancer cell growth, migration, invasion, and metastasis [102,103]. EGFR overexpression is an unfavorable prognostic marker in lung cancer [104], squamous cell carcinoma (SCC) [105], colorectal cancer [106,107], and others. Inhibition of EGFR activation through monoclonal antibodies (such as cetuximab and panitumumab) or small tyrosine kinase inhibitors (i.e., erlotinib and gefitinib, afatinib, and osimertinib) is an important pharmacological approach in several tumors, including those of the lung, pancreas, and colon, where the EGFR receptor is highly expressed, constitutively activated, or mutated [108,109,110].
Despite the initial favorable response to these treatments, most patients become resistant to the therapy [111]. Potential mechanisms involved in the appearance of resistance to EGFR inhibition include EGFR amplifications or mutations in the kinase domain [112,113]; constitutive activation of signaling pathways independent of EGFR, such as the coactivation of multiple TKRs [111]; promotion of the ERK5 pathway [114], or a pathway downstream of EGFR, such as the Ras/ERK1/2 and PI3K pathway [110,115,116]; and the activation of drug efflux mechanisms [113]. New studies are aimed at circumventing the resistance to EGFR inhibition by using combination therapies that engage EGFR inhibitors together with other treatment modalities targeting downstream EGFR signals.
In this scenario, extensive studies on the interplay between EGFR signaling and inflammation appear very important. Much evidence supports the idea that EGFR signaling and inflammation may be closely interconnected with each other. Both external inflammatory stimuli (extrinsic inflammation) and tumor-cell-derived inflammatory mediators (intrinsic inflammation) produced by tumor cells promote EGFR phosphorylation or the activation of EGFR molecular signaling.
For example, interleukin-1 beta (IL-1β), a well-known mediator of chronic inflammation, has been identified as a salivary biomarker for oral squamous cell carcinoma [117], and increased IL-1β levels have been related to the increased severity of oral malignant transformation in in vivo models [118]. In addition, oral squamous carcinoma cell lines secrete high levels of IL-1β, which promotes tumor growth in an autocrine manner (intrinsic inflammation). Lee and co-workers showed that IL-1β can modulate EGFR activation through IL-1β-dependent CXCL1 expression, which promotes carcinogenesis [119]. Similarly, it has been demonstrated that IL-1β promotes tissue factor (TF) production in adenocarcinoma cell lines through the induction of Src-mediated EGFR phosphorylation [120]. To outline the importance of intrinsic inflammation in EGFR activation, it has also been demonstrated that IL-1β promotes EGFR phosphorylation by increasing PGE2 levels through the induction of mPGES1 expression [99] (see Figure 3).
Similarly, leukotrienes, derived from the arachidonic acid cascade through the action of lipoxygenase (LOX), may play a role in cancer progression [121]. As for other inflammatory stimuli, the interplay between COX and LOX pathways has been described, and the two distinct eicosanoid groups are likely to be able to mutually foster each other’s effects [122,123]. Leukotrienes, particularly leukotriene B4 (LTB4) and leukotriene D4 (LTD4), have been reported to be highly involved in EGFR signaling, and some experimental data indicate that they are able to induce EGFR transactivation [124,125,126].
As described above, a particular loop has been reported between EGFR and the COX2/mPGES1/PGE2 axis. Both exogenous PGE2 (produced by the tumor microenvironment) and endogenous PGE2 (produced by the tumor cell itself) promote EGFR phosphorylation through direct or indirect mechanisms. On the other hand, EGFR activation alone promotes the upregulation of both COX2 and mPGES1 enzymes associated with increased PGE2 production and enhanced tumor aggressiveness [127,128,129]. In head and neck squamous cell carcinoma, EGF regulates metastasis through the induction of angiopoietin-like 4 (ANGPTL4), which was inhibited after genetic or pharmacological inhibition of COX-2 [130]. Similarly, COX2 regulates ovarian cancer cell invasion [131] and colorectal cancer cell migration induced by EGF [132].
In addition, PGE2-induced EGFR activation, through different EP receptors, may be responsible for the development of resistance to anti-EGFR therapies in some cancer patients [47].
On the other hand, it has been also demonstrated that in several tumor cell lines (SCC, colon, and lung), EGFR phosphorylation induces mPGES-1 upregulation through the activation of ERK1/2 and Egr-1 signaling, and that this enzyme drives the protumoral activity of EGFR [127]. In fact, in a vicious circle, the inhibition of the COX2/mPGES1/PGE2 pathway reduced the protumoral effects of EGFR activation [99]. In this light, dual inhibition of EGFR activation and PGE2 production may be a strategy to inhibit tumor progression and overcome EGFR therapy resistance.

4. PGE2 and EGFR as a Dual Target for Cancer Therapy

Several preclinical studies report the antitumoral activities of compounds, of natural or chemical origin, that act as dual inhibitors of EGFR and the COX2/mPGES1/PGE2 axis. For example, berberine, an isoquinoline alkaloid present in different species of plants, inhibits intestinal tumor development through the downregulation of Wnt, EGFR signaling pathways, and COX-2 expression [133]. Similarly, avenanthramides, isolated from Avena sativa, helioxantyne, from Taiwania cryptomerioides Hayata, and FAG, a 2-O-a-L-rhamnopyranosyl-hexacosanoate-b-D-glucopyranosyl ester from Ficus bengalensis, are natural EGFR inhibitors and reduce EGFR phosphorylation and COX-2 expression in tumoral cells and macrophages [128,134,135].
The efficacy of EGFR/COX-2 dual inhibition has been reported in several cancer cell models and animal models of different tumor types. In 2007, it has been reported that the use of NS-398, a selective COX-2 inhibitor, and AG1478, an EGFR inhibitor, reduced the migration of colorectal cancer cells. The authors suggested combining non-steroidal anti-inflammatory drugs with EGFR antagonists for future use in the clinic [136]. Due to these and similar observations, Qian et al. reported that, in in vitro and in vivo models of oral squamous cell carcinoma, the administration of low concentrations of cetuximab and celecoxib reduces the proliferation, migration, and invasion of cancer cells and decreases PEG2 levels. They showed that the combination of these drugs significantly induces apoptosis and reduces the phosphorylation of EGFR, PI3K, and Akt, which may contribute to the inhibition of tumor growth [137]. Li et al. demonstrated that a combination of erlotinib and celecoxib inhibits tumor growth of NSCLC (non-squamous cell lung carcinoma) in vitro and in vivo, with synergistic effects [138]. Importantly, they found that the combination of celecoxib and erlotinib leads to synergistic cell death only in EGFR-mutated cell lines harboring EGFR exon 19 deletions [138]. Similarly, it has been reported that, in human patients, high serum COX-2 levels may correlate with EGFR mutations, and that the efficacy of combined celecoxib and gefitinib is significantly greater in NSCLC cells with EGFR mutations than in wild-type NSCLC cells [139].
Recently, a similar approach has been proposed for prostate cancer, and it has been demonstrated that, in prostate cellular models, celecoxib reduces cell growth, induces apoptosis, and promotes EGFR degradation. In addition, celecoxib, in association with cetuximab, reduced the invasive phenotype of CRPC (castration-resistant prostate cancer) cells by modulating NF-kB activity and tumor growth in mice xenografts [140].
Despite encouraging preclinical data, clinical trials, in which the combination of COX-2 specific inhibitors (celecoxib and apricoxib) with EGFR inhibitors (erlotinib) was tested, have shown poor activity [141]. However, Gitlitz and co-workers reported that, in a selected patient population in which urinary prostaglandin M (PGE-M) was modulated in response to COX-2 inhibitor, the primary endpoint of the study was not met, with no difference between patients treated with apricoxib and erlotinib. Nevertheless, in a subset analysis of patients aged 65 years or younger, the combination of the drugs demonstrated a statistically significant benefit compared with placebo/erlotinib groups. [141]. The need for patient selection was also demonstrated by Reckamp and colleagues, who showed that the combination of erlotinib and celecoxib did not improve outcomes in an unselected population, but selection by elevated baseline PGEM led to an increase in progression-free survival (PFS) with the celecoxib combination. They also outlined that patients with EGFR wild-type status may benefit from the combination [142].
