Open Access

Bile reflux and hypopharyngeal cancer (Review)

  • Authors:
    • Dimitra P. Vageli
    • Sotirios G. Doukas
    • Panagiotis G. Doukas
    • Benjamin L. Judson
  • View Affiliations

  • Published online on: September 23, 2021     https://doi.org/10.3892/or.2021.8195
  • Article Number: 244
  • Copyright: © Vageli et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Laryngopharyngeal reflux, a variant of gastroesophageal reflux disease, has been considered a risk factor in the development of hypopharyngeal cancer. Bile acids are frequently present in the gastroesophageal refluxate and their effect has been associated with inflammatory and neoplastic changes in the upper aerodigestive tract. Recent in vitro and in vivo studies have provided direct evidence of the role of acidic bile refluxate in hypopharyngeal carcinogenesis and documented the crucial role of NF‑κB as a key mediator of early oncogenic molecular events in this process and also suggested a contribution of STAT3. Acidic bile can cause premalignant changes and invasive squamous cell cancer in the affected hypopharynx accompanied by DNA damage, elevated p53 expression and oncogenic mRNA and microRNA alterations, previously linked to head and neck cancer. Weakly acidic bile can also increase the risk for hypopharyngeal carcinogenesis by inducing DNA damage, exerting anti‑apoptotic effects and causing precancerous lesions. The most important findings that strongly support bile reflux as an independent risk factor for hypopharyngeal cancer are presented in the current review and the underlying mechanisms are provided.

Introduction

Tobacco smoke, chronic alcohol use and infection with human papillomavirus type 16 (HPV-16) are known risk factors for laryngopharyngeal cancer (14); however, there is a growing interest in identifying other risk factors.

Laryngopharyngeal reflux (LPR), a variant of gastro-esophageal reflux disease (GERD), has also been considered a potential risk factor in the last decade that may exert a carcinogenic effect on the upper aerodigestive tract (511). Recent findings have clarified the role of bile reflux as an independent risk factor in hypopharyngeal carcinogenesis through preclinical and clinical models (1218), and a model of the molecular mechanism of the bile-induced tumorigenic effect has been proposed (1925). Bile is produced in the liver and stored in the gallbladder (26) and its primary role is to assist in lipid digestion and absorption. Regurgitation of duodenal bile contents into the stomach and esophagus is known as bile reflux. The present review provides the latest knowledge regarding the association of bile reflux with hypopharyngeal cancer.

Bile as a potential carcinogen

The caustic nature of bile has long been recognized (27). In ancient times, the medical theory that excess, deficiency or ectopic bile in the body may affect human health was first stated by the father of medicine, Hippocrates (27). This medical theory remained popular for centuries through the writings of Galen (129–201 AD) (28) but was decisively displaced newly published theories of cellular pathology by Virchow and Rather in 1858 (29). In the past century, in 1938-40, Cook (30) proposed the possible role of bile acids in cancer. Years later (1974–1993) several studies supported the role of bile acids as carcinogens causing gastrointestinal cancer (3134). In parallel, clinical studies provided the first evidence of mixed gastric and bile (duodenal) fluids in refluxate of patients with GERD (3537). During this period, Gotley et al (38) determined an increased amount of conjugated bile acids in 87% of aspirates using high-performance liquid chromatography, suggesting an association between bile and acid concentration and esophageal mucosal injury. Nehra et al (35), Kauer et al (39,40) and Domellof et al (41) also characterized the concentration and composition of bile fluid in aspirates of patients with GERD. Of note, Fein et al (42) were the first who provided solid evidence of bile fluid as an independent carcinogen in the gastrointestinal tract by using a rat model and demonstrating that bile acids are able to induce esophageal adenocarcinoma.

Until the present day, the clinical prevalence and magnitude of bile reflux have remained to be fully determined; however, there is growing evidence of bile contents in GERD refluxate (31,4345). According to Covington et al (46), bile-containing enterogastric reflux is much more common than previously assumed. The increased information linking LPR and inflammatory/neoplastic disease of the upper aerodigestive tract as well as the lack of evidence for the carcinogenic effect of bile-containing refluxate into laryngopharynx led to further investigation.

Considerable research efforts to explore the carcinogenic effect of bile in the upper aerodigestive tract were made, including clinical and experimental studies. Galli et al (5) and Sasaki et al (11) suggested that during LPR, bile fluid reaches the epithelium of the upper aerodigestive tract, which may contribute to the development of inflammatory and neoplastic events. Furthermore, Lewin et al (47) reported a close association between LPR and patients with premalignant lesions or early carcinomas of the larynx. It is worth mentioning that although there have been efforts to link the effect of other gastroesophageal refluxate contents, such as pepsin, with pre-neoplastic events in the larynx and pharynx, the conclusions have been divergent (4854). A recent study indicated a possible contributory effect of slightly acidic or neutral pepsin to the inflammatory and neoplastic effects of LPR (54), but there is still no direct evidence of carcinogenesis induced by pepsin.

At the beginning of 2016, a series of in vitro and in vivo experiments questioned the role of bile reflux in hypopharyngeal carcinogenesis (1214). A study by our group from 2016 (13) established a murine model of wild-type mice, Mus musculus (C57BL/6J) and provided the first evidence that bile acids may cause preneoplastic lesions in the hypopharyngeal mucosa (HM). Using this model and long-term exposure to bile acids, the progressive mutagenic effect of biliary refluxate causing invasive cancer was subsequently observed (15,16). These and other studies provided direct evidence that bile fluid is a carcinogen, capable of inducing hypopharyngeal squamous cell carcinoma (HSCC) (15,16). The significance of these findings was in line with clinical findings derived from bile reflux-related HSCC (18). Specifically, a clinical pilot study demonstrated a characteristic bile-related molecular phenotype that was similarly identified in bile-exposed murine HM, which clearly differed from adjacent non-pathologic tissue (18).

Bile refluxate composition and acidity as critical factors of neoplastic events

Bile refluxate composition

According to Nehra et al (35) and Kauer et al (39,40), the majority of bile acids in esophageal aspirates of patients with GERD are conjugated with taurine or glycine and may be sulfated. Specifically, glycine conjugates of cholic, deoxycholic and chenodeoxycholic acids are the predominant bile acids aspirated from the esophagus of patients with GERD (the ratio of glycine to taurine conjugates in normal human bile is 3:1). It has also been indicated that unconjugated secondary bile acids, such as deoxycholic acid (DCA), may be present in the esophageal refluxate, particularly in patients with erosive esophagitis and Barrett's esophagus (35,41,55). It is known that pH affects the solubility of each bile component in various manners. At acidic pH (≤4), the conjugated bile acids are more un-ionized and therefore capable of penetrating or interacting with the cell membrane (taurine conjugates: pKa=1.8–1.9; glycine conjugates: pKa=4.3–5.2) (56). At pH <3.0, bile salts tend to precipitate, whereas between pH 5.5 and 7.0, most conjugated primary bile acids are found to be ionized and therefore relatively inactive. However, unconjugated secondary bile acid DCA remains unionized (pKa 5.5–6.2) and may therefore exert its harmful effects, causing mucosal injury even at a less acidic pH (12,16,17,55). According to Stamp (57), duodenal fluid, at a less acidic pH, may also contribute to gastrointestinal tract tumorigenesis. Specifically, glycine conjugates may remain un-charged and therefore be harmful to the epithelial cells at a less acidic pH. Ireland et al (58) indicated that duodenal fluid significantly contributes to the carcinogenic potential of methyl-N-amyl nitrosamine, particularly at less acidic pH. In addition, as bile acids are natural detergents when in high concentration, they may interact with the cell membrane even at a neutral pH.

Bile refluxate concentration

Although the bile composition is a crucial factor of bile reflux-related tumorigenesis, the concentration of bile may also potentiate its oncogenic effect. This view may be supported by previous in vivo studies indicating that the application of bile reflux components, chenodeoxycholic acid or DCA, at pharmacologic concentrations, both at a neutral pH of 7.0, to murine laryngopharyngeal mucosa was able to cause early premalignant changes, such as hyperplasia and dysplasia, as well as marked activation of NF-κB and its related oncogenic molecular phenotype (13,14).

Bile refluxate acidity

It appears that acidity of LPR refluxate is a critical factor for bile to induce a harmful effect on laryngopharyngeal mucosa (39,59,60). In the clinic, intraesophageal pH monitoring has been used extensively to identify reflux episodes. Although pH varies during gastroesophageal reflux episodes, according to Ulualp et al (61), 24-h ambulatory pH monitoring in the pharynx of patients with reflux laryngitis confirmed that a drop below pH 4.0 is common and is considered diagnostic of a reflux event, suggesting that acid may contribute to duodenogastric-induced inflammatory and neoplastic events. Lillemoe et al (62) demonstrated the injurious effect of the various duodenal components on rabbit esophageal mucosa at strongly acidic pH, supporting a synergism between bile and HCl. A study by our group from 2019 (17) documented that the tumorigenic effect of bile on hypopharyngeal cells is pH-dependent. Other studies from our group (1315) also demonstrated that a strongly acidic pH (≤4.0) serves a critical role in the bile-induced tumorigenic effect in murine laryngopharyngeal mucosa. It was indicated that chronic intermittent exposure of murine HM to a mixture of bile salts at a strongly acidic pH of 3.0 was able to progressively induce premalignant changes, microinvasion and invasive squamous cell carcinoma, causing increased DNA damage and oncogenic molecular alterations (15). Specifically, it was demonstrated that histopathologic changes caused by acidic bile were accompanied by underlying molecular alterations, such as increased levels of i) oxidative DNA/RNA damage and double-strand break (DSB) markers, ii) p53 and cell proliferation markers (Ki67, cytokeratin 14, and p63), as well as iii) alterations of the expression of cell adhesion molecules, like E-Cadherin and β-catenin, and iv) activation of NF-κB and other cancer-related transcription factors, such as signal transducer and activator of transcription 3 (STAT3) (1315). However, hypopharyngeal cells or mucosa exposed to the same mixture of conjugated primary bile acids at neutral pH (7.0) produced hyperplastic or mild dysplastic changes and significantly less intense underlying molecular changes compared to acidic bile salts (12,13,17). In parallel, it was indicated that chronic exposure to acid alone or concentrated glucose was not able to produce any histological changes (13,15). The negative or reduced effect of acid alone and/or bile salts at neutral pH, compared to acidic bile salts, indicates that the latter is particularly injurious.

Primary and secondary bile acids in refluxate

It is clear that the presence of conjugated primary bile acids in a highly acidic refluxate exerts a tumorigenic potency on the long-term exposed upper aerodigestive tract. This theory may explain findings from our group (13,15) indicating that chronic local exposure of murine HM to a mixture of conjugated primary bile acids, at concentrations previously measured in patients with GERD (35,40,43,35) at a strongly acidic pH (≤4.0), is able to progressively cause precancerous lesions and invasive cancer. Specifically, as taurine conjugates are active at low pH (≤4.0), it appears that taurine-conjugated bile acids may be responsible for the tumorigenic effect of bile at lower pH (15,17). There is also recent in vitro evidence supporting that acidity (pH ≤4.0) and bile composition may have a role in the progression of HSCC (63).