In a recent meta-analysis, the authors showed that celecoxib combined with palliative therapy is not able to improve patient survival or the local control of the tumor. However, they reported that EGFR wild-type patients had a prolonged PFS with celecoxib-combined therapy [143]. These observations suggest that despite the fact that initial clinical trials evaluating the efficacy of COX-2/EGFR dual inhibition were not successful, further studies are needed to take into account the differences in patient stratification (i.e., prostaglandin metabolite monitoring, EGFR status characterization).
Furthermore, cancer characterized by resistance to EGFR therapy may represent a possible application of EGFR/COX2 inhibition therapy. Xiao et al. demonstrated that COX2-TXA2 signaling prevents apoptosis and promotes gefitinib resistance in NSCLC. The combination of celecoxib, TOPK inhibitor pantoprazole, and gefitinib resulted in the ability to induce apoptosis in gefitinib-resistant cells and to inhibit tumor growth in vitro and in vivo [144]. Similarly, the clinical use of osimertinib, a third-generation irreversible EGFR inhibitor with important benefits for EGFR-mutated patients, may be limited by the acquisition of resistance. Han and colleagues demonstrated using in vitro and in vivo experiments that aspirin sensitizes osimertinib-resistant NSCLC cells to osimertinib by promoting apoptosis, providing evidence for the use of the combination of aspirin with osimertinib to overcome resistance in NSCLC patients [145]. The activity of aspirin in tumors resistant to targeted therapies has also been shown in other models, where it increased the sensitivity of resistant tumors to targeted drugs and significantly delayed the emergence of acquired resistance [146].

5. Conclusions

In summary, chronic inflammation and cancer appear as closely related diseases, and share important signaling pathways and molecules that act in synergy with each other. Inflammation has been recognized as an important hallmark of cancer, and ultimately serves to support tumor development. How and, primarily, why cancer onset and progression need to use inflammatory pathways is not understood, but it is now a fact.
Therefore, pharmacological approaches aimed to fight tumor progression through the inhibition of inflammation are under investigation, and strongly suggest that improving the combination of therapies that inhibit pathological inflammation and stimulate the antitumor response may be a successful strategy for the treatment of patients with cancer. Although the strong association between the COX-2/mPGES-1/PGE2 pathway and EGFR and its molecular signals in cancer is widely reported, more in-depth studies are necessary to better understand the physiopathological significance and the potential for cancer therapy.

Author Contributions

Conceptualization, F.F. and L.T; writing—original draft preparation, F.F., C.B., L.P. and M.P.; writing—review and editing, L.T. and F.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mollaei, M.; Abbasi, A.; Hassan, Z.M.; Pakravan, N. The Intrinsic and Extrinsic Elements Regulating Inflammation. Life Sci. 2020, 260, 118258. [Google Scholar] [CrossRef] [PubMed]
  2. Porta, C.; Larghi, P.; Rimoldi, M.; Grazia Totaro, M.; Allavena, P.; Mantovani, A.; Sica, A. Cellular and Molecular Pathways Linking Inflammation and Cancer. Immunobiology 2009, 214, 761–777. [Google Scholar] [CrossRef] [PubMed]
  3. Mantovani, A.; Pierotti, M.A. Cancer and Inflammation: A Complex Relationship. Cancer Lett. 2008, 267, 180–181. [Google Scholar] [CrossRef] [PubMed]
  4. Piotrowski, I.; Kulcenty, K.; Suchorska, W. Interplay between Inflammation and Cancer. Rep. Pract. Oncol. Radiother. 2020, 25, 422–427. [Google Scholar] [CrossRef]
  5. Korniluk, A.; Koper, O.; Kemona, H.; Dymicka-Piekarska, V. From Inflammation to Cancer. Ir. J. Med. Sci. 2017, 186, 57–62. [Google Scholar] [CrossRef]
  6. Herceg, Z.; Hainaut, P. Genetic and Epigenetic Alterations as Biomarkers for Cancer Detection, Diagnosis and Prognosis. Mol. Oncol. 2007, 1, 26–41. [Google Scholar] [CrossRef]
  7. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  8. Balkwill, F.; Mantovani, A. Inflammation and Cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
  9. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-Related Inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef]
  10. Dep Prete, A.; Allavena, P.; Santoro, G.; Fumarulo, R.; Corsi, M.M.; Mantovani, A. Inflammation: Molecular pathways in cancer-related inflammation. Biochem. Med. 2011, 21, 264–275. [Google Scholar] [CrossRef]
  11. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer: Review. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [PubMed]
  12. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  13. DeNardo, D.G.; Andreu, P.; Coussens, L.M. Interactions between Lymphocytes and Myeloid Cells Regulate Pro-versus Anti-Tumor Immunity. Cancer Metastasis Rev. 2010, 29, 309–316. [Google Scholar] [CrossRef]
  14. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, Inflammation, and Cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef]
  15. Qian, B.Z.; Pollard, J.W. Macrophage Diversity Enhances Tumor Progression and Metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [PubMed]
  16. Karnoub, A.E.; Dash, A.B.; Vo, A.P.; Sullivan, A.; Brooks, M.W.; Bell, G.W.; Richardson, A.L.; Polyak, K.; Tubo, R.; Weinberg, R.A. Mesenchymal Stem Cells within Tumour Stroma Promote Breast Cancer Metastasis. Nature 2007, 449, 557–563. [Google Scholar] [CrossRef]
  17. Wang, D.; Dubois, R.N. Role of Prostanoids in Gastrointestinal Cancer. J. Clin. Investig. 2018, 128, 2732–2742. [Google Scholar] [CrossRef]
  18. Wang, D.; Dubois, R.N. Prostaglandins and Cancer. Gut 2006, 55, 115–122. [Google Scholar] [CrossRef]
  19. Wang, D.; Dubois, R.N. Eicosanoids and Cancer. Nat. Rev. Cancer 2010, 10, 181–193. [Google Scholar] [CrossRef]
  20. Donnini, S.; Finetti, F.; Terzuoli, E.; Bazzani, L.; Ziche, M. Targeting PGE2 Signaling in Tumor Progression and Angiogenesis. Forum Immunopathol. Dis. Ther. 2014, 5, 223–232. [Google Scholar] [CrossRef]
  21. Finetti, F.; Travelli, C.; Ercoli, J.; Colombo, G.; Buoso, E.; Trabalzini, L. Prostaglandin E2 and Cancer: Insight into Tumor Progression and Immunity. Biology 2020, 9, 434. [Google Scholar] [CrossRef] [PubMed]
  22. Kang, W.; Xu, J.; Qian, Y.; Luk, S.T.Y.; Chen, H.; Li, W.; Zhao, L.; Zhang, X.; Chiu, P.W.Y.; Ng, E.K.W.; et al. Prostaglandin E2 Induces DNA Hypermethylation in Gastric Cancer in Vitro and in Vivo. Theranostics 2019, 9, 6256–6268. [Google Scholar] [CrossRef]
  23. So, J.Y.; Skrypek, N.; Yang, H.H.; Merchant, A.S.; Nelson, G.W.; Chen, W.D.; Ishii, H.; Chen, J.M.; Hu, G.; Achyut, B.R.; et al. Induction of DNMT3B by PGE2 and IL6 at Distant Metastatic Sites Promotes Epigenetic Modification and Breast Cancer Colonization. Cancer Res. 2020, 80, 2612–2627. [Google Scholar] [CrossRef] [PubMed]
  24. Terzuoli, E.; Donnini, S.; Finetti, F.; Nesi, G.; Villari, D.; Hanaka, H.; Radmark, O.; Giachetti, A.; Ziche, M. Linking Microsomal Prostaglandin E Synthase-1/PGE-2 Pathway with MiR-15a and -186 Expression: Novel Mechanism of VEGF Modulation in Prostate Cancer. Oncotarget 2016, 7, 44350–44364. [Google Scholar] [CrossRef]
  25. Park, Y.R.; Seo, S.Y.; Kim, S.L.; Zhu, S.M.; Chun, S.; Oh, J.M.; Lee, M.R.; Kim, S.H.; Kim, I.H.; Lee, S.O.; et al. MiRNA-206 Suppresses PGE2-Induced Colorectal Cancer Cell Proliferation, Migration, and Invasion by Targetting TM4SF1. Biosci. Rep. 2018, 38, BSR20180664. [Google Scholar] [CrossRef]
  26. Cen, B.; Lang, J.D.; Du, Y.; Wei, J.; Xiong, Y.; Bradley, N.; Wang, D.; DuBois, R.N. Prostaglandin E2 Induces MiR675-5p to Promote Colorectal Tumor Metastasis via Modulation of P53 Expression. Gastroenterology 2020, 158, 971–984.e10. [Google Scholar] [CrossRef]
  27. Li, P.; Shan, J.X.; Chen, X.H.; Zhang, D.; Su, L.P.; Huang, X.Y.; Yu, B.Q.; Zhi, Q.M.; Li, C.L.; Wang, Y.Q.; et al. Epigenetic Silencing of MicroRNA-149 in Cancer-Associated Fibroblasts Mediates Prostaglandin E2/Interleukin-6 Signaling in the Tumor Microenvironment. Cell Res. 2015, 25, 588–603. [Google Scholar] [CrossRef]
  28. Liu, X.; Ji, Q.; Ye, N.; Sui, H.; Zhou, L.; Zhu, H.; Fan, Z.; Cai, J.; Li, Q. Berberine Inhibits Invasion and Metastasis of Colorectal Cancer Cells via COX-2/PGE2 Mediated JAK2/STAT3 Signaling Pathway. PLoS ONE 2015, 10, e0123478. [Google Scholar] [CrossRef]
  29. Li, Y.; Qiu, M.; Li, B.; Xiao, J.; Yang, W.; Xie, S.; Du, Y.; Huang, K.; Nie, D. Growth of T-Cell Lymphoma Cells Is Inhibited by MPGES-1/PGE2 Suppression via JAK/STAT, TGF-β/Smad3 and PI3K/AKT Signal Pathways. Transl. Cancer Res. 2022, 11, 2175–2184. [Google Scholar] [CrossRef]
  30. Leone, V.; Di Palma, A.; Ricchi, P.; Acquaviva, F.; Giannouli, M.; Di Prisco, A.M.; Iuliano, F.; Acquaviva, A.M. 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, G673–G681. [Google Scholar] [CrossRef]
  31. Siegfried, J.M.; Gubish, C.T.; Rothstein, M.E.; De Oliveira, P.E.Q.; Stabile, L.P. Signaling Pathways Involved in Cyclooxygenase-2 Induction by Hepatocyte Growth Factor in Non-Small-Cell Lung Cancer. Mol. Pharmacol. 2007, 72, 769–779. [Google Scholar] [CrossRef] [PubMed]
  32. Pai, R.; Nakamura, T.; Moon, W.S.; Tarnawski, A.S. Prostaglandins Promote Colon Cancer Cell Invasion; Signaling by Cross-talk between Two Distinct Growth Factor Receptors. FASEB J. 2003, 17, 1640–1647. [Google Scholar] [CrossRef] [PubMed]
  33. Pai, R.; Soreghan, B.; Szabo, I.L.; Pavelka, M.; Baatar, D.; Tarnawski, A.S. Prostaglandin E2, Transactivates EGF Receptor: A Novel Mechanism for Promoting Colon Cancer Growth and Gastrointestinal Hypertrophy. Nat. Med. 2002, 8, 289–293. [Google Scholar] [CrossRef]
  34. Ye, Y.; Peng, L.; Vattai, A.; Deuster, E.; Kuhn, C.; Dannecker, C.; Mahner, S.; Jeschke, U.; von Schönfeldt, V.; Heidegger, H.H. Prostaglandin E2 Receptor 3 (EP3) Signaling Promotes Migration of Cervical Cancer via Urokinase-Type Plasminogen Activator Receptor (UPAR). J. Cancer Res. Clin. Oncol. 2020, 146, 2189–2203. [Google Scholar] [CrossRef]
  35. Gupta, R.A.; Dubois, R.N. Colorectal Cancer Prevention of Cyclooxygenase-2. Nat. Rev. Cancer 2001, 1, 11–21. [Google Scholar] [CrossRef] [PubMed]
  36. Inoue, H.; Tanabe, T.; Umesono, K. Feedback Control of Cyclooxygenase-2 Expression through PPARγ. J. Biol. Chem. 2000, 275, 28028–28032. [Google Scholar] [CrossRef] [PubMed]
  37. Newton, R.; Kuitert, L.M.E.; Bergmann, M.; Adcock, I.M.; Barnes, P.J. Evidence for Involvement of NF-ΚB in the Transcriptional Control of COX-2 Gene Expression by IL-1β. Biochem. Biophys. Res. Commun. 1997, 237, 28–32. [Google Scholar] [CrossRef]
  38. Olszowski, T.; Gutowska, I.; Baranowska-Bosiacka, I.; Piotrowska, K.; Korbecki, J.; Kurzawski, M.; Chlubek, D. The Effect of Cadmium on COX-1 and COX-2 Gene, Protein Expression, and Enzymatic Activity in THP-1 Macrophages. Biol. Trace Elem. Res. 2015, 165, 135–144. [Google Scholar] [CrossRef]
  39. Chen, R.; Zhao, L.D.; Liu, H.; Li, H.H.; Ren, C.; Zhang, P.; Guo, K.T.; Zhang, H.X.; Geng, D.Q.; Zhang, C.Y. Fluoride Induces Neuroinflammation and Alters Wnt Signaling Pathway in BV2 Microglial Cells. Inflammation 2017, 40, 1123–1130. [Google Scholar] [CrossRef]
  40. Dubois, R.N.; Abramson, S.B.; Crofford, L.; Gupta, R.A.; Simon, L.S.; van de Putte, L.B.A.; Lipsky, P.E. Cyclooxygenase in Biology and Disease. FASEB J. 1998, 12, 1063–1073. [Google Scholar] [CrossRef]
  41. Simmons, D.L.; Botting, R.M.; Hla, T. Cyclooxygenase Isozymes: The Biology of Prostaglandin Synthesis and Inhibition. Pharmacol. Rev. 2004, 56, 387–437. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, Y.Y.; da Yao, Y.; Luo, J.F.; Liu, Z.Q.; Huang, Y.M.; Wu, F.C.; Sun, Q.H.; Liu, J.X.; Zhou, H. Microsomal Prostaglandin E2 Synthase-1 and Its Inhibitors: Molecular Mechanisms and Therapeutic Significance. Pharmacol. Res. 2021, 175, 105977. [Google Scholar] [CrossRef] [PubMed]
  43. Tanioka, T.; Nakatani, Y.; Semmyo, N.; Murakami, M.; Kudo, I. Molecular Identification of Cytosolic Prostaglandin E2 Synthase That Is Functionally Coupled with Cyclooxygenase-1 in Immediate Prostaglandin E2 Biosynthesis. J. Biol. Chem. 2000, 275, 32775–32782. [Google Scholar] [CrossRef] [PubMed]