The above observations strongly support that controlling the pH during reflux episodes may have a protective effect by reducing the risk of bile-induced hypopharyngeal cancer. However, there is epidemiologic evidence that numerous patients with refractory GERD may also experience symptoms at a weakly acidic pH of 5.5–6.0 (60,64). Since unconjugated DCA and glycine-conjugated bile acids may be partially active at a weakly acidic pH, it appears that as the pH grows less acidic, approaching pH 5.5, the partially activated primary bile acids and the activated DCA may exert their influence (17). A recent study by our group (16) supported that DCA and glycine-conjugated bile acids are potent activators of DNA damage and oncogenic pathways in HM in a weakly acidic environment. Previous findings have demonstrated a similar association between DCA and its tumorigenic activity in the esophagus and colon (31,65,66). Regarding the hypopharynx, it has been documented that bile at a weakly acidic pH (5.0–5.5) with or without DCA, similarly to a strongly acidic pH 3.0, is able to increase the risk of bile-related hypopharyngeal neoplasia by promoting premalignant lesions, DNA damage and oncogenic molecular alterations, compared to controls (16). Of note, it was indicated that long-term exposure to a weakly acidic control (pH 5.5) was not able to induce any histological changes (16). This observation strongly supports that the oncogenic properties of biliary esophageal reflux on laryngopharyngeal mucosa may not be fully modified when antacid therapy is applied.

Although further exploration with clinical evidence is expected to strengthen these previous preclinical observations, investigation of the mechanism by which bile refluxate exerts its oncogenic properties is expected to contribute not only to a better understanding of the pathophysiology of hypopharyngeal cancer but also to alternative therapeutic strategies for patients with refractory GERD, using specific inhibitors of relevant molecular pathways or bile receptors.

Key role of NF-κB in bile reflux-related hypopharyngeal carcinogenesis

Several epidemiologic studies have supported the role of LPR in the neoplasia of the upper aerodigestive tract (511,67,68). However, the exact mechanism of LPR-related laryngopharyngeal carcinogenesis has remained elusive and unexplored until the last decade (6971). Studies including that by Huo et al (66) indicated that exposure of esophageal cells to DCA produced elevated levels of NF-κB in vitro, suggesting the role of NF-κB as a key molecule in esophageal cancer (7275). Thus, these observations supported the hypothesis of a possible mechanistic role of NF-κB in cancer of extraesophageal sites, such as hypopharynx.

NF-κB is a transcriptional factor complex consisting of homo- and heterodimers of five members of the Rel family [RelA (p65), RelB, c-Rel, NF-κB1 (p50/p105) and NF-κB2 (p52/p100)] (76). The canonical pathway of NF-κB activation includes phosphorylation of IκB-α, which leads to nuclear translocation of the heterodimers p50/Rela or p50/cRel and consequent binding to the promoters of target genes and regulation of their expression. Constitutive activation of NF-κB has been observed in various cancer types, linking inflammation to the neoplastic transformation of the epithelium (7779). In the initiation and progression of head and neck squamous cell carcinoma (HNSCC), several oncogenic pathways have been identified. These commonly include epidermal growth factor receptor (EGFR)/Ras/RAF/MAPK, PI3K/Akt1/mTOR, IΚΚ/NF-κB, STAT3 and Wnt/β-catenin (8089). It has been indicated that HNSCC exhibits abundant NF-κB activation and several studies indicate that NF-κB is upregulated in premalignant lesions and invasive cancer (80,9094).

The role of NF-κB in acidic bile reflux-related laryngopharyngeal carcinogenesis was first demonstrated in the last decade through in vitro and in vivo experimental models (1225). A study by our group from 2017 (21) documented the key role of NF-κB in mediating acidic bile-induced oncogenic molecular events in human hypopharyngeal cells (HHCs). Subsequently in 2018, a study by our group (22) also demonstrated that NF-κB is a crucial factor in controlling the levels of small regulatory molecules, such as microRNA (miRNA/miR) markers, in HHCs. A series of in vitro and in vivo studies also suggested that NF-κB inhibition may prevent inflammatory and neoplastic events in HHCs and HM, including STAT3 activation and significant deregulations of several cancer-related genes and miRNA markers (19,2325). Other non-specific stress factors, such as highly concentrated glucose or acidic pepsin, were not capable of inducing activation of genes with oncogenic function previously linked to HNSCC in vitro (12,13,53).

Bile reflux-induced NF-κB-related mRNA oncogenic phenotype

There is evidence that numerous types of cancer arise from sites of chronic inflammation (95). Specifically, a wide array of chronic inflammatory conditions predisposes susceptible cells of epithelial origin to neoplastic transformation (carcinomas) as a multistep process (focal proliferation of dysplastic cells with potential progression to malignant carcinoma). An example includes reflux esophagitis that may lead to DNA damage, development of Barrett's esophagus and esophageal carcinoma (96). In certain cases, the progenitors of the inflammation are known, such as bacterial infections or gastric acids that have been associated with increased risk of adenocarcinoma of the stomach and esophagus, respectively (97,98). It may be assumed that bile acids interact directly with HM, as described in a paragraph below, but may also be the progenitors of an LPR-induced chronic inflammatory microenvironment associated with an increased risk of hypopharyngeal cancer. It has been indicated that a chronic inflammatory microenvironment or harmful stimuli are able to induce a constitutive activation of NF-κB, which may lead to a cascade of molecular alterations. Specifically, constitutive activation of NF-κB may lead to subsequent transcriptional activation of genes that are implicated in a variety of signaling pathways via aberrant overexpression of cytokines, transcription factors and growth factor receptors, such as TNF-α, TLR and EGFR (76,77,7989,93,94,99103).

Preclinical studies from our group (1225) documented that acidic bile is able to induce activation of NF-κB and significant overexpression of several cancer-related genes. Specifically, acidic bile was reported to induce significant transcriptional activation of anti-apoptotic BCL2 and other genes previously linked to HNSCC, such as STAT3, EGFR, WNT5A, TNF-α, ΔNp63, cREL, IL6, IL1β, AKT1 and PTGS2 (8294,102111) (Fig. 1). Furthermore, a clinical pilot study revealed that bile-related HSCC had significantly higher levels of NF-κB and differential expression of the above genes compared to the adjacent non-pathologic tissue or bile-negative HSCC, providing further evidence of the central role of NF-κB (18).

Targeting NF-κB negatively affects the NF-κB signaling pathway and has been indicated to be an encouraging strategy to improve anticancer therapies (112). Several pharmacologic and dietary inhibitors of NF-κB are considered promising thera-peutic options, demonstrating chemo-preventive or chemo-sensitizing properties in head and neck cancer (99,112). BAY 11-7082 [(E)-3-(4-methylphenylsulphonyl)-1-propenenitrile] is a reliable inhibitor of the NF-κB pathway that has been widely used in numerous studies exploring the effect of NF-κB (112,113). It has been suggested that BAY 11-7082 offers the most rapid and potent anti-tumor effect among other NF-κB inhibitors (112) and it may be used as a sensitizer for anti-cancer treatment (114,115). Furthermore, curcumin is a turmeric natural supplement with known antioxidant, anti-inflammatory and anti-cancer properties, previously demonstrated to have potential chemo-preventive effects in head and neck malignancies (116), by blocking NF-κB activation and halting the proliferation of cancer cells (117) due to its pleiotropic properties (118).

Both BAY 11-7082 and dietary curcumin have been used in in vitro and in vivo experimental studies to investigate the underlying mechanism of bile reflux-induced carcinogenic effect into the hypopharynx. Studies by our group (19,25) suggested that application of BAY 11-7082 effectively suppressed cell proliferation rates, the activation of NF-κB and related oncogenic mRNA profiles induced by acidic bile exposure. This oncogenic phenotype included the significant overexpression of anti-apoptotic BCL2 and other genes implicated in the initiation and progression of HNSCC, including TNF-α, EGFR, STAT3, ΔNp63, cREL, IL6, IL1β, AKT1, PTGS2 and WNT5A (8285,87,8994,102111) (Fig. 1). Parallel evidence that acidic bile stimulus is able to activate NF-κB and its related pathways in HHCs arose from the in vitro treatment with curcumin, which successfully blocked the transcriptional activity of NF-κB (20), similar to BAY 11-7082 (21,23). A study by our group from 2020 (24) documented the preventive and therapeutic properties of curcumin on murine HM against the acidic bile effect, thus shaping the future translational development of effective targeted therapies using topical non-pharmacologic inhibitors of NF-κB.

Strong evidence that NF-κB activation is able to influence the acidic bile-induced oncogenic mRNA profile inspired a further study on whether synchronizing its inhibition with acidic bile exposure is significant. Thus, a study by our group from 2019 (23) reported the temporal characteristics of NF-κB inhibition in blocking the acidic bile-induced oncogenic molecular events in HHCs. A series of studies also documented that topical application of BAY 11-7082 or curcumin to HM, either before, after or simultaneous to acidic bile exposure, successfully prevented or suppressed cell proliferation and NF-κB-related molecular events (19,24,25).

These results revealed that the upregulation of RELA, BCL2, STAT3, EGFR, WNT5A, TNF-α, IL6 and PTGS2 is directly promoted by acidic bile through NF-κB, shortly after its exposure (19,24,25) (Fig. 1), and strongly suggested that it may be clinically feasible to topically apply NF-κB inhibitors, without any precise synchronization with acidic bile exposure, to prevent acidic bile-induced oncogenic molecular changes.

Interactions between NF-κB activation and other factors

The application of NF-κB inhibitors also revealed important information about possible interactions between acidic bile-induced NF-κB activation and other central molecules in head and neck cancer (Fig. 1).

The NF-κB/STAT3 crosstalk has been indicated to be fundamental in inflammation-associated carcinogenesis in head and neck cancer (108,119,120). Application of NF-κB inhibitors successfully prevented the acidic bile-induced activation and nuclear translocation of STAT3, which is an important regulator of cell proliferation, and reduced the transcriptional levels of IL6 and STAT3, in treated hypopharyngeal cells (1925) (Fig. 1). These data strongly support the theory that the acidic bile-induced activation of IL-6/STAT3 is NF-κB-dependent (108) (in a paragraph below, the role of STAT3 in bile-induced carcinogenesis is discussed). In addition, prior findings implied strong interactions between NF-κB and STAT3 in acidic bile-exposed premalignant HM (13,14), further supporting the theory that the inflammatory response induced by acidic bile may increase the risk of laryngopharyngeal cancer.

EGFR is frequently overexpressed in HNSCC (121,122). Crosstalk between NF-κB and downstream pathways of EGFR has been observed (123,124). Although the exact role of EGFR in bile-related hypopharyngeal carcinogenesis has remained to be elucidated, the application of NF-κB inhibitors resulted in the successful suppression of acidic bile-induced overexpression of EGFR, supporting the interactions between NF-κB and EGFR pathways during this process (Fig. 1). In addition, as both STAT3 and EGFR are important contributors to HNSCC pathogenesis (80,121), the above observations further emphasized the requirement to develop a therapeutic strategy for targeting NF-κB in head and neck malignancies and particularly in bile reflux-related HSCC.

Furthermore, NF-κB inhibition had a strong effect in suppressing the acidic bile-induced overexpression of WNT5A (Fig. 1), a factor related to cancer-associated inflammation and epithelial-to-mesenchymal transition (111), indicating that NF-κB is able to mediate acidic bile-induced changes in hypopharyngeal cell-cell interactions.

In addition, COX-2 is regularly highly overexpressed during inflammatory and neoplastic processes (125) and is significantly overexpressed in acidic bile-exposed HM (24,25). In vivo application of NF-κB inhibitor significantly abrogated the acidic bile-induced overexpression of PTGS2 (24,25) (Fig. 1), further supporting the regulatory role of NF-κB in early inflammatory and cancer-related pathways, such as COX-2 (126).