  44. Lazarus, M.; Kubata, B.K.; Eguchi, N.; Fujitani, Y.; Urade, Y.; Hayaishi, O. Biochemical Characterization of Mouse Microsomal Prostaglandin E Synthase-1 and Its Colocalization with Cyclooxygenase-2 in Peritoneal Macrophages. Arch. Biochem. Biophys. 2002, 397, 336–341. [Google Scholar] [CrossRef] [PubMed]
  45. Murakami, M.; Nakashima, K.; Kamei, D.; Masuda, S.; Ishikawa, Y.; Ishii, T.; Ohmiya, Y.; Watanabe, K.; Kudo, I. Cellular Prostaglandin E2 Production by Membrane-Bound Prostaglandin E Synthase-2 via Both Cyclooxygenases-1 and -2. J. Biol. Chem. 2003, 278, 37937–37947. [Google Scholar] [CrossRef] [PubMed]
  46. Nakanishi, M.; Rosenberg, D.W. Multifaceted Roles of PGE2 in Inflammation and Cancer. Semin. Immunopathol. 2013, 35, 123–137. [Google Scholar] [CrossRef]
  47. O’Callaghan, G.; Houston, A. Prostaglandin E2 and the EP Receptors in Malignancy: Possible Therapeutic Targets? Br. J. Pharmacol. 2015, 172, 5239–5250. [Google Scholar] [CrossRef]
  48. Sugimoto, Y.; Narumiya, S. Prostaglandin E Receptors. J. Biol. Chem. 2007, 282, 11613–11617. [Google Scholar] [CrossRef]
  49. Narumiya, S.; Sugimoto, Y.; Ushikubi, F. Prostanoid Receptors: Structures, Properties, and Functions. Physiol. Rev. 1999, 79, 1193–1226. [Google Scholar] [CrossRef]
  50. Ching, M.M.; Reader, J.; Fulton, A.M. Eicosanoids in Cancer: Prostaglandin E2 Receptor 4 in Cancer Therapeutics and Immunotherapy. Front. Pharmacol. 2020, 11, 819. [Google Scholar] [CrossRef]
  51. Tai, H.H. Prostaglandin Catabolic Enzymes as Tumor Suppressors. Cancer Metastasis Rev. 2011, 30, 409–417. [Google Scholar] [CrossRef] [PubMed]
  52. Fang, H.Y.; Lin, T.S.; Lin, J.P.; Wu, Y.C.; Chow, K.C.; Wang, L.S. Cyclooxygenase-2 in Human Non-Small Cell Lung Cancer. Eur. J. Surg. Oncol. 2003, 29, 171–177. [Google Scholar] [CrossRef]
  53. Yoshimatsu, K.; Subbaramaiah, K.; Dannenberg, A.J.; DeLellis, R.A.; Golijanin, D.; Paty, P.B.; Soslow, R.A.; Jakobsson, P.J. Inducible Microsomal Prostaglandin E Synthase Is Overexpressed in Colorectal Adenomas and Cancer. Clin. Cancer Res. 2001, 7, 3971–3976. [Google Scholar] [PubMed]
  54. Sano, H.; Kawahito, Y.; Wilder, R.L.; Hashiramoto, A.; Mukai, S.; Asai, K.; Kimura, S.; Kato, H.; Kondo, M.; Hla, T. Expression of Cyclooxygenà ¤ Se-1and -2 in Human Colorectal Cancer1. Cancer Res. 1995, 55, 3785–3790. [Google Scholar] [PubMed]
  55. Roelofs, H.M.J.; te Morsche, R.H.M.; van Heumen, B.W.H.; Nagengast, F.M.; Peters, W.H.M. Over-Expression of COX-2 MRNA in Colorectal Cancer. BMC Gastroenterol. 2014, 14, 3–8. [Google Scholar] [CrossRef]
  56. Half, E.; Tang, X.M.; Gwyn, K.; Sahin, A.; Wathen, K.; Sinicrope, F.A. Cyclooxygenase-2 Expression in Human Breast Cancers and Adjacent Ductal Carcinoma in Situ. Cancer Res. 2002, 62, 1676–1681. [Google Scholar]
  57. Mehrotra, S.; Morimiya, A.; Agarwal, B.; Konger, R.; Badve, S. Microsomal Prostaglandin E2 Synthase-1 in Breast Cancer: A Potential Target for Therapy. J. Pathol. 2006, 208, 356–363. [Google Scholar] [CrossRef]
  58. Majumder, M.; Nandi, P.; Omar, A.; Ugwuagbo, K.C.; Lala, P.K. EP4 as a Therapeutic Target for Aggressive Human Breast Cancer. Int. J. Mol. Sci. 2018, 19, 1019. [Google Scholar] [CrossRef]
  59. Khor, L.Y.; Bae, K.; Pollack, A.; Hammond, M.E.H.; Grignon, D.J.; Venkatesan, V.M.; Rosenthal, S.A.; Ritter, M.A.; Sandler, H.M.; Hanks, G.E.; et al. COX-2 Expression Predicts Prostate-Cancer Outcome: Analysis of Data from the RTOG 92-02 Trial. Lancet Oncol. 2007, 8, 912–920. [Google Scholar] [CrossRef]
  60. Finetti, F.; Terzuoli, E.; Giachetti, A.; Santi, R.; Villari, D.; Hanaka, H.; Radmark, O.; Ziche, M.; Donnini, S. MPGES-1 in Prostate Cancer Controls Stemness and Amplifies Epidermal Growth Factor Receptor-Driven Oncogenicity. Endocr. Relat. Cancer 2015, 22, 665–678. [Google Scholar] [CrossRef]
  61. Panza, E.; de Cicco, P.; Ercolano, G.; Armogida, C.; Scognamiglio, G.; Anniciello, A.M.; Botti, G.; Cirino, G.; Ianaro, A. Differential Expression of Cyclooxygenase-2 in Metastatic Melanoma Affects Progression Free Survival. Oncotarget 2016, 7, 57077–57085. [Google Scholar] [CrossRef] [PubMed]
  62. Ercoli, J.; Finetti, F.; Woodby, B.; Belmonte, G.; Miracco, C.; Valacchi, G.; Trabalzini, L. KRIT1 as a Possible New Player in Melanoma Aggressiveness. Arch. Biochem. Biophys. 2020, 691, 108483. [Google Scholar] [CrossRef] [PubMed]
  63. Koga, H.; Sakisaka, S.; Ohishi, M.; Kawaguchi, T.; Taniguchi, E.; Sasatomi, K.; Harada, M.; Kusaba, T.; Tanaka, M.; Kimura, R.; et al. Expression of Cyclooxygenase-2 in Human Hepatocellular Carcinoma: Relevance to Tumor Dedifferentiation. Hepatology 1999, 29, 688–696. [Google Scholar] [CrossRef] [PubMed]
  64. Zang, S.; Ni, M.; Lian, Y.; Zhang, Y.; Liu, J.; Huang, A. Expression of Microsomal Prostaglandin E2 Synthase-1 and Its Role in Human Hepatocellular Carcinoma. Hum. Pathol. 2013, 44, 1681–1687. [Google Scholar] [CrossRef]
  65. Takeda, H.; Miyoshi, H.; Tamai, Y.; Oshima, M.; Taketo, M.M. Simultaneous Expression of COX-2 and MPGES-1 in Mouse Gastrointestinal Hamartomas. Br. J. Cancer 2004, 90, 701–704. [Google Scholar] [CrossRef]
  66. Sasaki, Y.; Kamei, D.; Ishikawa, Y.; Ishii, T.; Uematsu, S.; Akira, S.; Murakami, M.; Hara, S. Microsomal Prostaglandin E Synthase-1 Is Involved in Multiple Steps of Colon Carcinogenesis. Oncogene 2012, 31, 2943–2952. [Google Scholar] [CrossRef]
  67. Oshima, H.; Oshima, M.; Inaba, K.; Taketo, M.M. Hyperplastic Gastric Tumors Induced by Activated Macrophages in COX-2/MPGES-1 Transgenic Mice. EMBO J. 2004, 23, 1669–1678. [Google Scholar] [CrossRef]
  68. Nakanishi, M.; Montrose, D.C.; Clark, P.; Nambiar, P.R.; Belinsky, G.S.; Claffey, K.P.; Xu, D.; Rosenberg, D.W. Genetic Deletion of MPGES-1 Suppresses Intestinal Tumorigenesis. Cancer Res. 2008, 68, 3251–3259. [Google Scholar] [CrossRef]
  69. Badawi, A.F.; Liu, Y.; Eldeen, M.B.; Morrow, W.; Razak, Z.R.; Maradeo, M.; Badr, M.Z. Age-Associated Changes in the Expression Pattern of Cyclooxygenase-2 and Related Apoptotic Markers in the Cancer Susceptible Region of Rat Prostate. Carcinogenesis 2004, 25, 1681–1688. [Google Scholar] [CrossRef]
  70. Sun, X.; Li, Q. Prostaglandin EP2 Receptor: Novel Therapeutic Target for Human Cancers (Review). Int. J. Mol. Med. 2018, 42, 1203–1214. [Google Scholar] [CrossRef]
  71. Mutoh, M.; Watanabe, K.; Kitamura, T.; Shoji, Y.; Takahashi, M.; Kawamori, T.; Sugimura, T.; Wakabayashi, K.; Tani, K.; Kobayashi, M.; et al. Involvement of Prostaglandin E Receptor Subtype EP4 in Colon Carcinogenesis. Cancer Res. 2002, 62, 28–32. [Google Scholar] [PubMed]
  72. Chell, S.D.; Witherden, I.R.; Dobson, R.R.; Moorghen, M.; Herman, A.A.; Qualtrough, D.; Williams, A.C.; Paraskeva, C. Increased EP4 Receptor Expression in Colorectal Cancer Progression Promotes Cell Growth and Anchorage Independence. Cancer Res. 2006, 66, 3106–3113. [Google Scholar] [CrossRef] [PubMed]
  73. Doherty, G.A.; Byrne, S.M.; Molloy, E.S.; Malhotra, V.; Austin, S.C.; Kay, E.W.; Murray, F.E.; Fitzgerald, D.J. Proneoplastic Effects of PGE2 Mediated by EP4 Receptor in Colorectal Cancer. BMC Cancer 2009, 9, 207. [Google Scholar] [CrossRef]
  74. Watanabe, K.; Kawamori, T.; Nakatsugi, S.; Ohta, T.; Ohuchida, S.; Yamamoto, H.; Maruyama, T.; Kondo, K.; Ushikubi, F.; Narumiya, S.; et al. Role of the Prostaglandin E Receptor Subtype EP1 in Colon Carcinogenesis. Cancer Res. 1999, 59, 5093–5096. [Google Scholar]
  75. Shoji, Y.; Takahashi, M.; Kitamura, T.; Watanabe, K.; Kawamori, T.; Maruyama, T.; Sugimoto, Y.; Negishi, M.; Narumiya, S.; Sugimura, T.; et al. Downregulation of Prostaglandin E Receptor Subtype EP3 during Colon Cancer Development. Gut 2004, 53, 1151–1158. [Google Scholar] [CrossRef] [PubMed]
  76. Sonoshita, M.; Takaku, K.; Sasaki, N.; Sugimoto, Y.; Ushikubi, F.; Narumiya, S.; Oshima, M.; Taketo, M.M. Acceleration of Intestinal Polyposis through Prostaglandin Receptor EP2 in ApcΔ716 Knockout Mice. Nat. Med. 2001, 7, 1048–1051. [Google Scholar] [CrossRef]
  77. Fujino, H.; Toyomura, K.; Chen, X.B.; Regan, J.W.; Murayama, T. Prostaglandin E2 Regulates Cellular Migration via Induction of Vascular Endothelial Growth Factor Receptor-1 in HCA-7 Human Colon Cancer Cells. Biochem. Pharmacol. 2011, 81, 379–387. [Google Scholar] [CrossRef]
  78. Mizuno, R.; Kawada, K.; Sakai, Y. Prostaglandin E2/EP Signaling in the Tumor Microenvironment of Colorectal Cancer. Int. J. Mol. Sci. 2019, 20, 6254. [Google Scholar] [CrossRef]
  79. Wei, J.; Zhang, J.; Wang, D.; Cen, B.; Lang, J.D.; DuBois, R.N. The COX-2–PGE2 Pathway Promotes Tumor Evasion in Colorectal Adenomas. Cancer Prev. Res. 2022, 15, 285–296. [Google Scholar] [CrossRef]
  80. Masato, M.; Miyata, Y.; Kurata, H.; Ito, H.; Mitsunari, K.; Asai, A.; Nakamura, Y.; Araki, K.; Mukae, Y.; Matsuda, T.; et al. Oral Administration of E-Type Prostanoid (EP) 1 Receptor Antagonist Suppresses Carcinogenesis and Development of Prostate Cancer via Upregulation of Apoptosis in an Animal Model. Sci. Rep. 2021, 11, 20279. [Google Scholar] [CrossRef]
  81. Miyata, Y.; Kanda, S.; Maruta, S.; Matsuo, T.; Sakai, H.; Hayashi, T.; Kanetake, H. Relationship between Prostaglandin E2 Receptors and Clinicopathologic Features in Human Prostate Cancer Tissue. Urology 2006, 68, 1360–1365. [Google Scholar] [CrossRef]
  82. Zelenay, S.; Reis e Sousa, C. Reducing Prostaglandin E2 Production to Raise Cancer Immunogenicity. Oncoimmunology 2016, 5, e1123370. [Google Scholar] [CrossRef]
  83. Böttcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of CDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037.e14. [Google Scholar] [CrossRef]
  84. Pelly, V.S.; Moeini, A.; Roelofsen, L.M.; Bonavita, E.; Bell, C.R.; Hutton, C.; Blanco-Gomez, A.; Banyard, A.; Bromley, C.P.; Flanagan, E.; et al. Anti-Inflammatory Drugs Remodel the Tumor Immune Environment to Enhance Immune Checkpoint Blockade Efficacy. Cancer Discov. 2021, 11, 2602–2619. [Google Scholar] [CrossRef]
  85. Oshima, H.; Hioki, K.; Popivanova, B.K.; Oguma, K.; van Rooijen, N.; Ishikawa, T.; Oshima, M. Prostaglandin E2 2 Signaling and Bacterial Infection Recruit Tumor-Promoting Macrophages to Mouse Gastric Tumors. Gastroenterology 2011, 140, 596–607.e7. [Google Scholar] [CrossRef]
  86. Fulton, A.M.; Ma, X.; Kundu, N. Targeting Prostaglandin E EP Receptors to Inhibit Metastasis. Cancer Res. 2006, 66, 9794–9797. [Google Scholar] [CrossRef]
  87. Majumder, M.; Xin, X.; Liu, L.; Girish, G.v.; Lala, P.K. 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, 1142–1151. [Google Scholar] [CrossRef]
  88. Albu, D.I.; Wang, Z.; Huang, K.C.; Wu, J.; Twine, N.; Leacu, S.; Ingersoll, C.; Parent, L.; Lee, W.; Liu, D.; et al. EP4 Antagonism by E7046 Diminishes Myeloid Immunosuppression and Synergizes with Treg-Reducing IL-2-Diphtheria Toxin Fusion Protein in Restoring Anti-Tumor Immunity. Oncoimmunology 2017, 6, e1338239. [Google Scholar] [CrossRef] [PubMed]
  89. Loo, T.M.; Kamachi, F.; Watanabe, Y.; Yoshimoto, S.; Kanda, H.; Arai, Y.; Nakajima-Takagi, Y.; Iwama, A.; Koga, T.; Sugimoto, Y.; et al. Gut Microbiota Promotes Obesity-Associated Liver Cancer through Pge2-Mediated Suppression of Antitumor Immunity. Cancer Discov. 2017, 7, 522–538. [Google Scholar] [CrossRef] [PubMed]
  90. Buchanan, F.G.; Wang, D.; Bargiacchi, F.; DuBois, R.N. Prostaglandin E2 Regulates Cell Migration via the Intracellular Activation of the Epidermal Growth Factor Receptor. J. Biol. Chem. 2003, 278, 35451–35457. [Google Scholar] [CrossRef] [PubMed]
  91. Donnini, S.; Finetti, F.; Solito, R.; Terzuoli, E.; Sacchetti, A.; Morbidelli, L.; Patrignani, P.; Ziche, M. EP2 Prostanoid Receptor Promotes Squamous Cell Carcinoma Growth through Epidermal Growth Factor Receptor Transactivation and INOS and ERK1/2 Pathways. FASEB J. 2007, 21, 2418–2430. [Google Scholar] [CrossRef] [PubMed]
  92. Sales, K.J.; Maudsley, S.; Jabbour, H.N. Elevated Prostaglandin EP2 Receptor in Endometrial Adenocarcinoma Cells Promotes Vascular Endothelial Growth Factor Expression via Cyclic 3′,5′-Adenosine Monophosphate-Mediated Transactivation of the Epidermal Growth Factor Receptor and Extracelluar Signal-Regulated Kinase 1/2 Signaling Pathways. Mol. Endocrinol. 2004, 18, 1533–1545. [Google Scholar] [CrossRef] [PubMed]
  93. Ding, Y.B.; Shi, R.H.; Tong, J.D.; Li, X.Y.; Zhang, G.X.; Xiao, W.M.; Yang, J.G.; Bao, Y.; Wu, J.; Yan, Z.G.; et al. PGE2 Up-Regulates Vascular Endothelial Growth Factor Expression in MKN28 Gastric Cancer Cells via Epidermal Growth Factor Receptor Signaling System. Exp. Oncol. 2005, 27, 108–113. [Google Scholar] [PubMed]