In addition to the above, the PI3K/Akt pathway (127) is one of the most frequently activated pathways in head and neck cancer (128). The successful suppression of acidic bile-induced AKT1 overexpression using topical application of NF-κB inhibitor on murine HM suggested that NF-κB may mediate acidic bile-induced deregulations of PI3K/Akt downstream pathways (127) (Fig. 1).

In summary, the NF-κB pathway is a core central pathway that interacts with multiple upstream and downstream signaling pathways linked to the carcinogenic process. Using both a specific NF-κB inhibitor, such as BAY 11-7082, and a more pleiotropic NF-κB inhibitor, such as curcumin, it was documented that the acidic bile-induced deregulations of cancer-related genes or inflammatory factors are mediated by the NF-κB (Fig. 1). Furthermore, as curcumin is able to prevent the bile-related anti-apoptotic effect independently of the pH status, it may have an advantage over other NF-κB inhibitors. Of note, curcumin specifically reduced a lower percentage of analyzed NF-κB signaling genes compared to BAY 11-7082 (25 vs. 85%) (20,21). Thus, curcumin may confer a clinical advantage by preventing generalized suppression of NF-κB signaling, which is essential to the basic metabolic function of healthy mucosa and thereby reducing global toxicity (24,129).

Bile reflux-induced NF-κB-related miRNA oncogenic phenotype

miRNA molecules have also been important in both inflammation and cancer (130), modulating the expression of genes by causing target mRNA degradation or inhibiting their translation (131). The expression levels of certain miRNAs, such as ‘oncomiRs’ and ‘tumor suppressor’ miRNAs, have been indicated to be altered in tumor cells compared to normal cells (upregulated or downregulated), and capable of contributing to carcinogenesis, thereby demonstrating a significant regulatory role in the multistep process of cancer initiation and progression (132).

There is further evidence that miRNA markers, such as ‘oncomiR’ miR-21 and ‘tumor suppressor’ miR-375, have a crucial role in the initiation and progression of HNSCC (14,22,133,134). Arantes et al (135) reported the fundamental role of miR-21 as a biomarker in head and neck carcinogenesis, while miR-375 has been proposed as a predictive biomarker for early diagnosis in laryngeal cancer (136). In addition, interactions between NF-κB and miRNA markers, such as miR-21, miR-34a and miR-451a, have been importantly described by others in human cancer cells, including HNSCC (104,137,138). Specifically, a cluster of miRNA markers was reported to be associated with NF-κB that may be associated with the aggressive biological behavior of HNSCC (104).

Explorations by our group (14,15,19,14) revealed that acidic bile caused deregulations of the expression of oncogenic miRNA markers, previously associated with laryngopharyngeal cancer (133141). Specifically, the ‘oncomiRs’ miR-21, miR-192 and miR-155, and the ‘tumor suppressors’ miR-34a, miR-375, miR-451a, miR-99a and miR-504 were indicated to be significantly altered in exposed HHCs and murine laryngopharyngeal mucosa (14,15,19,14) (Fig. 2). Of note, results from our group (14,15) highlighted the role of miR-21 and miR-375 deregulations in acidic bile-related neoplasia.

In addition, findings from HSCC tumor specimens from patients with documented bile reflux supported its strong association with upregulation of ‘oncomiR’ miR-21 and downregulation of ‘tumor suppressors’ miR-34a and particularly of miR-375, along with strong positivity for NF-κB (18) (Fig. 2). Bile reflux-associated HSCC was also associated with a marked reduction of ‘tumor suppressors’ miR-489, miR-504 and miR-99a compared with their adjacent non-pathologic tissue (18), suggesting their involvement in the onset and progression of HSCC (142,143). Finally, bile exposure-associated HSCC exhibited differential expression of miR-489 and miR-504, and particularly of miR-375, compared to bile-negative HSCC, which had significantly lower NF-κB levels. A previous view by our group (18) suggested that these miRNA markers may have a distinct role in biliary reflux-associated hypopharyngeal cancer.

Application of BAY 11-7082 in HHCs or murine HM was proven to prevent miRNA deregulations caused by acidic bile, providing insight into the interactions of transcriptionally active NF-κB with cancer-related miRNA markers (19,22,23,25). Specifically, it was demonstrated that BAY 11-7082 is able to effectively reverse the acidic bile-induced downregulation of ‘tumor suppressor’ miR-451a and miR-99a, and upregulation of ‘oncomiRs’ miR-21, miR-155 and miR-192 (19,22,23,25) (Fig. 2), which are considered important markers for poor prognosis in head and neck cancer or linked to gastroesophageal reflux (22,135,144150). Other studies have also indicated that NF-κB has a direct regulatory effect on the expression of miR-21 and miR-155 through their binding promoters (151,152). Through in vitro and in vivo applications of BAY 11-7082 on hypopharyngeal cells and mucosa, respectively, a direct effect of acidic bile on the above miRNAs was demonstrated shortly after exposure (22,23,25). This observation proposed the use of these miRNAs as biomarkers of early neoplastic events in acidic bile-exposed HM, strongly supporting the role of NF-κB as a mediator in this process. In addition, topical in vivo application of BAY 11-7082 either before, after or simultaneous to acidic bile significantly inhibited the acidic bile-induced upregulation of ‘oncomiR’ miR-192 (19,25), previously associated with GERD (148), and downregulation of ‘tumor suppressor’ miR-504, a promising target for HSCC (142) (Fig. 2). This observation also suggested the utility of these miRNAs as biomarkers of early neoplastic events in acidic bile-exposed HM.

In summary, the above preclinical data provided evidence of the role of NF-κB as a regulator of miR-192, miR-21, miR-155, miR-451a, miR-375, miR-99a and miR-504 (Fig. 2), and proposed these miRNAs as potential therapeutic targets of bile-related mutagenic evolution in the HM.

Interactions between NF-κB-related mRNA and miRNA phenotypes

Previous studies suggested that interactions between miRNAs and mRNA molecules may be NF-κB-dependent during carcinogenesis (141,153,154). Rokavec et al (153) proposed that miRNA molecules, such as miR-34a, interact with STAT3 in an NF-κB-dependent manner. According to Tili et al (154), permanent upregulation of miR-155 may mediate a prolonged inflammatory reaction leading to cancer.

It has been proposed that acidic bile-induced NF-κB activation, BCL2 overexpression and significant alterations of oncogenic EGFR, STAT3, TNF-α, IL6, IL1β and WNT5A may directly or indirectly interact with cancer-related miRNA markers, such as ‘oncomiRs’ miR-21 and miR-155, as well as ‘tumor suppressors’ miR-34a, miR-375 and miR-451a (1416,141) (Fig. 3). These observations suggest that inflammatory episodes caused by acidic bile may be associated with downstream oncogenic pathways and may be effectively prevented by NF-κB inhibition.

NF-κB as a challenging target for cancer therapy

Recent findings documenting the crucial role of NF-κB in bile reflux-related hypopharyngeal carcinogenesis, pose a challenge for researchers and clinicians on how to identify patients who are more likely to benefit from NF-κB inhibition treatment. An NF-κB-related gene expression signature associated with bile reflux in patients with HSCC (Figs. 1 and 2) may provide a better prediction for inhibition selection and also allow the development of diagnostic and prognostic biomarkers of NF-κB inhibition response.

Although NF-κB targeted therapy has been already applied in clinical practice with promising results in anti-cancer therapy (129,155,156), including HNSCC treatment (157,158), there is an increasing effort in the pharmaceutical industry to develop advanced NF-κB inhibitors (129,159,160). In particular, research focuses on the identification of IKK/NF-κB inhibitors for targeted therapy that would prevent NF-κB activation without affecting other signaling pathways and selectively affect malignant cells rather than normal cells. As one of the major adverse effects of using NF-κB inhibitors as anticancer drugs is their ability to impair innate immunity when applied in excessive and prolonged periods (129,159161), research should also focus on both minimizing systemic toxicity and prevention of long-term immunosuppression. Thus, an ideal inhibition of NF-κB should be transient and reversible, as well as effective when combined with other anti-cancer treatments. Prior in vivo explorations strongly supported the effectiveness of models using intermittent and topical treatment as opposed to prolonged and systemic treatment. Specifically, the marked efficacy of short-term topical treatment with NF-κB inhibitors, such as curcumin (2 mg/kg/day) and BAY 11-7082 (6.25 mg/kg/day), in suppressing bile reflux-induced early preneoplastic changes in the hypopharynx (19,24,25), strongly supports the view that non-systemic and transient NF-κB inhibition may be clinically feasible in preventing bile-reflux-related oncogenic effects. This area of research is progressing rapidly; however, another adverse effect is of high importance prior to the targeting of IKK or NF-κB in the clinic. This refers to the enhanced production of IL-1β and related cytokines by inhibitors of NF-κB activation during bacterial infections (161), suggesting the short-term use of NF-κB inhibitors in combination with antibiotics.

Role of STAT3 in bile reflux-related hypopharyngeal carcinogenesis

The STAT3 oncogene (162) is a transcription factor central to head and neck cancer (119,120,122). In addition to the significant role of NF-κB, recent preliminary data from our team indicate the important role of STAT3 in bile-related hypopharyngeal carcinogenesis by promoting early oncogenic molecular events, including cancer-related inflammatory molecules IL6, TNF-α and RELA (p65).

Specifically, using three different inhibitors with each blocking a different step of STAT3 upstream signaling, such as nifuroxazide, SI3-201 and STA-21 (163165), preliminary data from our group were obtained regarding the mechanism of the effects of bile. In detail, acidic bile is able to induce constitutive activation of STAT3 that may not be exclusively dependent on JAK/STAT3 upstream signaling (165), but it may also be stimulated by alternative signaling, such as EGFR. Although targeting STAT3, either by its knockdown or its pharmacological inhibition, had a minimal effect on nuclear or total phosphorylated NF-κB (p65 S536) protein levels, it was observed to contribute, among others, to the transcriptional activation of NF-κB. As mentioned above, previous findings of NF-κB inhibition, using BAY 11-7082, had determined a role of NF-κB in acidic bile-induced activation of STAT3 (1925).

All of these observations suggest possible molecular crosstalk between the NF-κB and STAT3 transcription factor associated signaling pathways in bile reflux-related inflammation and tumorigenesis in the hypopharynx, as similarly proposed in HNSCC (108,119,120).

Bile-induced DNA damage

One of the principal questions regarding the effects of bile on cellular physiology was how bile refluxate induces DNA damage. Dvorak et al (59), suggested that bile at acidic pH may potentially induce DNA damage in esophageal cells, speculating that chronic exposure to bile acids at low pH may result in increased genomic instability, abnormal cell signaling and resistance to apoptosis. According to Goldman et al (166), bile in combination with acid, but not acid alone, immediately activates all three isoforms of nitric oxide (NO) synthase, a family of enzymes catalyzing the production of NO, which links chronic inflammatory diseases and reactive oxygen/nitrogen species (ROS/RNS) with cancer. They also indicated that bile in combination with acid increased intracellular acidification and DNA damage in esophageal cells, which may lead to mutations and cancer progression (166). Bernstein et al (31,167) proposed that de-conjugated secondary bile acids, such as DCA, are capable of inducing DNA damage, giving rise to cancer due to the accumulation of mutations. Specifically, DCA was determined to induce increased intracellular production of ROS/RNS, resulting in increased oxidative stress and DNA damage (64,168,169).

Recent studies by our group (15,16,19) documented that conjugated bile acids led to the upregulation of γ-H2AX (pS139). γ-H2AX is a consistent marker of DNA DSBs (170,171), which was profoundly increased in bile-treated hypopharyngeal cells or HM at acidic pH, compared to neutral pH, acid alone or neutral control conditions (15,19). Subsequently, it was documented that bile-treated HM at weakly acidic pH, with or without DCA, also induced DNA damage in exposed HM (16). Specifically, premalignant and malignant lesions caused by acidic bile demonstrated increased levels of nuclear γ-H2AX, as well as DNA/RNA oxidative damage (Fig. 4) (15,16).

According to previous findings, increased oxidative damage may result in high levels of ROS or DSBs incurring direct DNA damage (168,172,173), which may potentially lead to tumor-initiating mutations in head and neck cancer (174). All of these results advocate the theory that acidic bile-induced DNA damage may contribute to its mutated phenotype (Fig. 4). In addition, ROS is able to activate several cancer-associated signaling pathways, including NF-κB (164), concluding that acidic bile may contribute to evasion of apoptosis and/or proliferation of mutated hypopharyngeal epithelial cells, resulting in malignant lesions of HM.

In parallel, chronic exposure of murine hypopharyngeal epithelium to acidic bile was observed to induce a systematic release of inflammatory molecules, such as IL-6 and TNF-α, which are considered central to head and neck carcinogenesis (91). This systematic release of inflammatory molecules is able to maintain the constitutive release of other cancer-related cytokines in the microenvironment of the exposed epithelium. It is known that the chronic inflammatory microenvironment may lead to the production of activated inflammatory cells that may also serve as sources of ROS and reactive nitrogen intermediates (RNI), which are capable of inducing DNA damage and genomic instability and so promote mutations in neighboring epithelial cells (175,176). In addition, inflammatory cytokines contribute to increased intracellular ROS and RNI production in pre-malignant cells. In conclusion, the chronic inflammatory microenvironment caused by bile reflux may be one of the main factors in hypopharyngeal carcinogenesis.

Possible interactions of bile refluxate with hypopharyngeal mucosa

How acidic bile interacts with HM to exert its harmful effect, causing DNA damage and promoting NF-κB-related anti-apoptotic processes, leading to its malignant transformation, has remained to be fully elucidated. According to Li and Cao (177), bile acids may interact with membrane receptors, such as Takeda G-protein coupled receptor (TGR5). Their study also indicated that TGR5 is able to mediate bile reflux-induced DNA damage in esophageal cells (177). Another cell membrane receptor that is able to interact with bile acids is the sphingosine-1-phosphate receptor 1, known as S1PR1 or S1P1 (178,179). Both above-mentioned receptors were previously associated with lower esophageal cancer related to bile reflux (180,181). In particular, TGR5 was reported to be expressed in both adenocarcinoma and squamous cell carcinoma of the lower esophagus (181), while S1PR1 has been associated with squamous cell carcinoma of the head and neck (179).

Other studies provided evidence that bile acids are able to activate nuclear farnesoid X receptors (FXRs) (182,183), suggesting their contribution to pre-neoplastic changes (183) of the lower esophagus. Although several nuclear receptors (NRs) have been identified in the head and neck (184), an association between NRs and bile acids has not yet been described in HNSCC. Prior studies suggested a mutually antagonistic relationship between FXR and NF-κB activation (185,186) and proposed FXR receptors as useful targets for esophageal adenocarcinoma (182). However, the exact mechanism by which FXR affects the expression of proinflammatory molecules, such as NF-κB in either the lower or upper esophagus remains elusive and the role of FXR in bile reflux-related carcinogenesis deserves further exploration. Further investigation in the hypopharynx may identify specific receptors activated by acidic bile and clarify the role of NRs, such as FXR, and cell membrane receptors, such as TGR5 and S1PR1, in this process.

Conclusion

Recent in vitro and in vivo data provide evidence on bile reflux-associated hypopharyngeal carcinogenesis. The composition of biliary refluxate, such as conjugated bile acids, and acidity are pivotal factors in promoting DNA damage, as well as histologic and molecular changes in the HM, most likely through the constitutive activation of NF-κB. Chronic acidic bile exposure can cause increased oxidative damage, DSBs, overproduction of cytokines and cell-cell interaction changes, which are critical elements of tumor initiation and progression (187), possibly through derangements in both pre-neoplastic/neoplastic cells and their microenvironment. In parallel, acidic bile-induced constitutive activation of NF-κB can promote oncogenic mRNA and miRNA phenotypes, contributing to the proliferation of mutated cells and thus giving rise to the malignant transformation of the exposed HM. Primary data also support the contributing role of STAT3 in this process. Further investigation of the proposed mechanisms mediating bile-induced DNA damage, the tumor microenvironment and downstream oncogenic signaling pathways in HM, as well as the identification of specific receptors that may interact with bile, will contribute to innovative approaches to the diagnosis and prevention of laryngopharyngeal malignancies, as well as to the improvement of current therapeutic approaches to LPR-related carcinogenesis.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Data sharing is not applicable.

Authors' contributions

DPV and SGD were involved in the conceptualization of this review article. DPV, SGD, PGD and BLJ were involved in searching the literature. DPV, PGD and SGD were involved in the writing of the original draft. DPV, SGD, PGD and BLJ reviewed and edited the article. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

HM

hypopharyngeal mucosa

HHCs

human normal hypopharyngeal cells

References

1 

Hashibe M, Boffetta P, Zaridze D, Shangina O, Szeszeni-Dabrowska N, Mates D, Fabiánová E, Rudnai P and Brennan P: Contribution of tobacco and alcohol to the high rates of squamous cell carcinoma of the supraglottis and glottis in Central Europe. Am J Epidemiol. 165:814–820. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Curado MP and Hashibe M: Recent changes in the epidemiology of head and neck cancer. Curr Opin Oncol. 21:194–200. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Talamini R, Bosetti C, La Vecchia C, Dal Maso L, Levi F, Bidoli E, Negri E, Pasche C, Vaccarella S, Barzan L and Franceschi S: Combined effect of tobacco and alcohol on laryngeal cancer risk: A case-control study. Cancer Causes Control. 13:957–964. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Pöschl G and Seitz HK: Alcohol and cancer. Alcohol Alcohol. 39:155–165. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Galli J, Cammarota G, De Corso E, Agostino S, Cianci R, Almadori G and Paludetti G: Biliary laryngopharyngeal reflux: A new pathological entity. Curr Opin Otolaryngol Head Neck Surg. 14:128–132. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Tutar H, Erdamar H, Köybaşioğlu A, Dinç AE, Ceylan A and Uslu S: Can bile acids be an etiological factor for laryngeal carcinoma? ORL J Otorhinolaryngol Relat Spec. 73:156–161. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Geterud A, Bove M and Ruth M: Hypopharyngeal acid exposure: An independent risk factor for laryngeal cancer? Laryngoscope. 113:2201–2205. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Sereg-Bahar M, Jerin A and Hocevar-Boltezar I: Higher levels of total pepsin and bile acids in the saliva as a possible risk factor for early laryngeal cancer. Radiol Oncol. 49:59–64. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Altman KW, Prufer N and Vaezi MF: A review of clinical practice guidelines for reflux disease: toward creating a clinical protocol for the otolaryngologist. Laryngoscope. 121:717–723. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Assimakopoulos D and Patrikakos G: The role of gastroesophageal reflux in the pathogenesis of laryngeal carcinoma. Am J Otolaryngol. 23:351–357. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Sasaki CT, Marotta J, Hundal J, Chow J and Eisen RN: Bile-induced laryngitis: Is there a basis in evidence? Ann Otol Rhinol Laryngol. 114:192–197. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Sasaki CT, Issaeva N and Vageli DP: In vitro model for gastroduodenal reflux-induced nuclear factor-kappaB activation and its role in hypopharyngeal carcinogenesis. Head Neck. 38 (Suppl 1):E1381–E1391. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Vageli DP, Prasad ML and Sasaki CT: Gastro-duodenal fluid induced nuclear Factor-κappaB activation and early pre-malignant alterations in murine hypopharyngeal mucosa. Oncotarget. 7:5892–5908. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Sasaki CT and Vageli DP: MiR-21, miR-155, miR-192, and miR-375 deregulations related to NF-kappaB activation in gastroduodenal Fluid-Induced early Preneoplastic lesions of laryngeal mucosa in vivo. Neoplasia. 18:329–338. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Sasaki CT, Doukas SG, Costa J and Vageli DP: The progressive mutagenic effects of acidic bile refluxate in hypopharyngeal squamous cell carcinogenesis: New insights. Cancers (Basel). 12:10642020. View Article : Google Scholar : PubMed/NCBI

16 

Sasaki CT, Doukas SG, Doukas PG and Vageli DP: Weakly acidic bile is a risk factor for hypopharyngeal carcinogenesis evidenced by DNA damage, antiapoptotic function, and premalignant dysplastic lesions in vivo. Cancers (Basel). 13:8522021. View Article : Google Scholar : PubMed/NCBI

17 

Doukas SG, Cardoso B, Tower JI, Vageli DP and Sasaki CT: Biliary tumorigenic effect on hypopharyngeal cells is significantly enhanced by pH reduction. Cancer Med. 8:4417–4427. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Sasaki CT, Doukas SG, Costa J and Vageli DP: Biliary reflux as a causal factor in hypopharyngeal carcinoma: New clinical evidence and implications. Cancer. 125:3554–3565. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Sasaki CT, Doukas SG and Vageli DP: In Vivo Short-Term topical application of BAY 11-7082 prevents the acidic Bile-Induced mRNA and miRNA oncogenic phenotypes in exposed Murine Hypopharyngeal Mucosa. Neoplasia. 20:374–386. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Vageli DP, Doukas SG, Spock T and Sasaki CT: Curcumin prevents the bile reflux-induced NF-κB-related mRNA oncogenic phenotype, in human hypopharyngeal cells. J Cell Mol Med. 22:4209–4220. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Vageli DP, Doukas SG and Sasaki CT: Inhibition of NF-kappaB prevents the acidic bile-induced oncogenic mRNA phenotype, in human hypopharyngeal cells. Oncotarget. 9:5876–5891. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Doukas SG, Vageli DP and Sasaki CT: NF-κB inhibition reverses acidic bile-induced miR-21, miR-155, miR-192, miR-34a, miR-375 and miR-451a deregulations in human hypopharyngeal cells. J Cell Mol Med. 22:2922–2934. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Doukas PG, Vageli DP, Doukas SG and Sasaki CT: Temporal characteristics of NF-κB inhibition in blocking bile-induced oncogenic molecular events in hypopharyngeal cells. Oncotarget. 10:3339–3351. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Doukas SG, Doukas PG, Sasaki CT and Vageli D: The in vivo preventive and therapeutic properties of curcumin in bile reflux-related oncogenesis of the hypopharynx. J Cell Mol Med. 24:10311–10321. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Vageli DP, Kasle D, Doukas SG, Doukas PG and Sasaki CT: The temporal effects of topical NF-κB inhibition, in the in vivo prevention of bile-related oncogenic mRNA and miRNA phenotypes in murine hypopharyngeal mucosa: A preclinical model. Oncotarget. 11:3303–3314. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Hofmann AF: Chemistry and enterohepatic circulation of bile acids. Hepatology. 4 (Suppl 5):4S–14S. 1984. View Article : Google Scholar : PubMed/NCBI

27 

Greek Medicine, . http://www.greekmedicine.net/b_p/Four_Humors.html

28 

Rather LJ: Disturbance of function (functio laesa): The legendary fifth cardinal sign of inflammation, added by Galen to the four cardinal signs of Celsus. Bull NY Acad Med. 47:303–322. 1971.PubMed/NCBI

29 

Virchow R and Rather LJ: Disease, Life, and Man: Selected Essays. Stanford University Press; Stanford, CA: 1958