  94. Fernández-Martínez, A.B.; Lucio-Cazaña, J. Intracellular EP2 Prostanoid Receptor Promotes Cancer-Related Phenotypes in PC3 Cells. Cell. Mol. Life Sci. 2015, 72, 3355–3373. [Google Scholar] [CrossRef]
  95. Bai, X.; Wang, J.; Guo, Y.; Pan, J.; Yang, Q.; Zhang, M.; Li, H.; Zhang, L.; Ma, J.; Shi, F.; et al. Prostaglandin E2 Stimulates Β1-Integrin Expression in Hepatocellular Carcinoma through the EP1 Receptor/PKC/NF-ΚB Pathway. Sci. Rep. 2014, 4, 6538. [Google Scholar] [CrossRef]
  96. Oshima, H.; Popivanova, B.K.; Oguma, K.; Kong, D.; Ishikawa, T.O.; Oshima, M. Activation of Epidermal Growth Factor Receptor Signaling by the Prostaglandin E2 Receptor EP4 Pathway during Gastric Tumorigenesis. Cancer Sci. 2011, 102, 713–719. [Google Scholar] [CrossRef]
  97. Bazzani, L.; Donnini, S.; Finetti, F.; Christofori, G.; Ziche, M. PGE<inf>2</Inf>/EP3/SRC Signaling Induces EGFR Nuclear Translocation and Growth through EGFR Ligands Release in Lung Adenocarcinoma Cells. Oncotarget 2017, 8, 31270–31287. [Google Scholar] [CrossRef]
  98. Tveteraas, I.H.; Müller, K.M.; Aasrum, M.; Ødegård, J.; Dajani, O.; Guren, T.; Sandnes, D.; Christoffersen, T. Mechanisms Involved in PGE2-Induced Transactivation of the Epidermal Growth Factor Receptor in MH1C1 Hepatocarcinoma Cells. J. Exp. Clin. Cancer Res. 2012, 31, 72. [Google Scholar] [CrossRef]
  99. Finetti, F.; Terzuoli, E.; Bocci, E.; Coletta, I.; Polenzani, L.; Mangano, G.; Alisi, M.A.; Cazzolla, N.; Giachetti, A.; Ziche, M.; et al. Pharmacological Inhibition of Microsomal Prostaglandin E Synthase-1 Suppresses Epidermal Growth Factor Receptor-Mediated Tumor Growth and Angiogenesis. PLoS ONE 2012, 7, e40576. [Google Scholar] [CrossRef]
  100. Lemmon, M.A.; Schlessinger, J.; Ferguson, K.M. The EGFR Family: Not so Prototypical Receptor Tyrosine Kinases. Cold Spring Harb. Perspect. Biol. 2014, 6, a020768. [Google Scholar] [CrossRef]
  101. Roskoski, R. The ErbB/HER Family of Protein-Tyrosine Kinases and Cancer. Pharmacol. Res. 2014, 79, 34–74. [Google Scholar] [CrossRef] [PubMed]
  102. Arteaga, C.L.; Engelman, J.A. ERBB Receptors: From Oncogene Discovery to Basic Science to Mechanism-Based Cancer Therapeutics. Cancer Cell 2014, 25, 282–303. [Google Scholar] [CrossRef] [PubMed]
  103. Appert-Collin, A.; Hubert, P.; Crémel, G.; Bennasroune, A. Role of ErbB Receptors in Cancer Cell Migration and Invasion. Front. Pharmacol. 2015, 6, 283. [Google Scholar] [CrossRef] [PubMed]
  104. Zheng, H.; Saito, H.; Masuda, S.; Yang, X.; Takano, Y. Phosphorylated GSK3β-Ser9 and EGFR Are Good Prognostic Factors for Lung Carcinomas. Anticancer Res. 2007, 27, 3561–3569. [Google Scholar]
  105. Monteiro, L.S.; Diniz-Freitas, M.; Warnakulasuriya, S.; Garcia-Caballero, T.; Forteza, J.; Fraga, M. An Immunohistochemical Score to Predict the Outcome for Oral Squamous Cell Carcinoma. J. Oral Pathol. Med. 2018, 47, 375–381. [Google Scholar] [CrossRef] [PubMed]
  106. Punt, C.J.A.; Koopman, M.; Vermeulen, L. From Tumour Heterogeneity to Advances in Precision Treatment of Colorectal Cancer. Nat. Rev. Clin. Oncol. 2016, 14, 235–246. [Google Scholar] [CrossRef]
  107. Theodoropoulos, G.E.; Karafoka, E.; Papailiou, J.G.; Stamopoulos, P.; Zambirinis, C.P.; Bramis, K.; Panoussopoulos, S.G.; Leandros, E.; Bramis, J. P53 and EGFR Expression in Colorectal Cancer: A Reappraisal of “old” Tissue Markers in Patients with Long Follow-Up. Anticancer Res. 2009, 29, 785–791. [Google Scholar]
  108. Yarden, Y.; Pines, G. The ERBB Network: At Last, Cancer Therapy Meets Systems Biology. Nat. Rev. Cancer 2012, 12, 553–563. [Google Scholar] [CrossRef]
  109. Guardiola, S.; Varese, M.; Sánchez-Navarro, M.; Giralt, E. A Third Shot at EGFR: New Opportunities in Cancer Therapy. Trends Pharmacol. Sci. 2019, 40, 941–955. [Google Scholar] [CrossRef]
  110. Martinelli, E.; Ciardiello, D.; Martini, G.; Troiani, T.; Cardone, C.; Vitiello, P.P.; Normanno, N.; Rachiglio, A.M.; Maiello, E.; Latiano, T.; et al. Implementing Anti-Epidermal Growth Factor Receptor (EGFR) Therapy in Metastatic Colorectal Cancer: Challenges and Future Perspectives. Ann. Oncol. 2020, 31, 30–40. [Google Scholar] [CrossRef]
  111. Wykosky, J.; Fenton, T.; Furnari, F.; Cavenee, W.K. Therapeutic Targeting of Epidermal Growth Factor Receptor in Human Cancer: Successes and Limitations. Chin. J. Cancer 2011, 30, 5–12. [Google Scholar] [CrossRef] [PubMed]
  112. Hrustanovic, G.; Lee, B.J.; Bivona, T.G. Mechanisms of Resistance to EGFR Targeted Therapies. Cancer Biol. Ther. 2013, 14, 304–314. [Google Scholar] [CrossRef]
  113. Hopper-Borge, E.A.; Nasto, R.E.; Ratushny, V.; Weiner, L.M.; Golemis, E.A.; Astsaturov, I. Mechanisms of Tumor Resistance to EGFR-Targeted Therapies. Expert Opin. Ther. Targets 2009, 13, 339–362. [Google Scholar] [CrossRef]
  114. Zhao, W.; Yu, D.; Chen, Z.; Yao, W.; Yang, J.; Ramalingam, S.S.; Sun, S.Y. Inhibition of MEK5/ERK5 Signaling Overcomes Acquired Resistance to the Third Generation EGFR Inhibitor, Osimertinib, via Enhancing Bim-Dependent Apoptosis. Cancer Lett. 2021, 519, 141–149. [Google Scholar] [CrossRef]
  115. Sato, H.; Yamamoto, H.; Sakaguchi, M.; Shien, K.; Tomida, S.; Shien, T.; Ikeda, H.; Hatono, M.; Torigoe, H.; Namba, K.; et al. Combined Inhibition of MEK and PI3K Pathways Overcomes Acquired Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer. Cancer Sci. 2018, 109, 3183–3196. [Google Scholar] [CrossRef]
  116. Sforza, V.; Martinelli, E.; Ciardiello, F.; Gambardella, V.; Napolitano, S.; Martini, G.; Corte, C.D.; Cardone, C.; Ferrara, M.L.; Reginelli, A.; et al. Mechanisms of Resistance to Anti-Epidermal Growth Factor Receptor Inhibitors in Metastatic Colorectal Cancer. World J. Gastroenterol. 2016, 22, 6345–6361. [Google Scholar] [CrossRef]
  117. Li, Y.; St. John, M.A.R.; Zhou, X.; Kim, Y.; Sinha, U.; Jordan, R.C.K.; Eisele, D.; Abemayor, E.; Elashoff, D.; Park, N.H.; et al. Salivary Transcriptome Diagnostics for Oral Cancer Detection. Clin. Cancer Res. 2004, 10, 8442–8450. [Google Scholar] [CrossRef]
  118. Lee, C.-H.; Chang, J.S.-M.; Syu, S.-H.; Wong, T.-S.; Chan, J.Y.-W.; Tang, Y.-C.; Yang, Z.-P.; Yang, W.-C.; Chen, C.-T.; Lu, S.-C.; et al. IL-1β Promotes Malignant Transformation and Tumor Aggressiveness in Oral Cancer. J. Cell Physiol. 2015, 230, 875–884. [Google Scholar] [CrossRef]