30 

Cook JW: Carcinogenic chemical agents. Yale J Biol Med. 11:1–13. 1938.PubMed/NCBI

31 

Bernstein H, Bernstein C, Payne CM, Dvorakova K and Garewal H: Bile acids as carcinogens in human gastrointestinal cancers. Mutat Res. 589:47–65. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Kuwahara A, Saito T and Kobayashi M: Bile acids promote carcinogenesis in the remnant stomach of rats. J Cancer Res Clin Oncol. 115:423–428. 1989. View Article : Google Scholar : PubMed/NCBI

33 

Hill MJ: Bile flow and colon cancer. Mutat Res. 238:313–320. 1990. View Article : Google Scholar : PubMed/NCBI

34 

Bayerdörffer E, Mannes GA, Ochsenkühn T, Dirschedl P, Wiebecke B and Paumgartner G: Unconjugated secondary bile acids in the serum of patients with colorectal adenomas. Gut. 36:268–273. 1995. View Article : Google Scholar : PubMed/NCBI

35 

Nehra D, Howell P, Williams CP, Pye JK and Beynon J: Toxic bile acids in gastro-oesophageal reflux disease: Influence of gastric acidity. Gut. 44:598–602. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Vaezi MF and Richter JE: Double reflux: Double trouble. Gut. 44:590–592. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Vaezi MF, Singh S and Richter JE: Role of acid and duodenogastric reflux in esophageal mucosal injury: A review of animal and human studies. Gastroenterology. 108:1897–1907. 1995. View Article : Google Scholar : PubMed/NCBI

38 

Gotley DC, Morgan AP and Cooper MJ: Bile acid concentrations in the refluxate of patients with reflux oesophagitis. Br J Surg. 75:587–590. 1988. View Article : Google Scholar : PubMed/NCBI

39 

Kauer WK, Peters JH, DeMeester TR, Ireland AP, Bremner CG and Hagen JA: Mixed reflux of gastric and duodenal juices is more harmful to the esophagus than gastric juice alone. The need for surgical therapy re-emphasized. Ann Surg. 222:525–531. 1995. View Article : Google Scholar : PubMed/NCBI

40 

Kauer WK, Peters JH, DeMeester TR, Feussner H, Ireland AP, Stein HJ and Siewert RJ: Composition and concentration of bile acid reflux into the esophagus of patients with gastroesophageal reflux disease. Surgery. 122:874–881. 1997. View Article : Google Scholar : PubMed/NCBI

41 

Domellof L, Reddy BS and Weisburger JH: Microflora and deconjugation of bile acids in alkaline reflux after partial gastrectomy. Am J Surg. 140:291–295. 1980. View Article : Google Scholar : PubMed/NCBI

42 

Fein M, Peters JH, Chandrasoma P, Ireland AP, Oberg S, Ritter MP, Bremner CG, Hagen JA and DeMeester TR: Duodenoesophageal reflux induces esophageal adenocarcinoma without exogenous carcinogen. J Gastrointest Surg. 2:260–268. 1998. View Article : Google Scholar : PubMed/NCBI

43 

McQuaid KR, Laine L, Fennerty MB, Souza R and Spechler SJ: Systematic review: The role of bile acids in the pathogenesis of gastro-oesophageal reflux disease and related neoplasia. Aliment Pharmacol Ther. 34:146–165. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Oh DS, Hagen JA, Fein M, Bremner CG, Dunst CM, Demeester SR, Lipham J and Demeester TR: The impact of reflux composition on mucosal injury and esophageal function. J Gastrointest Surg. 10:787–796. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Sweet MP, Patti MG, Hoopes C, Hays SR and Golden JA: Gastro-oesophageal reflux and aspiration in patients with advanced lung disease. Thorax. 64:167–173. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Covington MF, Krupinski E, Avery RJ and Kuo PH: Classification schema of symptomatic enterogastric reflux utilizing sincalide augmentation on hepatobiliary scintigraphy. J Nucl Med Technol. 42:198–202. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Lewin JS, Gillenwater AM, Garrett JD, Bishop-Leone JK, Nguyen DD, Callender DL, Ayers GD and Myers JN: Characterization of laryngopharyngeal reflux in patients with premalignant or early carcinomas of the larynx. Cancer. 97:1010–1014. 2003. View Article : Google Scholar : PubMed/NCBI

48 

Johnston N, Ondrey F, Rosen R, Hurley BP, Gould J, Allen J, DelGaudio J and Altman KW: Airway reflux. Ann N Y Acad Sci. 1381:5–13. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Adams J, Heintz P, Gross N, Andersen P, Everts E, Wax M and Cohen J: Acid/pepsin promotion of carcinogenesis in the hamster cheek pouch. Arch Otolaryngol Head Neck Surg. 126:405–409. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Johnston N, Dettmar PW, Ondrey FG, Nanchal R, Lee SH and Bock JM: Pepsin: Biomarker, mediator, and therapeutic target for reflux and aspiration. Ann NY Acad Sci. 1434:282–289. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Johnston N, Wells CW, Samuels TL and Blumin JH: Pepsin in nonacidic refluxate can damage hypopharyngeal epithelial cells. Ann Otol Rhinol Laryngol. 118:677–685. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Del Negro A, Araújo MR, Tincani AJ, Meirelles L, Martins AS and Andreollo NA: Experimental carcinogenesis on the oropharyngeal mucosa of rats with hydrochloric acid, sodium nitrate and pepsin. Acta Cir Bras. 23:337–342. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Sasaki CT, Toman J and Vageli D: The in vitro effect of Acidic-Pepsin on nuclear factor KappaB activation and its related oncogenic effect on normal human hypopharyngeal cells. PLoS One. 11:e01682692016. View Article : Google Scholar : PubMed/NCBI

54 

Doukas PG, Vageli DP, Sasaki CT and Judson BL: Pepsin promotes activation of epidermal growth factor receptor and downstream oncogenic pathways, at slightly acidic and neutral pH, in exposed hypopharyngeal cells. Int J Mol Sci. 22:42752021. View Article : Google Scholar : PubMed/NCBI

55 

Goldstein JL, Schlesinger PK, Mozwecz HL and Layden TJ: Esophageal mucosal resistance. A factor in esophagitis. Gastroenterol Clin North Am. 19:565–586. 1990. View Article : Google Scholar : PubMed/NCBI

56 

Stamp D and Jenkins G: An overview of bile-acid synthesis, chemistry and function. Bile Acids: Toxicology and Bioactivity. Jenkins GJ and Hardie L: Royal Society of Chemistry; Cambridge: 2008, Print: Issues in Toxicology; 4. https://pubs.rsc.org/en/content/ebook/978-0-85404-846-5 View Article : Google Scholar

57 

Stamp DH: Three hypotheses linking bile to carcinogenesis in the gastrointestinal tract: certain bile salts have properties that may be used to complement chemotherapy. Med Hypotheses. 59:398–405. 2002. View Article : Google Scholar : PubMed/NCBI

58 

Ireland AP, Peters JH, Smyrk TC, DeMeester TR, Clark GW, Mirvish SS and Adrian TE: Gastric juice protects against the development of esophageal adenocarcinoma in the rat. Ann Surg. 224:358–370. 1996. View Article : Google Scholar : PubMed/NCBI

59 

Dvorak K, Payne CM, Chavarria M, Ramsey L, Dvorakova B, Bernstein H, Holubec H, Sampliner RE, Guy N, Condon A, et al: Bile acids in combination with low pH induce oxidative stress and oxidative DNA damage: Relevance to the pathogenesis of Barrett's oesophagus. Gut. 56:763–771. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Kauer WK and Stein HJ: Role of acid and bile in the genesis of Barrett's esophagus. Chest Surg Clin N Am. 12:39–45. 2002. View Article : Google Scholar : PubMed/NCBI

61 

Ulualp SO, Roland PS, Toohill RJ and Shaker R: Prevalence of gastroesophagopharyngeal acid reflux events: An evidence-based systematic review. Am J Otolaryngol. 26:239–244. 2005. View Article : Google Scholar : PubMed/NCBI

62 

Lillemoe KD, Gadacz TR and Harmon JW: Bile absorption occurs during disruption of the esophageal mucosal barrier. J Surg Res. 35:57–62. 1983. View Article : Google Scholar : PubMed/NCBI

63 

Sasaki CT, Hajek M, Doukas SG and Vageli DP: The role of bile reflux and its related NF-κB activated pathway in progression of hypopharyngeal squamous cell cancer. Oral Oncol. 105:1046682020. View Article : Google Scholar : PubMed/NCBI

64 

Hemmink GJ, Bredenoord AJ, Weusten BL, Monkelbaan JF, Timmer R and Smout AJ: Esophageal pH-impedance monitoring in patients with therapy-resistant reflux symptoms: ‘On’ or ‘off’ proton pump inhibitor? Am J Gastroenterol. 103:2446–2453. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Bernstein H, Payne CM, Bernstein C, Schneider J, Beard SE and Crowley CL: Activation of the promoters of genes associated with DNA damage, oxidative stress, ER stress and protein malfolding by the bile salt, deoxycholate. Toxicol Lett. 108:37–46. 1999. View Article : Google Scholar : PubMed/NCBI

66 

Huo X, Juergens S, Zhang X, Rezaei D, Yu C, Strauch ED, Wang JY, Cheng E, Meyer F, Wang DH, et al: Deoxycholic acid causes DNA damage while inducing apoptotic resistance through NF-κB activation in benign Barrett's epithelial cells. Am J Physiol Gastrointest Liver Physiol. 301:G278–G286. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Langevin SM, Michaud DS, Marsit CJ, Nelson HH, Birnbaum AE, Eliot M, Christensen BC, McClean MD and Kelsey KT: Gastric reflux is an independent risk factor for laryngopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 22:1061–1068. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Coca-Pelaz A, Rodrigo JP, Takes RP, Silver CE, Paccagnella D, Rinaldo A, Hinni ML and Ferlito A: Relationship between reflux and laryngeal cancer. Head Neck. 35:1814–1818. 2013. View Article : Google Scholar : PubMed/NCBI

69 

Attwood SE, Smyrk TC, DeMeester TR, Mirvish SS, Stein HJ and Hinder RA: Duodenoesophageal reflux and the development of esophageal adenocarcinoma in rats. Surgery. 111:503–510. 1992.PubMed/NCBI

70 

Fein M, Fuchs KH, Stopper H, Diem S and Herderich M: Duodenogastric reflux and foregut carcinogenesis: Analysis of duodenal juice in a rodent model of cancer. Carcinogenesis. 21:2079–2084. 2000. View Article : Google Scholar : PubMed/NCBI

71 

Miwa K, Hattori T and Miyazaki I: Duodenogastric reflux and foregut carcinogenesis. Cancer. 75 (Suppl 6):S1426–S1432. 1995. View Article : Google Scholar

72 

Fang Y, Chen H, Hu Y, Djukic Z, Tevebaugh W, Shaheen NJ, Orlando RC, Hu J and Chen X: Gastroesophageal reflux activates the NF-κB pathway and impairs esophageal barrier function in mice. Am J Physiol Gastrointest Liver Physiol. 305:G58–G65. 2013. View Article : Google Scholar : PubMed/NCBI

73 

McAdam E, Haboubi HN, Griffiths AP, Baxter JN, Spencer-Harty S, Davies C and Jenkins GJ: Reflux composition influences the level of NF-κB activation and upstream kinase preference in oesophageal adenocarcinoma cells. Int J Cancer. 136:527–535. 2015.PubMed/NCBI

74 

Bus P, Siersema PD and van Baal JW: Cell culture models for studying the development of Barrett's esophagus: A systematic review. Cell Oncol (Dordr). 35:149–161. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Hormi-Carver K, Zhang X, Zhang HY, Whitehead RH, Terada LS, Spechler SJ and Souza RF: Unlike esophageal squamous cells, Barrett's epithelial cells resist apoptosis by activating the nuclear factor-κB pathway. Cancer Res. 69:672–677. 2009. View Article : Google Scholar : PubMed/NCBI

76 

Karin M: Nuclear factor-kappaB in cancer development and progression. Nature. 441:431–436. 2006. View Article : Google Scholar : PubMed/NCBI

77 

Wang H and Cho CH: Effect of NF-kappaB signaling on apoptosis in chronic inflammation-associated carcinogenesis. Curr Cancer Drug Targets. 10:593–599. 2010. View Article : Google Scholar : PubMed/NCBI

78 

DiDonato JA, Mercurio F and Karin M: NF-kappaB and the link between inflammation and cancer. Immunol Rev. 246:379–400. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Hoesel B and Schmid JA: The complexity of NF-kappaB signaling in inflammation and cancer. Mol Cancer. 12:862013. View Article : Google Scholar : PubMed/NCBI

80 

Nottingham LK, Yan CH, Yang X, Si H, Coupar J, Bian Y, Cheng TF, Allen C, Arun P, Gius D, et al: Aberrant IKKα and IKKβ cooperatively activate NF-κB and induce EGFR/AP1 signaling to promote survival and migration of head and neck cancer. Oncogene. 33:1135–1147. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Stadler ME, Patel MR, Couch ME and Hayes DN: Molecular biology of head and neck cancer: Risks and pathways. Hematol Oncol Clin North Am. 22:1099–1124. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Molinolo AA, Amornphimoltham P, Squarize CH, Castilho RM, Patel V and Gutkind JS: Dysregulated molecular networks in head and neck carcinogenesis. Oral Oncol. 45:324–334. 2009. View Article : Google Scholar : PubMed/NCBI

83 

King KE, Ponnamperuma RM, Allen C, Lu H, Duggal P, Chen Z, Van Waes C and Weinberg WC: The p53 homologue DeltaNp63alpha interacts with the nuclear factor-kappaB pathway to modulate epithelial cell growth. Cancer Res. 68:5122–5131. 2008. View Article : Google Scholar : PubMed/NCBI

84 

Jackson-Bernitsas DG, Ichikawa H, Takada Y, Myers JN, Lin XL, Darnay BG, Chaturvedi MM and Aggarwal BB: Evidence that TNF-TNFR1-TRADD-TRAF2-RIP-TAK1-IKK pathway mediates constitutive NF-kappaB activation and proliferation in human head and neck squamous cell carcinoma. Oncogene. 26:1385–1397. 2007. View Article : Google Scholar : PubMed/NCBI

85 

Dong J, Jimi E, Zeiss C, Hayden MS and Ghosh S: Constitutively active NF-kappaB triggers systemic TNFalpha-dependent inflammation and localized TNFalpha-independent inflammatory disease. Genes Dev. 24:1709–1717. 2010. View Article : Google Scholar : PubMed/NCBI

86 

Foxwell BM, Bondeson J, Brennan F and Feldmann M: Adenoviral transgene delivery provides an approach to identifying important molecular processes in inflammation: Evidence for heterogenecity in the requirement for NF-kappaB in tumour necrosis factor production. Ann Rheum Dis. 59 (Suppl 1):i54–i59. 2000. View Article : Google Scholar : PubMed/NCBI

87 

Guyer RA and Macara IG: Loss of the polarity protein PAR3 activates STAT3 signaling via an atypical protein kinase C (aPKC)/NF-κB/interleukin-6 (IL-6) axis in mouse mammary cells. J Biol Chem. 290:8457–8468. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Zhao Y, Zhang C, Huang Y, Yu Y, Li R, Li M, Liu N, Liu P and Qiao J: Up-regulated expression of WNT5a increases inflammation and oxidative stress via PI3K/AKT/NF-κB signaling in the granulosa cells of PCOS patients. J Clin Endocrinol Metab. 100:201–211. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Bo H, Zhang S, Gao L, Chen Y, Zhang J, Chang X and Zhu M: Upregulation of Wnt5a promotes epithelial-to-mesenchymal transition and metastasis of pancreatic cancer cells. BMC Cancer. 13:4962013. View Article : Google Scholar : PubMed/NCBI

90 

Klein JD and Grandis JR: The molecular pathogenesis of head and neck cancer. Cancer Biol Ther. 9:1–7. 2010. View Article : Google Scholar : PubMed/NCBI

91 

Allen CT, Ricker JL, Chen Z and Van Waes C: Role of activated nuclear factor-kappaB in the pathogenesis and therapy of squamous cell carcinoma of the head and neck. Head Neck. 29:959–971. 2007. View Article : Google Scholar : PubMed/NCBI

92 

Loercher A, Lee TL, Ricker JL, Howard A, Geoghegen J, Chen Z, Sunwoo JB, Sitcheran R, Chuang EY, Mitchell JB, et al: Nuclear factor-kappaB is an important modulator of the altered gene expression profile and malignant phenotype in squamous cell carcinoma. Cancer Res. 64:6511–6523. 2004. View Article : Google Scholar : PubMed/NCBI

93 

Chung CH, Parker JS, Ely K, Carter J, Yi Y, Murphy BA, Ang KK, El-Naggar AK, Zanation AM, Cmelak AJ, et al: Gene expression profiles identify epithelial-to-mesenchymal transition and activation of nuclear factor-kappaB signaling as characteristics of a high-risk head and neck squamous cell carcinoma. Cancer Res. 66:8210–8218. 2006. View Article : Google Scholar : PubMed/NCBI

94 

Lee TL, Yang XP, Yan B, Friedman J, Duggal P, Bagain L, Dong G, Yeh NT, Wang J, Zhou J, et al: A novel nuclear factor-kappaB gene signature is differentially expressed in head and neck squamous cell carcinomas in association with TP53 status. Clin Cancer Res. 13:5680–5691. 2007. View Article : Google Scholar : PubMed/NCBI

95 

Coussens LM and Werb Z: Inflammation and cancer. Nature. 420:860–867. 2002. View Article : Google Scholar : PubMed/NCBI

96 

Souza RF: From reflux esophagitis to esophageal adenocarcinoma. Dig Dis. 34:483–490. 2016. View Article : Google Scholar : PubMed/NCBI

97 

Wroblewski LE, Peek RM Jr and Wilson KT: Helicobacter pylori and gastric cancer: Factors that modulate disease risk. Clin Microbiol Rev. 23:713–739. 2010. View Article : Google Scholar : PubMed/NCBI

98 

Shaheen N and Ransohoff DF: Gastroesophageal reflux, Barrett esophagus, and esophageal cancer: Scientific review. JAMA. 287:1972–1981. 2002. View Article : Google Scholar : PubMed/NCBI

99 

Vander Broek R, Snow GE, Chen Z and Van Waes C: Chemoprevention of head and neck squamous cell carcinoma through inhibition of NF-kappaB signaling. Oral Oncol. 50:930–941. 2014. View Article : Google Scholar : PubMed/NCBI

100 

Baldwin AS Jr: The NF-kappa B and I kappa B proteins: New discoveries and insights. Annu Rev Immunol. 14:649–683. 1996. View Article : Google Scholar : PubMed/NCBI

101 

Lee TL, Yeh J, Friedman J, Yan B, Yang X, Yeh NT, Van Waes C and Chen Z: A signal network involving coactivated NF-kappaB and STAT3 and altered p53 modulates BAX/BCL-XL expression and promotes cell survival of head and neck squamous cell carcinomas. Int J Cancer. 122:1987–1998. 2008. View Article : Google Scholar : PubMed/NCBI

102 

Wheeler SE, Suzuki S, Thomas SM, Sen M, Leeman-Neill RJ, Chiosea SI, Kuan CT, Bigner DD, Gooding WE, Lai SY and Grandis JR: Epidermal growth factor receptor variant III mediates head and neck cancer cell invasion via STAT3 activation. Oncogene. 29:5135–5145. 2010. View Article : Google Scholar : PubMed/NCBI

103 

Yan B, Yang X, Lee TL, Friedman J, Tang J, Van Waes C and Chen Z: Genome-wide identification of novel expression signatures reveal distinct patterns and prevalence of binding motifs for p53, nuclear factor-kappaB and other signal transcription factors in head and neck squamous cell carcinoma. Genome Biol. 8:R782007. View Article : Google Scholar : PubMed/NCBI

104 

Yan B, Li H, Yang X, Shao J, Jang M, Guan D, Zou S, Van Waes C, Chen Z and Zhan M: Unraveling regulatory programs for NF-kappaB, p53 and microRNAs in head and neck squamous cell carcinoma. PLoS One. 8:e736562013. View Article : Google Scholar : PubMed/NCBI

105 

Sriuranpong V, Park JI, Amornphimoltham P, Patel V, Nelkin BD and Gutkind JS: Epidermal growth factor receptor-independent constitutive activation of STAT3 in head and neck squamous cell carcinoma is mediated by the autocrine/paracrine stimulation of the interleukin 6/gp130 cytokine system. Cancer Res. 63:2948–2956. 2003.PubMed/NCBI

106 

Yang X, Lu H, Yan B, Romano RA, Bian Y, Friedman J, Duggal P, Allen C, Chuang R, Ehsanian R, et al: ΔNp63 versatility regulates a Broad NF-κB gene program and promotes squamous epithelial proliferation, migration, and inflammation. Cancer Res. 71:3688–3700. 2011. View Article : Google Scholar : PubMed/NCBI

107 

Du J, Romano RA, Si H, Mattox A, Bian Y, Yang X, Sinha S, Van Waes C and Chen Z: Epidermal overexpression of transgenic ΔNp63 promotes type 2 immune and myeloid inflammatory responses and hyperplasia via NF-κB activation. J Pathol. 232:356–368. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Squarize CH, Castilho RM, Sriuranpong V, Pinto DS Jr and Gutkind JS: Molecular cross-talk between the NFkappaB and STAT3 signaling pathways in head and neck squamous cell carcinoma. Neoplasia. 8:733–746. 2006. View Article : Google Scholar : PubMed/NCBI

109 

Yap LF, Ahmad M, Zabidi MM, Chu TL, Chai SJ, Lee HM, Lim PV, Wei W, Dawson C, Teo SH, et al: Oncogenic effects of WNT5A in Epstein-Barr virus associated nasopharyngeal carcinoma. Int J Oncol. 44:1774–1780. 2014. View Article : Google Scholar : PubMed/NCBI

110 

Sherwood V, Chaurasiya SK, Ekström EJ, Guilmain W, Liu Q, Koeck T, Brown K, Hansson K, Agnarsdóttir M, Bergqvist M, et al: WNT5A-mediated β-catenin-independent signalling is a novel regulator of cancer cell metabolism. Carcinogenesis. 35:784–794. 2014. View Article : Google Scholar : PubMed/NCBI

111 

Asem MS, Buechler S, Wates RB, Miller DL and Stack MS: Wnt5a Signaling in Cancer. Cancers (Basel). 8:792016. View Article : Google Scholar : PubMed/NCBI

112 

Nakanishi C and Toi M: Nuclear factor-kappaB inhibitors as sensitizers to anticancer drugs. Nat Rev Cancer. 5:297–309. 2005. View Article : Google Scholar : PubMed/NCBI