  119. Lee, C.-H.; Syu, S.-H.; Liu, K.-J.; Chu, P.-Y.; Yang, W.-C.; Lin, P.; Shieh, W.-Y. Interleukin-1 Beta Transactivates Epidermal Growth Factor Receptor via the CXCL1-CXCR2 Axis in Oral Cancer. Oncotarget 2015, 6, 38866–38880. [Google Scholar] [CrossRef]
  120. Mechelke, T.; Wittig, F.; Ramer, R.; Hinz, B. Interleukin-1β Induces Tissue Factor Expression in A549 Cells via Egfr-Dependent and-Independent Mechanisms. Int. J. Mol. Sci. 2021, 22, 6606. [Google Scholar] [CrossRef]
  121. Murdocca, M.; De Masi, C.; Pucci, S.; Mango, R.; Novelli, G.; Di Natale, C.; Sangiuolo, F. LOX-1 and Cancer: An Indissoluble Liaison. Cancer Gene Ther. 2021, 28, 1088–1098. [Google Scholar] [CrossRef]
  122. Ohd, J.F.; Wikstrom, K.; Sjolander, A. Leukotrienes Induce Cell-Survival Signaling in Intestinal Epithelial Cells. Gastroenterology 2000, 119, 1007–1018. [Google Scholar] [CrossRef]
  123. Cabral, M.; Martín-Venegas, R.; Moreno, J.J. Leukotriene D4-Induced Caco-2 Cell Proliferation Is Mediated by Prostaglandin E2 Synthesis. Physiol. Rep. 2015, 3, e12417. [Google Scholar] [CrossRef]
  124. Dholia, N.; Yadav, U.C.S. Lipid Mediator Leukotriene D4-Induces Airway Epithelial Cells Proliferation through EGFR/ERK1/2 Pathway. Prostaglandins Other Lipid Mediat. 2018, 136, 55–63. [Google Scholar] [CrossRef]
  125. McGovern, T.; Risse, P.A.; Tsuchiya, K.; Hassan, M.; Frigola, G.; Martin, J.G. LTD4 Induces HB-EGF-Dependent CXCL8 Release through EGFR Activation in Human Bronchial Epithelial Cells. Am. J. Physiol. Cell. Mol. Physiol. 2010, 299, L808–L815. [Google Scholar] [CrossRef]
  126. Yang, C.C.; Chang, K.W. Eicosanoids and HB-EGF/EGFR in Cancer. Cancer Metastasis Rev. 2018, 37, 385–395. [Google Scholar] [CrossRef]
  127. Donnini, S.; Finetti, F.; Terzuoli, E.; Giachetti, A.; Ĩiguez, M.A.; Hanaka, H.; Fresno, M.; Rådmark, O.; Ziche, M. EGFR Signaling Upregulates Expression of Microsomal Prostaglandin e Synthase-1 in Cancer Cells Leading to Enhanced Tumorigenicity. Oncogene 2012, 31, 3457–3466. [Google Scholar] [CrossRef]
  128. Trabalzini, L.; Ercoli, J.; Trezza, A.; Schiavo, I.; Macr, G.; Moglia, A.; Spiga, O.; Finetti, F. Pharmacological and In Silico Analysis of Oat Avenanthramides as EGFR Inhibitors: Effects on EGF-Induced Lung Cancer Cell Growth and Migration. Int. J. Mol. Sci. 2022, 23, 8534. [Google Scholar] [CrossRef]
  129. Hsu, J.-Y.; Chang, K.-Y.; Chen, S.-H.; Lee, C.-T.; Chang, S.-T.; Cheng, H.-C.; Chang, W.-C.; Chen, B.-K. Epidermal Growth Factor-Induced Cyclooxygenase-2 Enhances Head and Neck Squamous Cell Carcinoma Metastasis through Fibronectin up-Regulation. Oncotarget 2015, 6, 1723. [Google Scholar] [CrossRef]
  130. Chiang, K.H.; Shieh, J.M.; Shen, C.J.; Chang, T.W.; Wu, P.T.; Hsu, J.Y.; Tsai, J.P.; Chang, W.C.; Chen, B.K. Epidermal Growth Factor-Induced COX-2 Regulates Metastasis of Head and Neck Squamous Cell Carcinoma through Upregulation of Angiopoietin-like 4. Cancer Sci. 2020, 111, 2004–2015. [Google Scholar] [CrossRef]
  131. Qiu, X.; Cheng, J.C.; Chang, H.M.; Leung, P.C.K. COX2 and PGE2 Mediate EGF-Induced E-Cadherin-Independent Human Ovarian Cancer Cell Invasion. Endocr. Relat. Cancer 2014, 21, 533–543. [Google Scholar] [CrossRef] [PubMed]
  132. Bocca, C.; Bozzo, F.; Miglietta, A. COX2 Inhibitor NS398 Reduces HT-29 Cell Invasiveness by Modulating Signaling Pathways Mediated by EGFR and HIF1-α. Anticancer Res. 2014, 34, 1793–1800. [Google Scholar] [PubMed]
  133. Cao, H.; Song, S.; Zhang, H.; Zhang, Y.; Qu, R.; Yang, B.; Jing, Y.; Hu, T.; Yan, F.; Wang, B. Chemopreventive Effects of Berberine on Intestinal Tumor Development in Apc Min/+ Mice. BMC Gastroenterol. 2013, 13, 163. [Google Scholar] [CrossRef] [PubMed]
  134. Alaaeldin, R.; Hassan, H.A.; Abdel-Rahman, I.M.; Mohyeldin, R.H.; Youssef, N.; Allam, A.E.; Abdelwahab, S.F.; Zhao, Q.L.; Fathy, M. A New EGFR Inhibitor from Ficus Benghalensis Exerted Potential Anti-Inflammatory Activity via Akt/PI3K Pathway Inhibition. Curr. Issues Mol. Biol. 2022, 44, 2967–2981. [Google Scholar] [CrossRef] [PubMed]
  135. Lin, Y.M.; Kuo, W.W.; Velmurugan, B.K.; Hsien, H.H.; Hsieh, Y.L.; Hsu, H.H.; Tu, C.C.; Bau, D.T.; Viswanadha, V.P.; Huang, C.Y. Helioxanthin Suppresses the Cross Talk of COX-2/PGE2 and EGFR/ERK Pathway to Inhibit Arecoline-Induced Oral Cancer Cell (T28) Proliferation and Blocks Tumor Growth in Xenografted Nude Mice. Environ. Toxicol. 2016, 31, 2045–2056. [Google Scholar] [CrossRef] [PubMed]
  136. Banu, N.; Buda, A.; Chell, S.; Elder, D.; Moorghen, M.; Paraskeva, C.; Qualtrough, D.; Pignatelli, M.; Pignatelli, M. Inhibition of COX-2 with NS-398 Decreases Colon Cancer Cell Motility through Blocking Epidermal Growth Factor Receptor Transactivation: Possibilities for Combination Therapy. Cell Prolif. 2007, 40, 768–779. [Google Scholar] [CrossRef] [PubMed]
  137. Qian, M.; Qian, D.; Jing, H.; Li, Y.; Ma, C.; Zhou, Y. Combined Cetuximab and Celecoxib Treatment Exhibits a Synergistic Anticancer Effect on Human Oral Squamous Cell Carcinoma in Vitro and in Vivo. Oncol. Rep. 2014, 32, 1681–1688. [Google Scholar] [CrossRef]
  138. Li, Y.X.; Wang, J.L.; Gao, M.; Tang, H.; Gui, R.; Fu, Y.F. Celecoxib-Erlotinib Combination Delays Growth and Inhibits Angiogenesis in EGFR-Mutated Lung Cancer. Am. J. Cancer Res. 2016, 6, 1494–1510. [Google Scholar]
  139. Li, N.; Li, H.; Su, F.; Li, J.; Ma, X.; Gong, P. Relationship between Epidermal Growth Factor Receptor (EGFR) Mutation and Serum Cyclooxygenase-2 Level, and the Synergistic Effect of Celecoxib and Gefitinib on EGFR Expression in Non-Small Cell Lung Cancer Cells. Int. J. Clin. Exp. Pathol. 2015, 8, 9010. [Google Scholar]
  140. Benelli, R.; Barboro, P.; Costa, D.; Astigiano, S.; Barbieri, O.; Capaia, M.; Poggi, A.; Ferrari, N. Multifocal Signal Modulation Therapy by Celecoxib: A Strategy for Managing Castration-Resistant Prostate Cancer. Int. J. Mol. Sci. 2019, 20, 6091. [Google Scholar] [CrossRef]
  141. Gitlitz, B.J.; Bernstein, E.; Santos, E.S.; Otterson, G.A.; Milne, G.; Syto, M.; Burrows, F.; Zaknoen, S. A Randomized, Placebo-Controlled, Multicenter, Biomarker-Selected, Phase 2 Study of Apricoxib in Combination with Erlotinib in Patients with Advanced Non-Small-Cell Lung Cancer. J. Thorac. Oncol. 2014, 9, 577–582. [Google Scholar] [CrossRef] [PubMed]