113 

Meng Z, Lou S, Tan J, Xu K, Jia Q and Zheng W: Nuclear factor-kappa B inhibition can enhance apoptosis of differentiated thyroid cancer cells induced by 131I. PLoS One. 7:e335972012. View Article : Google Scholar : PubMed/NCBI

114 

Lin Y, Bai L, Chen W and Xu S: The NF-κB activation pathways, emerging molecular targets for cancer prevention and therapy. Expert Opin Ther Targets. 14:45–55. 2010. View Article : Google Scholar : PubMed/NCBI

115 

Van Waes C: Nuclear factor-kappaB in development, prevention, and therapy of cancer. Clin Cancer Res. 13:1076–1082. 2007. View Article : Google Scholar : PubMed/NCBI

116 

Wilken R, Veena MS, Wang MB and Srivatsan ES: Curcumin: A review of anti-cancer properties and therapeutic activity in head and neck squamous cell carcinoma. Mol Cancer. 10:122011. View Article : Google Scholar : PubMed/NCBI

117 

LoTempio MM, Veena MS, Steele HL, Ramamurthy B, Ramalingam TS, Cohen AN, Chakrabarti R, Srivatsan ES and Wang MB: Curcumin suppresses growth of head and neck squamous cell carcinoma. Clin Cancer Res. 11:6994–7002. 2005. View Article : Google Scholar : PubMed/NCBI

118 

Naksuriya O, Okonogi S, Schiffelers RM and Hennink WE: Curcumin nanoformulations: A review of pharmaceutical properties and preclinical studies and clinical data related to cancer treatment. Biomaterials. 35:3365–3383. 2014. View Article : Google Scholar : PubMed/NCBI

119 

Fan Y, Mao R and Yang J: NF-κB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell. 4:176–185. 2013. View Article : Google Scholar : PubMed/NCBI

120 

Gaykalova DA, Manola JB, Ozawa H, Zizkova V, Morton K, Bishop JA, Sharma R, Zhang C, Michailidi C, Considine M, et al: NF-κB and stat3 transcription factor signatures differentiate HPV-positive and HPV-negative head and neck squamous cell carcinoma. Int J Cancer. 137:1879–1889. 2015. View Article : Google Scholar : PubMed/NCBI

121 

Khaznadar SS, Khan M, Schmid E, Gebhart S, Becker ET, Krahn T and von Ahsen O: EGFR overexpression is not common in patients with head and neck cancer. Cell lines are not representative for the clinical situation in this indication. Oncotarget. 9:28965–28975. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Johnson DE, Burtness B, Leemans CR, Lui VW, Bauman JE and Grandis JR: Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 6:922020. View Article : Google Scholar : PubMed/NCBI

123 

Vander Broek R, Mohan S, Eytan DF, Chen Z and Van Waes C: The PI3K/Akt/mTOR axis in head and neck cancer: Functions, aberrations, cross-talk, and therapies. Oral Dis. 21:815–825. 2015. View Article : Google Scholar : PubMed/NCBI

124 

Li Z, Yang Z, Passaniti A, Lapidus RG, Liu X, Cullen KJ and Dan HC: A positive feedback loop involving EGFR/Akt/mTORC1 and IKK/NF-kB regulates head and neck squamous cell carcinoma proliferation. Oncotarget. 7:31892–31906. 2016. View Article : Google Scholar : PubMed/NCBI

125 

Harris RE: Cyclooxygenase-2 (cox-2) and the inflammogenesis of cancer. Subcell Biochem. 42:93–126. 2007. View Article : Google Scholar : PubMed/NCBI

126 

Lim JW, Kim H and Kim KH: Nuclear factor-kappaB regulates cyclooxygenase-2 expression and cell proliferation in human gastric cancer cells. Lab Invest. 81:349–360. 2001. View Article : Google Scholar : PubMed/NCBI

127 

Meng F, Liu L, Chin PC and D'Mello SR: Akt is a downstream target of NF-kappa B. J Biol Chem. 277:29674–29680. 2002. View Article : Google Scholar : PubMed/NCBI

128 

Ahmad A, Biersack B, Li Y, Kong D, Bao B, Schobert R, Padhye SB and Sarkar FH: Targeted regulation of PI3K/Akt/mTOR/NF-κB signaling by indole compounds and their derivatives: Mechanistic details and biological implications for cancer therapy. Anticancer Agents Med Chem. 13:1002–1013. 2013. View Article : Google Scholar : PubMed/NCBI

129 

Baud V and Karin M: Is NF-κB a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discov. 8:33–40. 2009. View Article : Google Scholar : PubMed/NCBI

130 

Tili E, Michaille JJ and Croce CM: MicroRNAs play a central role in molecular dysfunctions linking inflammation with cancer. Immunol Rev. 253:167–184. 2013. View Article : Google Scholar : PubMed/NCBI

131 

Filipowicz W, Bhattacharyya SN and Sonenberg N: Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nat Rev Genet. 9:102–114. 2008. View Article : Google Scholar : PubMed/NCBI

132 

Svoronos AA, Engelman DM and Slack FJ: OncomiR or tumor suppressor? The duplicity of microRNAs in cancer. Cancer Res. 76:3666–3670. 2016. View Article : Google Scholar : PubMed/NCBI

133 

Hu A, Huang JJ, Xu WH, Jin XJ, Li JP, Tang YJ, Huang XF, Cui HJ and Sun GB: MiR-21 and miR-375 microRNAs as candidate diagnostic biomarkers in squamous cell carcinoma of the larynx: Association with patient survival. Am J Transl Res. 6:604–613. 2014.PubMed/NCBI

134 

Hu A, Huang JJ, Xu WH, Jin XJ, Li JP, Tang YJ, Huang XF, Cui HJ, Sun GB, Li RL and Duan JL: MiR-21/miR-375 ratio is an independent prognostic factor in patients with laryngeal squamous cell carcinoma. Am J Cancer Res. 5:1775–1785. 2015.PubMed/NCBI

135 

Arantes LM, Laus AC, Melendez ME, de Carvalho AC, Sorroche BP, De Marchi PR, Evangelista AF, Scapulatempo-Neto C, de Souza Viana L and Carvalho AL: MiR-21 as prognostic biomarker in head and neck squamous cell carcinoma patients undergoing an organ preservation protocol. Oncotarget. 8:9911–9921. 2017. View Article : Google Scholar : PubMed/NCBI

136 

Wu Y, Yu J, Ma Y, Wang F and Liu H: MiR-148a and miR-375 may serve as predictive biomarkers for early diagnosis of laryngeal carcinoma. Oncol Lett. 12:871–878. 2016. View Article : Google Scholar : PubMed/NCBI

137 

Li J, Wang K, Chen X, Meng H, Song M, Wang Y, Xu X and Bai Y: Transcriptional activation of microRNA-34a by NF-kappa B in human esophageal cancer cells. BMC Mol Biol. 13:42012. View Article : Google Scholar : PubMed/NCBI

138 

Li HP, Zeng XC, Zhang B, Long JT, Zhou B, Tan GS, Zeng WX, Chen W and Yang JY: MiR-451 inhibits cell proliferation in human hepatocellular carcinoma through direct suppression of IKK-β. Carcinogenesis. 34:2443–2451. 2013. View Article : Google Scholar : PubMed/NCBI

139 

Zhou P, Zeng F, Liu J, Lv D and Liu S: Correlation betweenmir-21 expression and laryngeal carcinoma risks: A meta-analysis. J Evid Based Med. 9:32–37. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Shen Z, Zhan G, Ye D, Ren Y, Cheng L, Wu Z and Guo J: MicroRNA-34a affects the occurrence of laryngeal squamous cell carcinoma by targeting the antiapoptotic gene survivin. Med Oncol. 29:2473–2480. 2010. View Article : Google Scholar : PubMed/NCBI

141 

Chen Y, Xian PF, Yang L and Wang SX: MicroRNA-21 promotes proliferation of fibroblast-like synoviocytes through mediation of NF-κB nuclear translocation in a rat model of collagen-induced rheumatoid arthritis. Biomed Res Int. 2016:92790782016.PubMed/NCBI

142 

Kikkawa N, Kinoshita T, Nohata N, Hanazawa T, Yamamoto N, Fukumoto I, Chiyomaru T, Enokida H, Nakagawa M, Okamoto Y and Seki N: MicroRNA-504 inhibits cancer cell proliferation via targeting CDK6 in hypopharyngeal squamous cell carcinoma. Int J Oncol. 44:2085–2092. 2014. View Article : Google Scholar : PubMed/NCBI

143 

Kikkawa N, Hanazawa T, Fujimura L, Nohata N, Suzuki H, Chazono H, Sakurai D, Horiguchi S, Okamoto Y and Seki N: MiR-489 is a tumour-suppressive miRNA target PTPN11 in hypopharyngeal squamous cell carcinoma (HSCC). Br J Cancer. 103:877–884. 2010. View Article : Google Scholar : PubMed/NCBI

144 

Fukumoto I, Kinoshita T, Hanazawa T, Kikkawa N, Chiyomaru T, Enokida H, Yamamoto N, Goto Y, Nishikawa R, Nakagawa M, et al: Identification of tumour suppressive microRNA-451a in hypopharyngeal squamous cell carcinoma based on microRNA expression signature. Br J Cancer. 111:386–394. 2014. View Article : Google Scholar : PubMed/NCBI

145 

Hui AB, Lenarduzzi M, Krushel T, Waldron L, Pintilie M, Shi W, Perez-Ordonez B, Jurisica I, O'Sullivan B, Waldron J, et al: Comprehensive microRNA profiling for head and neck squamous cell carcinomas. Clin Cancer Res. 16:1129–1139. 2010. View Article : Google Scholar : PubMed/NCBI

146 

Tai J, Xiao X, Huang ZG, Yu ZK, Chen XH, Zhou WG, Chen XJ, Rao YS, Fang JG and Ni X: MicroRNAs regulate epithelial-mesenchymal transition of supraglottic laryngeal cancer. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 48:499–503. 2013.(In Chinese). PubMed/NCBI

147 

Zhao XD, Zhang W, Liang HJ and Ji WY: Overexpression of miR-155 promotes proliferation and invasion of human laryngeal squamous cell carcinoma via targeting SOCS1 and STAT3. PLoS One. 8:e563952013. View Article : Google Scholar : PubMed/NCBI

148 

Luzna P, Gregar J, Uberall I, Radova L, Prochazka V and Ehrmann J Jr: Changes of microRNAs-192, 196a and 203 correlate with Barrett's esophagus diagnosis and its progression compared to normal healthy individuals. Diagn Pathol. 6:1142011. View Article : Google Scholar : PubMed/NCBI

149 

Bus P, Siersema PD, Verbeek RE and van Baal JW: Upregulation of miRNA-143, −145, −192, and −194 in esophageal epithelial cells upon acidic bile salt stimulation. Dis Esophagus. 27:591–600. 2014. View Article : Google Scholar : PubMed/NCBI

150 

Chen D, Cabay RJ, Jin Y, Wang A, Lu Y, Shah-Khan M and Zhou X: MicroRNA deregulations in head and neck squamous cell carcinomas. J Oral Maxillofac Res. 4:e22013. View Article : Google Scholar : PubMed/NCBI

151 

Yang Z, Fang S, Di Y, Ying W, Tan Y and Gu W: Modulation of NF-κB/miR-21/PTEN pathway sensitizes non-small cell lung cancer to cisplatin. PLoS One. 10:e01215472015. View Article : Google Scholar : PubMed/NCBI

152 

Xiao T, Ling M, Xu H, Luo F, Xue J, Chen C, Bai J, Zhang Q, Wang Y, Bian Q and Liu Q: NF-κB-regulation of miR-155, via SOCS1/STAT3, is involved in the PM2.5-accelerated cell cycle and proliferation of human bronchial epithelial cells. Toxicol Appl Pharmacol. 377:1146162019. View Article : Google Scholar : PubMed/NCBI

153 

Rokavec M, Öner MG, Li H, Jackstadt R, Jiang L, Lodygin D, Kaller M, Horst D, Ziegler PK, Schwitalla S, et al: IL-6R/STAT3/miR-34a feedback loop promotes EMT-mediated colorectal cancer invasion and metastasis. J Clin Invest. 124:1853–1867. 2014. View Article : Google Scholar : PubMed/NCBI

154 

Tili E, Croce CM and Michaille JJ: MiR-155: On the crosstalk between inflammation and cancer. Int Rev Immunol. 28:264–284. 2009. View Article : Google Scholar : PubMed/NCBI

155 

Bharti AC, Donato N, Singh S and Aggarwal BB: Curcumin (diferuloylmethane) down-regulates the constitutive activation of nuclear factor-kappa B and IkappaBalpha kinase in human multiple myeloma cells, leading to suppression of proliferation and induction of apoptosis. Blood. 101:1053–1062. 2003. View Article : Google Scholar : PubMed/NCBI

156 

Dai Y, Pei XY, Rahmani M, Conrad DH, Dent P and Grant S: Interruption of the NF-kappaB pathway by Bay 11-7082 promotes UCN-01-mediated mitochondrial dysfunction and apoptosis in human multiple myeloma cells. Blood. 103:2761–2770. 2004. View Article : Google Scholar : PubMed/NCBI

157 

Monisha J, Roy NK, Bordoloi D, Kumar A, Golla R, Kotoky J, Padmavathi G and Kunnumakkara AB: Nuclear factor kappa B: A potential target to persecute head and neck cancer. Curr Drug Targets. 18:232–253. 2017. View Article : Google Scholar : PubMed/NCBI

158 

Karin M, Yamamoto Y and Wang QM: The IKK NF-kappa B system: A treasure trove for drug development. Nat Rev Drug Discov. 3:17–26. 2004. View Article : Google Scholar : PubMed/NCBI

159 

Ramadass V, Vaiyapuri T and Tergaonkar V: Small molecule NF-κB pathway inhibitors in clinic. Int J Mol Sci. 21:51642020. View Article : Google Scholar : PubMed/NCBI

160 

Greten FR, Arkan MC, Bollrath J, Hsu LC, Goode J, Miething C, Göktuna SI, Neuenhahn M, Fierer J, Paxian S, et al: NF-kappaB is a negative regulator of IL-1beta secretion as revealed by genetic and pharmacological inhibition of IKKbeta. Cell. 130:918–931. 2007. View Article : Google Scholar : PubMed/NCBI

161 

Bromberg JF, Wrzeszczynska MH, Devgan G, Zhao Y, Pestell RG, Albanese C and Darnell JE Jr: Stat3 as an oncogene. Cell. 98:295–303. 1999. View Article : Google Scholar : PubMed/NCBI

162 

Geiger JL, Grandis JR and Bauman JE: The STAT3 pathway as a therapeutic target in head and neck cancer: Barriers and innovations. Oral Oncol. 56:84–92. 2016. View Article : Google Scholar : PubMed/NCBI

163 

Nelson EA, Walker SR, Kepich A, Gashin LB, Hideshima T, Ikeda H, Chauhan D, Anderson KC and Frank DA: Nifuroxazide inhibits survival of multiple myeloma cells by directly inhibiting STAT3. Blood. 112:5095–5102. 2008. View Article : Google Scholar : PubMed/NCBI

164 

Siddiquee KA, Gunning PT, Glenn M, Katt WP, Zhang S, Schrock C, Sebti SM, Jove R, Hamilton AD and Turkson J: An oxazole-based small-molecule Stat3 inhibitor modulates Stat3 stability and processing and induces antitumor cell effects. ACS Chem Biol. 2:787–798. 2007. View Article : Google Scholar : PubMed/NCBI

165 

Johnson DE, O'Keefe RA and Grandis JR: Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 15:234–248. 2018. View Article : Google Scholar : PubMed/NCBI

166 

Goldman A, Shahidullah M, Goldman D, Khailova L, Watts G, Delamere N and Dvorak K: A novel mechanism of acid and bile acid-induced DNA damage involving Na+/H+ exchanger: Implication for Barrett's oesophagus. Gut. 59:1606–1616. 2010. View Article : Google Scholar : PubMed/NCBI

167 

Bernstein H, Bernstein C, Payne CM and Dvorak K: Bile acids as endogenous etiologic agents in gastrointestinal cancer. World J Gastroenterol. 15:3329–3340. 2009. View Article : Google Scholar : PubMed/NCBI

168 

Lechner S, Müller-Ladner U, Schlottmann K, Jung B, McClelland M, Rüschoff J, Welsh J, Schölmerich J and Kullmann F: Bile acids mimic oxidative stress induced upregulation of thioredoxin reductase in colon cancer cell lines. Carcinogenesis. 23:1281–1288. 2002. View Article : Google Scholar : PubMed/NCBI

169 

Kinner A, Wu W, Staudt C and Iliakis G: Gamma-H2AX in recognition and signaling of DNA double-strand breaks in the context of chromatin. Nucleic Acids Res. 36:5678–5694. 2008. View Article : Google Scholar : PubMed/NCBI

170 

Rothkamm K and Löbrich M: Evidence for a lack of DNA double-strand break repair in human cells exposed to very low x-ray doses. Proc Natl Acad Sci USA. 100:5057–5062. 2003. View Article : Google Scholar : PubMed/NCBI

171 

Nikitaki Z, Hellweg CE, Georgakilas AG and Ravanat JL: Stress-induced DNA damage biomarkers: Applications and limitations. Front Chem. 3:352015. View Article : Google Scholar : PubMed/NCBI

172 

Tsantoulis PK, Kotsinas A, Sfikakis PP, Evangelou K, Sideridou M, Levy B, Mo L, Kittas C, Wu XR, Papavassiliou AG and Gorgoulis VG: Oncogene-induced replication stress preferentially targets common fragile sites in preneoplastic lesions. A genome-wide study. Oncogene. 27:3256–3264. 2008. View Article : Google Scholar : PubMed/NCBI

173 

Choudhari SK, Chaudhary M, Gadbail AR, Sharma A and Tekade S: Oxidative and antioxidative mechanisms in oral cancer and precancer: A review. Oral Oncol. 50:10–18. 2014. View Article : Google Scholar : PubMed/NCBI

174 

Janssens S and Tschopp J: Signals from within: The DNA-damage-induced NF-kappaB response. Cell Death Differ. 13:773–784. 2006. View Article : Google Scholar : PubMed/NCBI

175 

Kawanishi S, Ohnishi S, Ma N, Hiraku Y and Murata M: Crosstalk between DNA damage and inflammation in the multiple steps of carcinogenesis. Int J Mol Sci. 18:18082017. View Article : Google Scholar : PubMed/NCBI

176 

Tafani M, Sansone L, Limana F, Arcangeli T, De Santis E, Polese M, Fini M and Russo MA: The interplay of reactive oxygen species, hypoxia, inflammation, and sirtuins in cancer initiation and progression. Oxid Med Cell Longev. 2016:39071472016. View Article : Google Scholar : PubMed/NCBI

177 

Li D and Cao W: Bile acid receptor TGR5, NADPH Oxidase NOX5-S and CREB mediate bile acid-induced DNA damage in Barrett's esophageal adenocarcinoma cells. Sci Rep. 6:315382016. View Article : Google Scholar : PubMed/NCBI

178 

Nema R, Vishwakarma S, Agarwal R, Panday RK and Kumar A: Emerging role of sphingosine-1-phosphate signaling in head and neck squamous cell carcinoma. Onco Targets Ther. 9:3269–3280. 2016.PubMed/NCBI

179 

Tamashiro PM, Furuya H, Shimizu Y, Iino K and Kawamori T: The impact of sphingosine kinase-1 in head and neck cancer. Biomolecules. 3:481–513. 2013. View Article : Google Scholar : PubMed/NCBI

180 

Liu R, Li X, Hylemon PB and Zhou H: Conjugated bile acids promote invasive growth of esophageal adenocarcinoma cells and cancer stem cell expansion via sphingosine 1-phosphate receptor 2-mediated yes-associated protein activation. Am J Pathol. 188:2042–2058. 2018. View Article : Google Scholar : PubMed/NCBI

181 

Pang C, LaLonde A, Godfrey TE, Que J, Sun J, Wu TT and Zhou Z: Bile salt receptor TGR5 is highly expressed in esophageal adenocarcinoma and precancerous lesions with significantly worse overall survival and gender differences. Clin Exp Gastroenterol. 10:29–37. 2017. View Article : Google Scholar : PubMed/NCBI

182 

Guan B, Li H, Yang Z, Hoque A and Xu X: Inhibition of farnesoid X receptor controls esophageal cancer cell growth in vitro and in nude mouse xenografts. Cancer. 119:1321–1329. 2013. View Article : Google Scholar : PubMed/NCBI

183 

Yu JH, Zheng JB, Qi J, Yang K, Wu YH, Wang K, Wang CB and Sun XJ: Bile acids promote gastric intestinal metaplasia by upregulating CDX2 and MUC2 expression via the FXR/NF-κB signaling pathway. Int J Oncol. 54:879–892. 2019.PubMed/NCBI

184 

Schweitzer A, Knauer SK and Stauber RH: Nuclear receptors in head and neck cancer: Current knowledge and perspectives. Int J Cancer. 126:801–809. 2010.PubMed/NCBI

185 

Gadaleta RM, Oldenburg B, Willemsen EC, Spit M, Murzilli S, Salvatore L, Klomp LW, Siersema PD, van Erpecum KJ and van Mil SW: Activation of bile salt nuclear receptor FXR is repressed by pro-inflammatory cytokines activating NF-κB signaling in the intestine. Biochim Biophys Acta. 1812:851–858. 2011. View Article : Google Scholar : PubMed/NCBI

186 

Wang YD, Chen WD, Wang M, Yu D, Forman BM and Huang W: Farnesoid X receptor antagonizes nuclear factor kappaB in hepatic inflammatory response. Hepatology. 48:1632–1643. 2008. View Article : Google Scholar : PubMed/NCBI

187 

Wang Z, Li Y and Sarkar FH: Signaling mechanism(s) of reactive oxygen species in epithelial-mesenchymal transition reminiscent of cancer stem cells in tumor progression. Curr Stem Cell Res Ther. 5:74–80. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2021
Volume 46 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Vageli DP, Doukas SG, Doukas PG and Judson BL: Bile reflux and hypopharyngeal cancer (Review). Oncol Rep 46: 244, 2021
APA
Vageli, D.P., Doukas, S.G., Doukas, P.G., & Judson, B.L. (2021). Bile reflux and hypopharyngeal cancer (Review). Oncology Reports, 46, 244. https://doi.org/10.3892/or.2021.8195
MLA
Vageli, D. P., Doukas, S. G., Doukas, P. G., Judson, B. L."Bile reflux and hypopharyngeal cancer (Review)". Oncology Reports 46.5 (2021): 244.
Chicago
Vageli, D. P., Doukas, S. G., Doukas, P. G., Judson, B. L."Bile reflux and hypopharyngeal cancer (Review)". Oncology Reports 46, no. 5 (2021): 244. https://doi.org/10.3892/or.2021.8195