  142. Reckamp, K.L.; Koczywas, M.; Cristea, M.C.; Dowell, J.E.; Wang, H.J.; Gardner, B.K.; Milne, G.L.; Figlin, R.A.; Fishbein, M.C.; Elashoff, R.M.; et al. Randomized Phase 2 Trial of Erlotinib in Combination with High-Dose Celecoxib or Placebo in Patients with Advanced Non-Small Cell Lung Cancer. Cancer 2015, 121, 3298–3306. [Google Scholar] [CrossRef] [PubMed]
  143. Ye, S.Y.; Li, J.Y.; Li, T.H.; Song, Y.X.; Sun, J.X.; Chen, X.W.; Zhao, J.H.; Li, Y.; Wu, Z.H.; Gao, P.; et al. The Efficacy and Safety of Celecoxib in Addition to Standard Cancer Therapy: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Curr. Oncol. 2022, 29, 6137–6153. [Google Scholar] [CrossRef] [PubMed]
  144. Xiao, J.; Wang, F.; Lu, H.; Xu, S.; Zou, L.; Tian, Q.; Fu, Y.; Lin, X.; Liu, L.; Yuan, P.; et al. Targeting the COX2/MET/TOPK Signaling Axis Induces Apoptosis in Gefitinib-Resistant NSCLC Cells. Cell Death Dis. 2019, 10, 777. [Google Scholar] [CrossRef] [PubMed]
  145. Han, R.; Hao, S.; Lu, C.; Zhang, C.; Lin, C.; Li, L.; Wang, Y.; Hu, C.; He, Y. Aspirin Sensitizes Osimertinib-Resistant NSCLC Cells in Vitro and in Vivo via Bim-Dependent Apoptosis Induction. Mol. Oncol. 2020, 14, 1152–1169. [Google Scholar] [CrossRef]
  146. Li, L.; Hu, M.; Wang, T.; Chen, H.; Xu, L. Repositioning Aspirin to Treat Lung and Breast Cancers and Overcome Acquired Resistance to Targeted Therapy. Front Oncol. 2020, 9, 1503. [Google Scholar] [CrossRef]
Figure 1. Biosynthesis of prostaglandins. Arachidonic acid (AA) is a phospholipid present in cell membranes and is liberated from the cellular membranes by cytoplasmic phospholipase A2 (PLA2). Free AA is converted to PGE2 through the COX pathway. AA is metabolized to the intermediate prostaglandin G2 (PGG2), which is then reduced to PGH2 by the peroxidase activity of COX. PGH2 is sequentially metabolized to PGE2 or other eicosanoids by specific synthases.
Figure 1. Biosynthesis of prostaglandins. Arachidonic acid (AA) is a phospholipid present in cell membranes and is liberated from the cellular membranes by cytoplasmic phospholipase A2 (PLA2). Free AA is converted to PGE2 through the COX pathway. AA is metabolized to the intermediate prostaglandin G2 (PGG2), which is then reduced to PGH2 by the peroxidase activity of COX. PGH2 is sequentially metabolized to PGE2 or other eicosanoids by specific synthases.
Cancers 15 02374 g001
Figure 2. Schematic representation of EP receptors. PGE2 activity is mediated by the interaction with four G-protein-coupled receptors (GPCRs), namely, EP1–EP4 receptors. Each EP receptor possesses a distinct signaling pathway.
Figure 2. Schematic representation of EP receptors. PGE2 activity is mediated by the interaction with four G-protein-coupled receptors (GPCRs), namely, EP1–EP4 receptors. Each EP receptor possesses a distinct signaling pathway.
Cancers 15 02374 g002
Figure 3. Crosstalk between PGE2 and EGFR. PGE2 promotes EGFR phosphorylation and internalization through the activation of different signaling pathways. PGE2 promotes EGFR phosphorylation either by EP-mediated Src or PKA activation, or by inducing the release of EGFR ligands (MMPs activation). Similarly, IL-1β signaling promotes EGFR activation through the increased production of CXCL1, which in turn activates EGFR through CXCR2, or by a Src-mediated mechanism. AC = adenylate cyclase; PGE2 = prostaglandin E2; EP = prostaglandin E2 receptor; cAMP = cyclic adenosine monophosphate; PKA = protein kinase A; Src = SRC proto-oncogene; EGFR = epidermal growth factor receptor; nEGFR = nuclear EGFR; EGF = epidermal growth factor; PI3K = phosphatidylinositol 4,5-bisphosphate 3-kinase; p42/44 MAPK = p42/44 mitogen-activated protein kinases; TF = tissue factor; MMPs = matrix metalloproteinases; IL-1β = interleukin 1β; CXCR2 = CXC motif chemokine receptor 2; CXCL1 = C-X-C motif chemokine ligand 1; COX-2 = cyclooxygenase 2; mPGES1 = microsomal prostaglandin E synthase 1; iNOS = inducible nitric oxide synthase.
Figure 3. Crosstalk between PGE2 and EGFR. PGE2 promotes EGFR phosphorylation and internalization through the activation of different signaling pathways. PGE2 promotes EGFR phosphorylation either by EP-mediated Src or PKA activation, or by inducing the release of EGFR ligands (MMPs activation). Similarly, IL-1β signaling promotes EGFR activation through the increased production of CXCL1, which in turn activates EGFR through CXCR2, or by a Src-mediated mechanism. AC = adenylate cyclase; PGE2 = prostaglandin E2; EP = prostaglandin E2 receptor; cAMP = cyclic adenosine monophosphate; PKA = protein kinase A; Src = SRC proto-oncogene; EGFR = epidermal growth factor receptor; nEGFR = nuclear EGFR; EGF = epidermal growth factor; PI3K = phosphatidylinositol 4,5-bisphosphate 3-kinase; p42/44 MAPK = p42/44 mitogen-activated protein kinases; TF = tissue factor; MMPs = matrix metalloproteinases; IL-1β = interleukin 1β; CXCR2 = CXC motif chemokine receptor 2; CXCL1 = C-X-C motif chemokine ligand 1; COX-2 = cyclooxygenase 2; mPGES1 = microsomal prostaglandin E synthase 1; iNOS = inducible nitric oxide synthase.
Cancers 15 02374 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Finetti, F.; Paradisi, L.; Bernardi, C.; Pannini, M.; Trabalzini, L. Cooperation between Prostaglandin E2 and Epidermal Growth Factor Receptor in Cancer Progression: A Dual Target for Cancer Therapy. Cancers 2023, 15, 2374. https://doi.org/10.3390/cancers15082374

AMA Style

Finetti F, Paradisi L, Bernardi C, Pannini M, Trabalzini L. Cooperation between Prostaglandin E2 and Epidermal Growth Factor Receptor in Cancer Progression: A Dual Target for Cancer Therapy. Cancers. 2023; 15(8):2374. https://doi.org/10.3390/cancers15082374

Chicago/Turabian Style

Finetti, Federica, Lucrezia Paradisi, Clizia Bernardi, Margherita Pannini, and Lorenza Trabalzini. 2023. "Cooperation between Prostaglandin E2 and Epidermal Growth Factor Receptor in Cancer Progression: A Dual Target for Cancer Therapy" Cancers 15, no. 8: 2374. https://doi.org/10.3390/cancers15082374

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop