Next Article in Journal
Extracranial Germ Cell Tumors in Children: Ten Years of Experience in Three Children’s Medical Centers in Shanghai
Next Article in Special Issue
Treatment Outcomes and Risk Factors for Incomplete Treatment after Definitive Chemoradiotherapy for Non-Resectable or Metastatic Esophageal Cancer
Previous Article in Journal
Treatment of Acute Myeloid Leukemia in Older Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Biology and Clinical Management of Esophageal Adenocarcinoma

by
Shulin Li
1,2,
Sanne Johanna Maria Hoefnagel
3 and
Kausilia Krishnawatie Krishnadath
4,5,*
1
Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
2
Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
3
Department of Internal Medicine, CWZ, 6532 SZ Nijmegen, The Netherlands
4
Department of Gastroenterology and Hepatology, Antwerp University Hospital, 2650 Edegem, Belgium
5
Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, 2000 Antwerpen, Belgium
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(22), 5410; https://doi.org/10.3390/cancers15225410
Submission received: 7 September 2023 / Revised: 10 November 2023 / Accepted: 12 November 2023 / Published: 14 November 2023
(This article belongs to the Special Issue New Trends in Esophageal Cancer Management (Volume II))

Abstract

:

Simple Summary

Esophageal adenocarcinoma (EAC) is one of the two main subtypes of esophageal cancer. EAC is a highly lethal disease with rising incidence in Western countries. EAC is associated with chronic gastroesophageal reflux disease and Barrett’s esophagus and mostly occurs in the distal esophagus. In the past decades, much effort has been made to improve treatment strategies, including regimens with chemoradiotherapy, targeted and immune therapies. Despite the multi-modal therapies, the survival of EAC patients has improved only marginally, and major breakthroughs in EAC treatment have not been achieved. We aim to summarize the literature on the comprehensive molecular landscape of EAC to elucidate factors underlying the EAC malignant behavior and poor outcomes. We discuss in detail the etiology, genetics, epigenetics and histology of EAC, as well as the currently employed therapies. Better knowledge about the molecular biology of EAC learned from this review may provide leads for developing novel therapies in the future.

Abstract

Esophageal adenocarcinoma (EAC) is a highly lethal malignancy. Due to its rising incidence, EAC has become a severe health challenge in Western countries. Current treatment strategies are mainly chosen based on disease stage and clinical features, whereas the biological background is hardly considered. In this study, we performed a comprehensive review of existing studies and discussed how etiology, genetics and epigenetic characteristics, together with the tumor microenvironment, contribute to the malignant behavior and dismal prognosis of EAC. During the development of EAC, several intestinal-type proteins and signaling cascades are induced. The anti-inflammatory and immunosuppressive microenvironment is associated with poor survival. The accumulation of somatic mutations at the early phase and chromosomal structural rearrangements at relatively later time points contribute to the dynamic and heterogeneous genetic landscape of EAC. EAC is also characterized by frequent DNA methylation and dysregulation of microRNAs. We summarize the findings of dysregulations of specific cytokines, chemokines and immune cells in the tumor microenvironment and conclude that DNA methylation and microRNAs vary with each different phase of BE, LGD, HGD, early EAC and invasive EAC. Furthermore, we discuss the suitability of the currently employed therapies in the clinic and possible new therapies in the future. The development of targeted and immune therapies has been hampered by the heterogeneous genetic characteristics of EAC. In view of this, the up-to-date knowledge revealed by this work is absolutely important for future EAC studies and the discovery of new therapeutics.

1. Introduction

Esophageal cancer (EC) is predominantly divided into histological subtypes: esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC). Different from ESCC, which mostly occurs in the upper and middle esophagus, EAC is mainly (>90%) located in the lower esophagus in the proximity of the gastro-esophageal junction [1,2,3]. This review focuses on EAC. The incidence rate of EAC has been increasing in Western countries [4]. EAC is a highly aggressive cancer and known for rapid tumor dissemination leading to distant metastases, which is associated with poor survival [5]. Barrett’s esophagus (BE) induced by gastroesophageal reflux disease (GERD) is the only known precursor of EAC. EAC develops via a stepwise process, commonly in compliance with the well-known sequence of GERD/BE/BE with dysplasia/adenocarcinoma. Obesity and GERD are two main risk factors associated with BE and EAC [6,7,8,9]. Other risk factors include male sex, smoking and Caucasian ethnicity [10]. In the past decades, much effort has been made in identifying and monitoring BE patients via endoscopic surveillance in order to detect early progression to dysplasia and prevent the occurrence of EAC in a timely manner. Although the majority of individuals with BE (90–95%) under surveillance do not progress to EAC or are treated successfully in the case of dysplasia or mucosal cancers, a small portion of BE patients (5–10%) even in careful surveillance can rapidly develop EAC [11]. The lesions may be missed potentially because of a lack of visible endoscopic abnormalities and sampling errors when randomly taking endoscopic biopsies [12]. Moreover, genetic hits in tumor driver genes may lead to accelerated malignant degeneration. The majority of EAC patients are incurable because of late-stage disease at the time of diagnosis, because of a relatively late onset of symptoms. The proportion of patients with late-stage disease at the time of diagnosis has been increasing over time [2,13].
For patients with EAC undergoing treatment with curative intent, neoadjuvant chemoradiotherapy (CROSS) or perioperative chemotherapy (FLOT) regimens are the main strategies employed in the clinic. However, the majority of EAC patients poorly respond to the currently employed therapies, with a ten-year survival of 36% in the Western population according to the CROSS regimen [14]. Notably, 82% of deceased patients following the CROSS regimen are attributed to disease relapse after treatment [14]. There is an urgent clinical need to better understand specific biological mechanisms that underlie dismal outcomes and acquired resistance following treatment of EAC patients.
Targeted and immune therapies based on the unique molecular features and genomic profiles of EAC may be beneficial to EAC patients. The only targeted therapy currently applied for both locally advanced and metastatic EAC is the HER2 antibody Trastuzumab. Around 30% of EAC patients are HER2 positive and may potentially respond to this therapy [15,16]. Pembrolizumab, which blocks the interaction between PD-1 and PD-L1 is an FDA-approved immune checkpoint inhibitor for metastatic EAC. This agent is potentially beneficial to 18% of EAC patients who express PD-L1 [17]. Identifying potential molecular targets could be beneficial to the future development of personalized therapies.
This is a comprehensive and up-to-date review, aiming to provide the latest knowledge about EAC for both researchers and clinicians. This review discusses multiple aspects of EAC, including etiology, biology, genetics and epigenetics, and clinical measures to reduce the burden of BE and EAC. Regarding the etiology, we discuss the three predominant contributors of EAC occurrence: obesity, GERD and BE. In the aspect of the biology and tumor microenvironment of EAC, we discuss driver molecules and signaling pathways associated with GERD leading to the premalignant condition BE and malignant EAC. We discuss changes in the tumor microenvironment and the consequences during the development and progression of EAC. For the review on genetics and epigenetics, we discuss the literature on distinct genetic characteristics associated with EAC among various studies. Especially for epigenetics, important findings appear in each phase of BE, LGD, HGD, early EAC and invasive EAC. Importantly, we discuss the latest measures for reducing GERD and BE, including surveillance of BE, treatment of BE, treatment of EAC and new therapies to be expected in the future.

2. Epidemiological and Biological Characteristics

Summary: In this part, besides the epidemiological characteristics of EAC, we predominantly discuss the origin of EAC and BE, as well as the association between EAC with obesity, GERD and BE. We also discuss main driver molecules, signaling pathways and the changes in the tumor microenvironment for the development and progression of EAC. Furthermore, we discuss genetic and epigenetic characteristics by reviewing existing studies and explore possible molecular targets for personalized therapy. Why is the incidence of EAC rising despite surveillance and treatment of GERD and BE? Which driver molecules or pathways could have potential as therapeutic targets?

2.1. Epidemiology and Etiology

Although EAC cases only account for 14% of EC cases worldwide, it is the predominant subtype in Western countries including Western and Northern Europe, Australia, Canada and the USA [18]. In contrast to ESCC, the annual incidence of EAC has drastically increased by 767% with an average rate of 5.11% per year, from 1973 to 2017 in the USA [19]. In 2012, there were approximately 52,000 EAC cases (41,000 male and 11,000 female) worldwide [4]. In 2020, this increased to approximately 85,700, among which males are four times more often affected compared to females. Moreover, young EAC patients (< 50 years) are increasingly frequent, while there is no correlation between the age of diagnosis and survival [20]. The distribution of EAC is also affected by geographic and racial factors, suggesting a genetic predisposition. For instance, in the USA, the incidence rate of EAC from high to low displayed the following sequence: non-Hispanic whites/Hispanic whites/(American Indian/Alaska Native)/blacks/(Asian/Pacific islanders) [21]. Approximately 7% of BE and EAC cases are associated within families and presumably attributed to inherited factors [22,23]. A germline variant S631G encoded by the gene VSIG10L has been identified as a susceptible source of familial EAC or BE [24], which provides a lead for the detection of BE and EAC via screening in families carrying inherited factors. In addition, germline alteration in MGST1 or FOXP1 may be predisposing factors for BE and EAC [25,26].
Common symptoms of EAC patients are dysphagia and weight loss. A total of 79% of EAC patients present with dysphagia and 53% with weight loss, whereas 47% of patients present with a combination of both [3]. The onset of these symptoms is associated with worse prognosis of EAC [3]. GERD, obesity and smoking are risk factors for EAC [6,7,8,27,28]. The increasing incidence of EAC is likely attributed to the increasing prevalence of GERD and obesity. Studies found that abdominal obesity rather than the body mass index (BMI) predisposes to the increased risk for BE and presumably EAC [29,30,31]. GERD is most prominent in the distal esophagus [32]. Abdominal obesity can increase symptoms of GERD through increased intra-abdominal pressure [33]. Moreover, a high-fat diet underlying obesity promotes esophageal carcinogenesis in BE by inducing dysplasia due to alterations in the esophageal microenvironment and the gut microbiome [34]. Other risk factors include cigarette smoking, which approximately doubles the risk of EAC, and a low intake of vegetables and fruit [35]. Epidemiological studies showed that Helicobacter pylori infection rates are negatively associated with the incidence of EAC [36].
There is an ongoing debate regarding the cell of origin in BE even if many original studies have been performed in the past decades (Table 1). Nevertheless, it is affirmative that BE is a metaplastic condition in which the normal esophageal squamous mucosa is replaced by metaplastic columnar types of epithelia [37]. BE occurs in approximately 10% of individuals with GERD [38], whereas 40% of BE patients have no symptoms of GERD [39]. GERD not only accelerates the development of Barrett’s-like metaplasia and dysplasia [40] but also strongly contributes to the progression of BE to EAC [6]. Although 50% of EAC patients present without BE [41,42], and 40–48% of EAC patients have no or infrequent GERD symptoms [35], EAC may originate from BE even if BE is absent or unapparent at the diagnosis of EAC [43]. Studies found that EAC patients without a history of intestinal metaplasia/BE have more aggressive behavior and poorer prognosis [41,42].
Dysplasia is a key feature of BE, which is associated with an increased risk of progression to EAC. Progression to EAC is via cellular changes of low-grade dysplasia (LGD) and high-grade dysplasia (HGD). The presence of epithelial dysplasia remarkably affects the risk of the cancerous transformation in BE patients. The incidence rate of EAC in BE patients with no dysplasia, low-grade dysplasia and high-grade dysplasia are around 0.6%, 13.4% and 25%, respectively [10]. Other studies support that the risk rate of BE with no dysplasia on progression to EAC lies between 0.2 and 0.7% per patient per year [49,50]. The diagnosis of LGD, HGD and adenocarcinoma based on histopathological features is contentious due to low inter-observer agreement, especially for LGD [51]. Therefore, reported rates of malignant progression in patients with LGD are highly variable. One study reports a progression rate of LGD to HGD or EAC of 13.4% per patient per year [52], whereas another study reports an incidence rate of 0.84% [53]. To minimize low inter-observer agreement, AGA guidelines recommend that Barrett’s dysplasia should be confirmed by a second expert gastrointestinal pathologist [54]. Definitive and simplified histopathological criteria and larger biopsy specimens to avoid sampling bias are needed to improve inter-observer agreement. Barrett’s segment length is another risk factor for neoplastic progression in BE. Barrett’s segment length is significantly associated with the risk of malignant progression of non-dysplastic Barrett’s esophagus (NDBE) [55]. One study found that the risk of malignant progression increases by 19% per centimeter of Barrett’s segment length [56]. One study found that the incidence rates of EAC progressed from NDBE with short-segment BE (< 3 cm) and long-segment BE (≥ 3 cm) are 0.24% and 0.76%, respectively [57].

2.2. Histology

According to WHO classification, EAC is classified into three subgroups based on the percentage of gland formation: poorly differentiated (<50%), moderately differentiated (50–95%) and well differentiated (≥95%) [58]. This classification shows no correlation with survival [59]. Moreover, EAC can be classified according to Lauren’s classification which distinguishes the intestinal type, diffuse type and mixed type of EAC. One study showed that Lauren’s classification subtypes can successfully predict response to chemotherapy and survival [60]. EAC classified as diffuse and mixed types are associated with less pathological response to chemoradiotherapy and poor prognosis [61]. A more analytical approach to identifying histological subtypes that provide useful prognostic information is an ongoing topic of research. One study classified EAC cases into histological subtypes including mucinous muconodular carcinomas, invasive mucinous carcinomas, diffuse desmoplastic carcinomas, diffuse anaplastic carcinomas and mixed carcinomas, which showed prognostic significance [59]. Also, another histologic classification of EAC into papillary, tubular, mucinous and signet ring subtypes has been proposed, but this classification lacks evidence of clinical impact [62]. Subgrouping based on other features than histology is discussed later on in “Genetics” and “Epigenetics”.

2.3. Biology and Immunology

EAC initiation is driven by GERD through reprogramming of cell proliferation and differentiation in the esophageal mucosa [63]. Long-standing GERD causes metaplasia of the esophageal squamous epithelium in a subset of patients through dysregulation of multiple driver molecules and pathways (Figure 1). GERD activates the NF-κB signaling in esophageal squamous cells [64,65,66], a pathway that is associated with abnormal cell proliferation and differentiation, treatment resistance and metastasis in multiple cancers [67]. In EAC patients, the expression of NF-κB is negatively correlated with complete pathologic response to neoadjuvant chemoradiotherapy [68]. GERD also increases the expression of CDX2 [69], which drives the development of an intestinal-type metaplasia [70,71,72]. One study identified activation of CDX2 in the development of BE [43]. GERD activates the CDX2 promotor through the activation of NF-κB and upregulates the expression of the CDX2 protein, leading to the production of the intestinal-type protein MUC2 [73]. GERD induces the expression of Notch ligand Delta-like1 (Dll1) via a CDX2-dependent pathway in the development of BE, and Dll1 expression is significantly higher in BE than in normal human esophageal squamous epithelium [74]. Dll1 expression facilitates intestinal-type metaplasia in esophageal squamous cells in combination with CDX2 expression. BMPs, especially BMP4, are highly expressed in BE and EAC, and BMP4 alone or in combination with CDX2 drives the differentiation of columnar epithelia [75,76]. BMP2 and BMP4 are both highly expressed in EAC, which may enhance the aggressiveness of EAC through triggering of non-canonical BMP signaling [77]. Moreover, COX-2 regulated by NF-κB exerts an important role in carcinogenesis in multiple cancers [78]. The expression of the COX-2 protein is significantly higher in patients with BE and EAC compared to normal squamous esophageal epithelia in healthy patients [79,80]. Genetic variants of the COX-2 gene are significantly associated with an increased risk of EAC [81,82]. Targeting COX-2 has been suggested as a potential therapy in EAC. Inhibition of COX-2 with the inhibitor MF-Tricyclic reduces the incidence of EAC in a mice model of BE [83]. Interestingly, one study reported that epithelial-to-mesenchymal transition (EMT) is associated with the progression from early EAC to invasive EAC. The measurement of EMT markers prior to intervention may help to make the clinical decision between endoscopic submucosal dissection or esophagectomy as the preferred treatment [84].
Cytokines and chemokines are important components of the tumor microenvironment. Cytokines mediate the inflammatory context in the tumor microenvironment, whereas chemokines are directly associated with the malignant behavior of tumor cells, for instance, cell migration. The change in cytokines and chemokines caused by GERD in the esophageal epithelium contributes to the development and progression of BE and EAC. GERD causes overexpression of the pro-inflammatory cytokines IL-1β, IL-8 and IL-6 in esophageal epithelium cells [85]. IL-1β is overexpressed in both BE and EAC compared to normal squamous esophagus [32,86]. IL-1β overexpression in the mouse esophagus induces overexpression of BMP4, SHH and CDX2, and the gene expression profile of the IL-1β-overexpressed mouse esophagus closely resembles that of human BE and EAC [40]. This suggests that IL-1β induces the development and progression of BE and EAC. In addition, IL-8 expression is significantly higher in patients with BE or EAC than in those who have only GERD symptoms [87]. The maximal degree of inflammation with a significant increase in pro-inflammatory IL-1β and IL-8 expression is located at the new squamo-columnar junction, whereas the minimal degree of inflammation with a significant increase in the anti-inflammatory IL-10 expression is located at the distal portion of the BE segment where most EAC occur [32]. IL-10 expression in EAC is associated with worse overall survival [88]. One study reports that IL-6 produced by cancer-associated fibroblasts is significantly higher in EAC than in normal tissue, which leads to resistance of EAC cells to chemoradiotherapy (CROSS regimen) [89]. The anti-inflammatory microenvironment existing in EAC and its peritumoral tissue is negatively associated with the survival of patients [88]. Moreover, one study found that the expression of CXCR4, a chemokine receptor, increases with the progression from BE to LGD and HGD and EAC in a mouse model [85]. CXCR4 and its chemokine ligand CXCL12 are highly expressed in EAC, and the expression is associated with poor prognosis and lymph node metastases [85,90]. High expression of the chemokine receptor CXCR7 and its chemokine ligand CXCL12 in EAC is also associated with poor prognosis [91]. In addition, the chemokines, chemokine receptors, cytokines and interleukins CXCR1, CXCR2, CXCL1, CXCL2, CXCL3, CXCL6, CXCL8, CCL15, CCR4, IL-2, IL-6, IL-7, IL-8, IL-15 and IL-18 are significantly higher expressed in HGD/EAC compared to LGD/BE [92].
The immune microenvironment plays a key role in the development and progression of BE and EAC. The immunosuppressive Foxp3 FOXP3 is significantly higher expressed at the mRNA level in BE than in normal squamous epithelium [93]. The number of FOXP3+ lymphocytes is significantly higher in BE and EAC than in non-metaplastic esophagitis [94] and is associated with high Ki-67 expression [95]. Ki-67 is higher expressed in EAC than in BE and higher in BE than in healthy person [96]. In addition, the number of FOXP3+ lymphocytes is significantly higher in HGD than in LGD and higher in LGD than in NDBE [95]. The number of CD3+ lymphocytes is also higher in BE tissues adjacent to EAC than in BE tissues without EAC and is associated with the absence of Barrett’s metaplasia [95]. Another study also found that the number of CD3+ FOXP3+ regulatory T cells significantly increases from BE to LGD and to EAC [92]. A high CD8+ lymphocyte number in BE adjacent to EAC is associated with worse overall survival and lymph node metastasis [95]. CD8+ cytotoxic T cells and CD163+ macrophages are both decreased in EAC compared to HGD [92]. BE is associated with a transition from a pro-inflammatory Th1-type immune response to an anti-inflammatory Th2-type immune response [97]. The number of Th2 effector cells (plasma cells and mast cells) is higher in BE than in reflux esophagitis, whereas the number of Th1 effector cells (macrophages and CD8+ T cells) is significantly lower in BE than in reflux esophagitis [98]. Another study also confirmed that BE harbors a Th2-predominant cytokine profile compared to the pro-inflammatory nature of esophagitis [99]. In addition, CTLA4 expressing in regulatory T cells with an anti-inflammatory role in EAC is upregulated, which is associated with poorer overall survival [88]. M1 macrophages are associated with tumor inhibition through intrinsic phagocytosis and increased anti-tumor inflammatory responses, whereas M2 macrophages are assumed to be tumor-promoting by involvement in stromal activation and remodeling, angiogenesis, neovascularization and immuno-suppression [100]. The infiltration of CD8+ T cells and M1 macrophages is significantly lower in EAC compared to the adjacent normal esophageal tissue [101]. M2 macrophages are significantly increased in EAC compared to HGD [92]. Eosinophils are significantly decreased in EAC compared to BE, LGD or HGD [92]. Eosinophilic infiltration in cancer has been associated with better prognosis [102]. Compared to the adjacent normal esophageal tissue, the microenvironment of EAC is characterized by infiltration with T regulatory cells and effector T cells, expansion of plasmacytoid dendritic cells, increased expression of cancer-associated fibroblasts and reduction in endothelial cells. Interestingly, a reversal of these characteristics happens following treatment with the FLOT regimen [103]. Another study shows that poor infiltration of cytotoxic effector cells and increased immune inhibitory signaling are the main characteristics of the EAC microenvironment [92].

2.4. Genetics

The esophageal mucosa is recurrently exposed to gastric and bile acid refluxates caused by GERD. This may cause DNA damage and the formation of mutational patterns, such as an A > C transversion [104,105]. A common mutational pattern T: A > G : C also exists in EAC [105,106]. This mutational signature is similarly enriched in SNVs of both BE and EAC, suggesting a common pathogenic etiology [107], which might be the result of long-standing gastroesophageal reflux [105,106,108].
High inter- and intra-tumor heterogeneity is a significant feature of EAC. Extensive genomic heterogeneity commonly presents in the advanced stage of cancer [109]. EAC is characterized by a high mutational burden, chromosomal instability (CIN), copy number variations (CNVs) and highly variable mutational signatures. The mutational rate in EAC is more significant compared to other types of cancer including gastric cancer, pancreatic cancer, colorectal cancer and hepatocellular carcinoma [110]. EAC harbors a median of 26,161 genome-wide mutations per tumor with a median mutational frequency of 9.9 mutations/Mb (range of 7.1–25.2/Mb) [105]. In comparison, 2.64 mutations/Mb (range 0.65–28.2/Mb) are seen in pancreatic cancer and 5.801 mutations/Mb (range 0.725–88.470/Mb) in gastric cancer [111,112]. One study showed a significant correlation between mutation frequency and the pathological grade of dysplasia (NDBE vs. DBEs) [113]. However, another study found that the mutation rate poorly correlates with dysplasia grade [107].
Genetic clonal diversity is a significant feature of EAC [114]. The prevailing view in EAC is that a gradual accumulation of mutations drives the transformation of precancerous lesions to EAC. Inactivation of the tumor suppressor CDKN2A, and clonal or polyclonal expansion in NDBE, followed by a dysplastic clone with TP53 inactivation and other somatic genetic variations, is one possible sequence of EAC development [11,115]. However, another study demonstrated that EAC initiation is through a more direct way in which TP53-mutant cells undergo genome doubling, followed by the acquisition of oncogenic amplifications rather than through the gradual accumulation of tumor-suppressor alterations [113]. It is known that carcinogenic factors drive abnormal clones that erode and occupy most regions of esophageal mucosa through clonal expansion. Genomic analyses of the normal esophageal epithelium (upper- and mid-esophagus) showed that strong expansion of clones with cancer-associated mutations in Notch1 and TP53 occurs in up to 80% and 37% of healthy middle-aged and elderly people, respectively [116]. Notably, the mutational burden of Notch1 is several times higher in the normal esophagus than in EC [116]. This indicates a complex somatic clonal evolution within normal esophageal tissue. Measures of genetic clonal diversity have been identified as robust biomarkers to stratify for cancer risk in endoscopically surveyed BE patients [117].
Notably, point mutations are reported to be dominant in EAC [105]. Some susceptibility genetic loci involved in the embryonic development of the esophagus, for instance, FOXF, BARX1 and ABCC5, were identified to be associated with BE and EAC risk [118,119]. To date, many mutated genes of EAC have been identified from EAC genome sequencing studies. We identified seven studies considering the analysis of mutated genes in EAC. The most frequently reported significantly mutated genes in EAC by DNA sequencing include TP53, SMAD4 and CDKN2A, which are reported in all seven EAC studies [1,105,106,120,121,122,123]. The rest mutated genes are reported in one or few studies (Table 2). The discrepancy in mutated genes identified from studies is likely due to different detecting methods, small study cohorts (between 112 and 551 patients) and distinct demographics. Targeted sequencing or exome sequencing as used in the three studies mentioned above [1,105,122] is not able to detect large-scale structural rearrangements and heterogeneity that exist in EAC [108]. In contrast, whole-genome sequencing (WGS) as applied in four studies [106,120,121,123] is able to more comprehensively profile genomic alterations in EAC.
TP53 is a tumor suppressor gene and is widely mutated in a variety of human cancers including EAC [124]. One study found that the majority of mutated genes in EAC are also mutated at the NDBE stage, with the exception of TP53, which is only mutated in HGD (72%) and EAC (69%), and SMAD4, which is only mutated in the EAC stage (13%) [106]. TP53 mutation cannot differentiate between high-grade dysplasia and EAC. However, in EAC, TP53 mutations are significantly correlated with the grade of histologic differentiation and worse survival [125]. SMAD4 is the only identified gene mutation that is able to differentiate between EAC and precancerous stages. However, SMAD4 mutations are only found in about 13% of EAC [106]. SMAD4 loss in EAC is also correlated with disease recurrence and poor survival [77,126].
One study reported that chromosomal structural rearrangements and copy number variations drive the development of EAC [123]. The genes with CNVs in EAC are reported in multiple studies (Table 3). The genomes of EAC are highly rearranged and complex (median of 263 structural variants (SVs) per tumor, range 126–776) [108]. As a pattern of chromosomal structural rearrangements, chromothripsis has been reported to present in 32.5% of EAC [108], whereas the prevalence of chromothripsis in other cancers is only 5% [127]. One study reports that the number of chromosomal aberrations is significantly correlated to patient survival in EAC [128].
In contrast to the gradual accumulation of somatic mutations (short deletions/insertions or single-nucleotide substitutions), chromosomal structural rearrangements occur relatively late in cancer progression and may occur within a short time [131,132]. Copy number variations are significantly increased in EAC compared to BE [107]. The levels of genomic instability are positively correlated with advanced stages of EAC [129]. A longitudinal study of BE to EAC progression found that progressors acquire significantly more somatic chromosomal alterations and genomic diversity during a median period of four years before EAC diagnosis, whereas the genomes of non-progressors remain stable over prolonged periods of time [11]. The frequency of copy number variations has been found to be low in the case of no dysplasia (1.3%) and increases along with the stages of dysplasia (4.7%) and reach high levels in EAC (30%) [130]. In EAC, copy number gains occur mostly for chromosomes 7, 8, 19 and 20, whereas chromosomes 5, 9, 18 and 21 are more common for copy number losses [130]. Several structural genetic alterations in specific genes, including copy number variation and loss of heterozygosity, are associated with the progression of BE to EAC. The loss of heterozygosity of the TP53 gene at chromosome 17p (17p (p53) LOH) is a predictor of progression to EAC [133]. Moreover, analysis of the abnormalities of the genes CDKN2A and MYC and aneusomy significantly improved risk prediction for the progression of BE to EAC [134]. Furthermore, a combination of 9p LOH, 17p LOH and DNA content tetraploidy and aneuploidy proved to be significant, independent predictors for EAC risk [135]. Moreover, amplification of potential therapeutic targeted genes, for instance, receptor tyrosine kinases (RTKs) HER2, EGFR, MET and FGFR, widely exists in EAC [105,123,136]. For instance, high-level amplifications of HER2 and EGFR are found in 17% and 11% of EAC, respectively [123]. HER2 amplification and overexpression are associated with poor survival of EAC patients [137]. Co-amplification of different RTKs, for instance, HER2 and EGFR, is frequent in EAC and may cause resistance to anti-RTK therapies [123]. Therefore, a combination of RTK antibodies/inhibitors to circumvent tumor resistance may be an effective therapeutic strategy. Interestingly, several studies found that shorter telomere length in BE patients is associated with an increased risk of progression to EAC [138,139,140]. Obesity and cigarette smoking can reduce telomere length [141,142]. In addition, telomere integrity analysis based on whole-genome sequencing found somatic telomere shortening in EAC, which is associated with complex chromosomal rearrangements [108]. This means that telomere length could be applicable to the risk stratification of Barrett patients.
Subclassifications are of paramount importance for the development of new targeted therapies and enable identification of new targets in a specific subset of patients. Nones et al. categorize EAC into three subtypes using structural rearrangement patterns: unstable genomes (tumors with ≥450 structural variants (SVs)), scattered (<450 SVs evenly distributed across the genome) and complex localized (with a concentration of SVs in a single or few chromosomes) [108]. The association between these subtypes and prognosis or therapy response needs to be investigated in the future. Based on mutational profiles, EAC can be divided into three subtypes: (i) enriched for BRCA signature in the homologous recombination pathway; (ii) a dominant T > G mutational pattern; and (iii) a C > A / T mutational pattern. These groups provide a strategy for therapy selection [123].
Next to genomic abnormalities, transcriptomic profiling by microarray or RNA sequencing has been an important topic of research [143]. Based on RNA profiling, the TCGA research group investigated gastric adenocarcinoma and proposed a new classification with four subtypes: chromosomal instability (CIN), microsatellite instability (MSI), genomic stability (GS) and Epstein–Barr-virus-positive cancers [144]. In an additional study, the TCGA group showed that EAC has a high frequency of chromosomal instability (CIN) and therefore resemble one of the four subtypes of gastric cancer. In this study, 71 out of 72 EAC cases were identified to have CIN indicating similarities between EAC and gastric cancers with CIN [1]. More recently, our research group was able to further differentiate EAC into three molecular subgroups, characterized by (i) p38 MAPK/Toll-like receptor signaling, (ii) an activated immune system and (iii) impaired cell adhesion, and this classification was associated with response to neo-adjuvant treatment [143].
Although much effort has been made to understand the mechanism underlying genetic differences, only few critical mutations and chromosomal structural rearrangements have been identified from the comparison of pre-EAC lesions and EAC. It is challenging to identify molecular biomarkers for predicting the progression of precursor lesions to EAC. These biomarkers are critical, for instance, as tools for the identification of high-risk populations for population screening programs. At present, next to RNA biomarkers, the analysis of epigenetics may hold promise to identify EAC subtypes and provide therapeutic targets for EAC.

2.5. Epigenetics

The role of DNA methylation has been studied in BE and the development of EAC (Table 4). The levels of DNA methylation in BE and EAC are similar and significantly higher than in normal esophageal tissue [145]. Genes that are frequently methylated in many cancers including APC, ID4, MGMT, SFRP1, TIMP3 and TMEFF2 have similar methylation frequencies in EAC and BE, whereas CDKN2A and RUNX3 are significantly more frequently methylated in EAC than in BE [146]. The hypermethylation of the CDKN2A promoter is associated with the progression of BE to EAC [147,148]. Moreover, in many EAC, the MT3 gene is hypermethylated, which is associated with advanced tumor stages [149]. One study reported that BE progressing to EAC harbors widespread DNA methylation that particularly occurs in 70% of known imprinted genes, compared to BE cases that do not progress to EAC [150]. Based on the number of methylations of four genes, respectively, SLC22A18, RIN2, PIGR and GJA12, stratification of BE patients into three risk groups (low, intermediate and high) with prognostic significance could be performed [150].
MicroRNAs (miRNAs) have been identified as oncogenes or tumor suppressors through their roles in regulating the expression of target genes in EAC. Altered expression of microRNAs can affect the development and progression of EAC (Table 5). One study identified 13 differentially expressed miRNAs during the development of BE (when comparing BE with normal squamous esophageal epithelium). These genes include upregulated miR-215, miR-560, miR-615, miR-192, miR-326 and miR-147 and downregulated miR-100, miR-23, miR-605, miR-99, miR-205, let-7c and miR-203 [151]. MiR-101, miR-125, miR-197, miR-200 and miR-513 are upregulated, and miR-20, miR-23, miR-181, miR-193, miR-203 and miR-636 are downregulated in the progression from BE with LGD to BE with HGD [152]. MiR-28, miR-30 miR-126, miR-143, miR-145, miR-181 and miR-199 are upregulated, and Let-7, miR-193, miR-345 and miR-494 are downregulated in the progression from HGD to EAC [152]. MiR-25, miR-93 and miR-106b are overexpressed in EAC compared to BE [153]. MiR-200a is overexpressed in EAC compared to the normal esophagus. Its expression decreases with advanced stages of EAC, which suggests that miR-200a may be involved in the early phase of the development of EAC [154]. MiR-133b is downregulated in EAC compared to the normal esophagus [155]. Besides differential expression, targeted genes and pathways of a subset of miRNAs were investigated. MiR-133b may regulate the proliferation and apoptosis of EAC cells by targeting the pro-survival gene MCL-1 and the receptor tyrosine kinase MET signaling pathway [156,157]. Moreover, miR-21 is overexpressed in EAC and is thought to play a role in the initiation and development of EAC by disrupting apoptosis due to activation of Ras/MEK/ERK signaling and NF-κB signaling [155,158,159,160]. Our group studied several miRNAs in EAC and demonstrated that miR-125a reduced MHC-I expression, which is required for antigen presentation and T-cell response. Reduced MHC-1 expression and adaptive immune system markers were associated with improved patient outcomes in EAC [161].

3. Management of EAC

Summary: Effective clinical management of BE and EAC is essential to reduce EAC incidence and improve the quality of life and prognosis of EAC patients. In this part, we discuss the latest clinical advancements for the prevention and treatment of EAC. The implementation of CROSS and FLOT chemo(radio)therapy regimens brought benefits to the potentially resectable EAC patients, whereas almost no progress in the treatment for patients with distant metastatic EAC has been made. What studies should be performed to improve the treatment of EAC patients with distant metastasis?

3.1. Prevention of EAC in Patients with BE

Decreasing risk factors by maintaining an appropriate BMI, frequent intake of vegetables and fruit and prohibition of cigarette smoking are general lifestyle measures for the prevention of EAC. Endoscopy as a population screening method to detect patients with BE has not been recommended yet considering the relatively high costs and risks of complications [162]. This means that many cases of BE are not diagnosed, especially because a large part of the BE population never have GERD symptoms or have only mild symptoms until the advent of EAC. There is an unmet clinical need for screening tools in the general population to detect BE. Low-cost and low-risk methods such as non-endoscopic screening by sponge capsules and breath tests are under investigation [163,164].
Early diagnosis and successive monitoring of BE through endoscopic surveillance do play an effective role in the prevention of EAC. Surveillance programs are effective, and the incidence of invasive EAC is lower, while outcomes of EAC patients detected in BE surveillance cohorts are better [165]. Patients with BE undergoing endoscopic surveillance can be treated in the early curative stages of EAC and have significant survival benefits compared to EAC patients who have not been detected in endoscopic surveillance programs [166,167,168]. Besides endoscopic surveillance, chemo-preventive medicines can effectively reduce progression to EAC in BE patients. The use of proton pump inhibitors (PPIs) effectively reduced the risk of EAC progression in patients with BE [169,170,171]. Aspirin and other nonsteroidal anti-inflammatory drugs (NSAIDs) have been demonstrated to reduce the risk of EAC compared to controls [135,172,173,174,175]. Aspirin and NSAIDs both inhibit the generation of cyclooxygenase (COX) [176], and aspirin also inhibits the activity of NF-κB signaling [177].

3.2. Treatment of EAC

Patients with BE without dysplasia are included in long-term endoscopic surveillance programs and are treated with PPIs. BE patients with dysplasia may undergo endoscopic therapies, including photodynamic therapy, cryotherapy, radiofrequency ablation (RFA) and endoscopic mucosal resection (EMR). EMR combined with RFA is the main therapeutic strategy currently applied. One study showed that photodynamic therapy is highly effective for eradicating dysplastic BE, but it is associated with a relatively high rate of complications [178]. Cryotherapy displayed effective results as a treatment for BE [179] and enables eradication of intestinal metaplasia in 55% of patients, dysplasia in 85–90% and HGD in 95–100% [180]. RFA achieves complete eradication of dysplasia in 81–91% of patients and is characterized by low recurrence rates [181]. BE patients with early-stage cancers (tumor in situ, staged as T1a) either undergo endoscopic treatment or surgery [182].
Patients with EAC undergo tumor staging to select the most appropriate treatment regimen. Endoscopy with endoscopic ultrasound and computed tomography (CT) or positron emission tomography (PET) with [18F]2-fluoro-2-deoxy-d-glucose (FDG) are frequently used in the clinic [183]. Early, locally advanced EAC without distant lymph node involvement and distant metastasis can be amenable to treatment with curative intent. Neoadjuvant chemoradiotherapy followed by surgery (CROSS regimen) is applied in the clinic. Neoadjuvant chemoradiotherapy (nCRT) with the administration of carboplatin and paclitaxel for 5 weeks and concurrent radiotherapy (41.4 Gy in 23 fractions, 5 days per week) lead to a significant survival benefit compared to surgery alone [184]. Another successful and clinically applied perioperative chemoregimen is FLOT (fluorouracil plus leucovorin, oxaliplatin and docetaxel), which improves overall survival compared to perioperative ECF/ECX (epirubicin and cisplatin plus either fluorouracil or capecitabine) [185,186]. A comparison of CROSS and FLOT has also been investigated in clinical trials, with one study showing comparable survival benefits [187], while other studies are ongoing [188].
Regarding the surgical approach for EAC, radical transthoracic esophagectomy in combination with lymphadenectomy is the first choice of treatment [183]. In addition, minimally invasive esophagectomy (MIO), which is characterized by a lower post-operative morbidity, better quality of life and quicker functional recovery compared to open esophagectomy, has been introduced to clinical practice recently [189,190].
Definitive CRT is an option for those EAC patients who are not operable due to age or co-morbidities or are unwilling to undergo surgery [191]. For these therapies, adjusted CROSS and FOLFOX regimens or a regimen of fluorouracil plus cisplatin are frequently used [192].
For EAC patients with stage IV disease due to distant metastasis, palliative treatment with systemic therapy is currently the treatment of choice. This treatment includes fluoropyrimidine (fluorouracil or capecitabine) combined with either oxaliplatin or cisplatin and is a regimen according to the NCCN guidelines [193,194]. The FDA also approved Lonsurf (trifluridine/tipiracil) for the treatment of metastatic EAC. This is based on a phase III trial that showed that trifluridine/tipiracil significantly improves overall survival in metastatic EAC compared to a placebo group [195]. Platinum-fluoropyrimidine doublet (oxaliplatin and cisplatin plus fluoropyrimidines) can also be considered as a treatment for advanced and metastatic EAC [196].
As for now, only few targeted and immune therapies have been approved for the treatment of EAC (Table 6). HER2, VEGFR2 and PD-1 are the only three molecular targets. Given that currently employed targeted and immune therapies marginally improved the outcomes of patients, exploring more available molecular targets is still an imperative clinical need.

4. Discussion

The majority of treatments for EAC currently employed in the clinic are not specifically designed for EAC based on its biological features. Although the CROSS and FLOT regimens displayed better outcomes than other regimens, their establishment is derived from clinical trials and is less related to the unique molecular features of EAC. Chemoradiotherapy is a common approach for the treatment of most cancer types. The inherent non-specificity of chemoradiotherapy can cause severe side effects and discontinuation of the treatment. Also, it is thought that chemoresistance induced by chemoradiotherapy may accelerate the metastatic behavior of residual cancer cells. Nevertheless, the currently employed targeted and immune therapies play only an auxiliary role in the treatment of EAC. The therapeutics Trastuzumab (anti-HER2), Ramucirumab (anti-VEGFR2), Nivolumab (anti-PD1) and Pembrolizumab (anti-PD1), which are used in the clinic for EAC, are monoclonal antibodies, whose frequent side effects have been increasingly drawing the attention of researchers and clinicians and whose optimal alternatives are under investigation [203]. The potential therapeutic targets, for instance, the receptor tyrosine kinases EGFR, MET and FGFR in EAC, still hold a challenge due to their failed clinical trials [33]. For the prevention and treatment of EAC, there are certainly other methods. At present, endoscopy is the only surveillance method for BE in the clinic. Unfortunately, it has not been recommended for screening people with GERD or at risk for EAC as it is costly and invasive. Therefore, a subset of BE patients who develop EAC are missed during the optional phase of progression for effective endoscopic treatment. There is still a big gap between fundamental research and the ideal therapy for EAC. Therefore, the development of more effective therapies is an unmet medical need.
This work systematically summarized and extracted valuable findings from a large number of existing studies. As such, we hope to provide a framework for future research. This work might accelerate key research and the design of novel translational studies. On the molecular level, we discussed the most important factor associated with BE and EAC, which is GERD. GERD is known to erode and damage esophageal mucosa and induce BE. It also acts on the tumor microenvironment in EAC. However, the underlying comprehensive biological mechanisms of GERD on BE and EAC are less reported. Previous studies vaguely describe premalignant promotion as the main biological mechanism of GERD driving EAC without mentioning specific molecules and pathways [204]. We summarized that GERD acts by impacting both esophageal squamous epithelial cells and the tumor microenvironment, which includes important molecular and signaling pathways, such as CDX2, COX2, BMP4, MUC2, Dll1 and the NFkB pathway. In current clinical practice, chemopreventive medicines (Aspirin and NSAIDs) against COX2 and NFkB have been used for the preventive treatment of GERD and BE [176,177]. This review provides the rationale for future translational studies to focus on targeting CDX2, BMP4, MUC2 and Dll1; especially CDX2 and BMP4 are targets of interest, considering that MUC2 and Dll1 are inhibited in the case of inhibition of CDX2. Moreover, many studies have focused on the change and role of cytokines and chemokines in the tumor microenvironment of EAC. We found that the biological interaction between these factors in the tumor microenvironment is a complex network. For instance, IL-1β mainly secreted by M2 macrophages might upregulate BMP4 and CDX2 expression in esophageal epithelial cells and SHH expression in cancer-associated fibroblasts. BMP4, CDX2 and SHH are both involved in the pathogenesis of BE and EAC [77,205,206]. Therefore, targeting IL-1β or IL-1β-secreting cells is an interesting potential treatment strategy. Previous studies showed that EAC is characterized by an anti-inflammatory and immunosuppressive microenvironment with potential therapeutic targets within this microenvironment. FOXP3, as an immunosuppressive protein expressed in BE cells, promotes dysplasia and cell proliferation. FOXP3 is expressed in lymphocytes, especially in regulatory T cells, and its expression inhibits the immune system and reduces the destruction of cancer cells. Fortunately, an anti-FOXP3 inhibitor has been tested in a Phase I clinical trial, which showed promising anti-tumor effects [207]. Therefore, inhibition of FOXP3 could be an attractive approach to prevent and treat EAC. Tumor-associated macrophages are an essential determinant of the development and progression of tumors and display anti-tumor potential in other types of cancer [208]. In this review, we found that M1 macrophages act as Th1 effector cells and have tumor-inhibiting effects in the tumor microenvironment in EAC. Macrophage-centered therapy has been investigated in several types of cancer but not in EAC [209]. In the future, studies should focus on M1-promoting and M2-inhibiting polarization in the tumor microenvironment of EAC. Specifically, it should be determined which cytokines and chemokines are involved in the polarization of M1 or M2 macrophages to enable the development of corresponding inhibitors or activators.
There is a controversy about whether the cell of origin of BE originates from gland ducts or from the gastric cardia. The resolution of this controversy will be a milestone in better understanding the pathogenesis of BE and EAC in the future. The difficulty in the confirmation of the origin of BE is mainly attributed to the inability to observe metaplastic transformations in vivo due to a lack of reliable animal models [210]. Our group developed a mouse model resembling BE that regenerates the neo-columnar epithelium [211], which may accelerate the progression of this kind of study.
By summarizing alterations on the genetic and epigenetic level in EAC, we reviewed the potential clinically applicable biomarkers to complement the insufficiency of histopathological grading of EAC. However, this is a difficulty for EAC considering that the genetics of EAC is characterized by a high mutational burden and a variety of mutational signatures, complemented by a high level of chromosomal instability. Also, the epigenetics of EAC is featured by massive microRNA variations and various DNA hypermethylations. In light of this, the purpose of precision treatment through carrying out more accurate gene stratification might be beneficial for the treatment of EAC. In different studies, it has been demonstrated that EAC can be divided into various molecular subgroups. One subclassification of EAC can be used to predict response to neoadjuvant treatment for EAC patients [143]. However, the subclassifications in several studies seem to have less clinical significance [108,123]. The definition of the molecular subtypes of EAC will be a milestone in the future. By reviewing the genetic and epigenetic studies, TP53, SMAD4 and CDKN2A are concluded to be biomarkers with more potential for the prediction of prognosis and treatment of EAC than other mutated genes as they are the most reported mutated genes. Studies reported that TP53 and SMAD4 are both prognostic factors, which can predict survival time or response to neoadjuvant therapy in EAC [126,212], whereas no study has reported CDKN2A as a prognostic factor in EAC. Other mutated genes discovered by one or few studies need to be further investigated. Besides detection methods and cohort size, mixtures with non-tumor cells (for instance, stromal cells and immune cells) in biopsies or resected specimens is an important factor that causes inconsistent findings between sequencing or gene expression data. Inconsistent gene profiling data could impact the identification of subgroups and the assignment of more suitable tailor-made therapies. This should be handled, for instance, by cell sorting or deconvolution methods including the implementation of single-cell sequencing. Epigenetic changes vary with each distinct phase of BE, LGD, HGD, early EAC and late EAC. New biomarkers that can be developed based on specific epigenetic changes will be suitable for patients in the specific phase of the disease. We found that some studies reported on genomic abnormalities as observed in single samples taken at one location and at one time point and neglected the temporal and spatial differences that are present in BE and EAC. Cancer studies of slow progressive precancerous lesions require longitudinal studies considering sufficient sample size, reasonable negative control populations and tissue sampling with follow-up that is synchronized with disease progression. Such studies may be more effective in revealing potential druggable targets compared to cross-sectional studies which are prone to bias due to patient heterogeneity.

5. Conclusions

In this work, we revealed up-to-date knowledge of biological characteristics and treatment strategies of esophageal adenocarcinoma, including molecular pathogenesis, tumor microenvironment, genetics, epigenetics and therapy advancement. Importantly, we discussed the questions that arose from these studies and the clinical practice and the scientific milestones, which may be useful to improve EAC prevention and treatment in the future.

Author Contributions

Conceptualization, S.L. and K.K.K.; methodology, S.L., S.J.M.H. and K.K.K.; writing—original draft preparation, S.L.; writing—review and editing, S.J.M.H. and K.K.K.; visualization, S.L.; supervision, S.J.M.H. and K.K.K.; project administration, K.K.K.; funding acquisition, S.L. and K.K.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the European Research Council, grant number ERC-POC 632258; Dutch government grant, grant number LSH-TKI-PPP 2017; China Scholarship Council, grant number 201806170097.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. The Cancer Genome Atlas Research Network. Integrated genomic characterization of oesophageal carcinoma. Nature 2017, 541, 169–175. [Google Scholar] [CrossRef] [PubMed]
  2. Dikken, J.L.; Lemmens, V.E.; Wouters, M.W.; Wijnhoven, B.P.; Siersema, P.D.; Nieuwenhuijzen, G.A.; van Sandick, J.W.; Cats, A.; Verheij, M.; Coebergh, J.W.; et al. Increased incidence and survival for oesophageal cancer but not for gastric cardia cancer in the Netherlands. Eur. J. Cancer 2012, 48, 1624–1632. [Google Scholar] [CrossRef] [PubMed]
  3. Gibbs, J.F.; Rajput, A.; Chadha, K.S.; Douglas, W.G.; Hill, H.; Nwogu, C.; Nava, H.R.; Sabel, M.S. The changing profile of esophageal cancer presentation and its implication for diagnosis. J. Natl. Med. Assoc. 2007, 99, 620–626. [Google Scholar] [PubMed]
  4. Coleman, H.G.; Xie, S.H.; Lagergren, J. The Epidemiology of Esophageal Adenocarcinoma. Gastroenterology 2018, 154, 390–405. [Google Scholar] [CrossRef] [PubMed]
  5. Schumacher, S.; Bartenhagen, C.; Hoffmann, M.; Will, D.; Fischer, J.C.; Baldus, S.E.; Vay, C.; Fluegen, G.; Dizdar, L.; Vallbohmer, D.; et al. Disseminated tumour cells with highly aberrant genomes are linked to poor prognosis in operable oesophageal adenocarcinoma. Br. J. Cancer 2017, 117, 725–733. [Google Scholar] [CrossRef] [PubMed]
  6. Lagergren, J.; Bergstrom, R.; Lindgren, A.; Nyren, O. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N. Engl. J. Med. 1999, 340, 825–831. [Google Scholar] [CrossRef] [PubMed]
  7. Ma, S.; Zhou, B.; Yang, Q.; Pan, Y.; Yang, W.; Freedland, S.J.; Ding, L.W.; Freeman, M.R.; Breunig, J.J.; Bhowmick, N.A.; et al. A Transcriptional Regulatory Loop of Master Regulator Transcription Factors, PPARG, and Fatty Acid Synthesis Promotes Esophageal Adenocarcinoma. Cancer Res. 2021, 81, 1216–1229. [Google Scholar] [CrossRef]
  8. Quante, M.; Graham, T.A.; Jansen, M. Insights Into the Pathophysiology of Esophageal Adenocarcinoma. Gastroenterology 2018, 154, 406–420. [Google Scholar] [CrossRef]
  9. Sharma, N.; Ho, K.Y. Risk Factors for Barrett’s Oesophagus. Gastrointest. Tumors 2016, 3, 103–108. [Google Scholar] [CrossRef]
  10. Gregson, E.M.; Bornschein, J.; Fitzgerald, R.C. Genetic progression of Barrett’s oesophagus to oesophageal adenocarcinoma. Br. J. Cancer 2016, 115, 403–410. [Google Scholar] [CrossRef]
  11. Li, X.; Galipeau, P.C.; Paulson, T.G.; Sanchez, C.A.; Arnaudo, J.; Liu, K.; Sather, C.L.; Kostadinov, R.L.; Odze, R.D.; Kuhner, M.K.; et al. Temporal and spatial evolution of somatic chromosomal alterations: A case-cohort study of Barrett’s esophagus. Cancer Prev. Res. 2014, 7, 114–127. [Google Scholar] [CrossRef] [PubMed]
  12. Corley, D.A.; Mehtani, K.; Quesenberry, C.; Zhao, W.; de Boer, J.; Weiss, N.S. Impact of endoscopic surveillance on mortality from Barrett’s esophagus-associated esophageal adenocarcinomas. Gastroenterology 2013, 145, 312–319.e311. [Google Scholar] [CrossRef] [PubMed]
  13. Joseph, A.; Raja, S.; Kamath, S.; Jang, S.; Allende, D.; McNamara, M.; Videtic, G.; Murthy, S.; Bhatt, A. Esophageal adenocarcinoma: A dire need for early detection and treatment. Cleve. Clin. J. Med. 2022, 89, 269–279. [Google Scholar] [CrossRef]
  14. Eyck, B.M.; van Lanschot, J.J.B.; Hulshof, M.; van der Wilk, B.J.; Shapiro, J.; van Hagen, P.; van Berge Henegouwen, M.I.; Wijnhoven, B.P.L.; van Laarhoven, H.W.M.; Nieuwenhuijzen, G.A.P.; et al. Ten-Year Outcome of Neoadjuvant Chemoradiotherapy Plus Surgery for Esophageal Cancer: The Randomized Controlled CROSS Trial. J. Clin. Oncol. 2021, 39, 1995–2004. [Google Scholar] [CrossRef]
  15. Bang, Y.J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet 2010, 376, 687–697. [Google Scholar] [CrossRef] [PubMed]
  16. Valkema, M.J.; Mostert, B.; Lagarde, S.M.; Wijnhoven, B.P.L.; van Lanschot, J.J.B. The effectivity of targeted therapy and immunotherapy in patients with advanced metastatic and non-metastatic cancer of the esophagus and esophago-gastric junction. Updates Surg. 2023, 75, 313–323. [Google Scholar] [CrossRef] [PubMed]
  17. Jin, Z.; Yoon, H.H. The promise of PD-1 inhibitors in gastro-esophageal cancers: Microsatellite instability vs. PD-L1. J. Gastrointest. Oncol. 2016, 7, 771–788. [Google Scholar] [CrossRef] [PubMed]
  18. Morgan, E.; Soerjomataram, I.; Rumgay, H.; Coleman, H.G.; Thrift, A.P.; Vignat, J.; Laversanne, M.; Ferlay, J.; Arnold, M. The Global Landscape of Esophageal Squamous Cell Carcinoma and Esophageal Adenocarcinoma Incidence and Mortality in 2020 and Projections to 2040: New Estimates From GLOBOCAN 2020. Gastroenterology 2022, 163, 649–658.e642. [Google Scholar] [CrossRef]
  19. Hang, T.P.; Spiritos, Z.; Gamboa, A.M.; Chen, Z.; Force, S.; Patel, V.; Chawla, S.; Keilin, S.; Saba, N.F.; El-Rayes, B.; et al. Epidemiology of early esophageal adenocarcinoma. Clin. Endosc. 2022, 55, 372–380. [Google Scholar] [CrossRef]
  20. Strauss, A.; Min, E.J.; Long, Q.; Gabriel, P.; Yang, Y.X.; Falk, G.W. Is the age of diagnosis of esophageal adenocarcinoma getting younger? Analysis at a tertiary care center. Dis. Esophagus 2020, 33, doz112. [Google Scholar] [CrossRef]
  21. Cook, M.B.; Chow, W.H.; Devesa, S.S. Oesophageal cancer incidence in the United States by race, sex, and histologic type, 1977–2005. Br. J. Cancer 2009, 101, 855–859. [Google Scholar] [CrossRef] [PubMed]
  22. To, H.; Clemons, N.J.; Duong, C.P.; Trainer, A.H.; Phillips, W.A. The Genetics of Barrett’s Esophagus: A Familial and Population-Based Perspective. Dig. Dis. Sci. 2016, 61, 1826–1834. [Google Scholar] [CrossRef] [PubMed]
  23. Verbeek, R.E.; Spittuler, L.F.; Peute, A.; van Oijen, M.G.; Ten Kate, F.J.; Vermeijden, J.R.; Oberndorff, A.; van Baal, J.W.; Siersema, P.D. Familial clustering of Barrett’s esophagus and esophageal adenocarcinoma in a European cohort. Clin. Gastroenterol. Hepatol. 2014, 12, 1656–1663.e1651. [Google Scholar] [CrossRef] [PubMed]
  24. Fecteau, R.E.; Kong, J.; Kresak, A.; Brock, W.; Song, Y.; Fujioka, H.; Elston, R.; Willis, J.E.; Lynch, J.P.; Markowitz, S.D.; et al. Association Between Germline Mutation in VSIG10L and Familial Barrett Neoplasia. JAMA Oncol. 2016, 2, 1333–1339. [Google Scholar] [CrossRef] [PubMed]
  25. Buas, M.F.; He, Q.; Johnson, L.G.; Onstad, L.; Levine, D.M.; Thrift, A.P.; Gharahkhani, P.; Palles, C.; Lagergren, J.; Fitzgerald, R.C.; et al. Germline variation in inflammation-related pathways and risk of Barrett’s oesophagus and oesophageal adenocarcinoma. Gut 2017, 66, 1739–1747. [Google Scholar] [CrossRef] [PubMed]
  26. Dai, J.Y.; de Dieu Tapsoba, J.; Buas, M.F.; Onstad, L.E.; Levine, D.M.; Risch, H.A.; Chow, W.H.; Bernstein, L.; Ye, W.; Lagergren, J.; et al. A newly identified susceptibility locus near FOXP1 modifies the association of gastroesophageal reflux with Barrett’s esophagus. Cancer Epidemiol. Biomark. Prev. 2015, 24, 1739–1747. [Google Scholar] [CrossRef] [PubMed]
  27. MacInnis, R.J.; English, D.R.; Hopper, J.L.; Giles, G.G. Body size and composition and the risk of gastric and oesophageal adenocarcinoma. Int. J. Cancer 2006, 118, 2628–2631. [Google Scholar] [CrossRef] [PubMed]
  28. Whiteman, D.C.; Sadeghi, S.; Pandeya, N.; Smithers, B.M.; Gotley, D.C.; Bain, C.J.; Webb, P.M.; Green, A.C.; Australian Cancer, S. Combined effects of obesity, acid reflux and smoking on the risk of adenocarcinomas of the oesophagus. Gut 2008, 57, 173–180. [Google Scholar] [CrossRef]
  29. Corley, D.A.; Kubo, A.; Levin, T.R.; Block, G.; Habel, L.; Zhao, W.; Leighton, P.; Quesenberry, C.; Rumore, G.J.; Buffler, P.A. Abdominal obesity and body mass index as risk factors for Barrett’s esophagus. Gastroenterology 2007, 133, 34–41. [Google Scholar] [CrossRef]
  30. Edelstein, Z.R.; Farrow, D.C.; Bronner, M.P.; Rosen, S.N.; Vaughan, T.L. Central adiposity and risk of Barrett’s esophagus. Gastroenterology 2007, 133, 403–411. [Google Scholar] [CrossRef]
  31. El-Serag, H.B.; Kvapil, P.; Hacken-Bitar, J.; Kramer, J.R. Abdominal obesity and the risk of Barrett’s esophagus. Am. J. Gastroenterol. 2005, 100, 2151–2156. [Google Scholar] [CrossRef] [PubMed]
  32. Fitzgerald, R.C.; Abdalla, S.; Onwuegbusi, B.A.; Sirieix, P.; Saeed, I.T.; Burnham, W.R.; Farthing, M.J. Inflammatory gradient in Barrett’s oesophagus: Implications for disease complications. Gut 2002, 51, 316–322. [Google Scholar] [CrossRef]
  33. Smyth, E.C.; Lagergren, J.; Fitzgerald, R.C.; Lordick, F.; Shah, M.A.; Lagergren, P.; Cunningham, D. Oesophageal cancer. Nat. Rev. Dis. Primers 2017, 3, 17048. [Google Scholar] [CrossRef] [PubMed]
  34. Munch, N.S.; Fang, H.Y.; Ingermann, J.; Maurer, H.C.; Anand, A.; Kellner, V.; Sahm, V.; Wiethaler, M.; Baumeister, T.; Wein, F.; et al. High-Fat Diet Accelerates Carcinogenesis in a Mouse Model of Barrett’s Esophagus via Interleukin 8 and Alterations to the Gut Microbiome. Gastroenterology 2019, 157, 492–506.e492. [Google Scholar] [CrossRef]
  35. Reid, B.J.; Li, X.; Galipeau, P.C.; Vaughan, T.L. Barrett’s oesophagus and oesophageal adenocarcinoma: Time for a new synthesis. Nat. Rev. Cancer 2010, 10, 87–101. [Google Scholar] [CrossRef] [PubMed]
  36. Arnold, M.; Laversanne, M.; Brown, L.M.; Devesa, S.S.; Bray, F. Predicting the Future Burden of Esophageal Cancer by Histological Subtype: International Trends in Incidence up to 2030. Am. J. Gastroenterol. 2017, 112, 1247–1255. [Google Scholar] [CrossRef] [PubMed]
  37. Correia, A.C.P.; Straub, D.; Read, M.; Hoefnagel, S.J.M.; Romero-Pinedo, S.; Abadia-Molina, A.C.; Clemons, N.J.; Wang, K.; Calpe, S.; Phillips, W.; et al. Inhibition of BMP2 and BMP4 Represses Barrett’s Esophagus While Enhancing the Regeneration of Squamous Epithelium in Preclinical Models. Cell. Mol. Gastroenterol. Hepatol. 2023, 15, 1199–1217. [Google Scholar] [CrossRef] [PubMed]
  38. Lin, E.C.; Holub, J.; Lieberman, D.; Hur, C. Low Prevalence of Suspected Barrett’s Esophagus in Patients With Gastroesophageal Reflux Disease Without Alarm Symptoms. Clin. Gastroenterol. Hepatol. 2019, 17, 857–863. [Google Scholar] [CrossRef]
  39. Rajendra, S. Diagnosis and Management of Barrett’s Esophagus: An Updated ACG Guideline. Am. J. Gastroenterol. 2022, 117, 1880. [Google Scholar] [CrossRef]
  40. Quante, M.; Bhagat, G.; Abrams, J.A.; Marache, F.; Good, P.; Lee, M.D.; Lee, Y.; Friedman, R.; Asfaha, S.; Dubeykovskaya, Z.; et al. Bile acid and inflammation activate gastric cardia stem cells in a mouse model of Barrett-like metaplasia. Cancer Cell 2012, 21, 36–51. [Google Scholar] [CrossRef]
  41. Sawas, T.; Katzka, D.A. Esophageal adenocarcinoma phenotypes and risk factors. Curr. Opin. Gastroenterol. 2022, 38, 423–427. [Google Scholar] [CrossRef] [PubMed]
  42. Sawas, T.; Killcoyne, S.; Iyer, P.G.; Wang, K.K.; Smyrk, T.C.; Kisiel, J.B.; Qin, Y.; Ahlquist, D.A.; Rustgi, A.K.; Costa, R.J.; et al. Identification of Prognostic Phenotypes of Esophageal Adenocarcinoma in 2 Independent Cohorts. Gastroenterology 2018, 155, 1720–1728.e1724. [Google Scholar] [CrossRef] [PubMed]
  43. Nowicki-Osuch, K.; Zhuang, L.; Jammula, S.; Bleaney, C.W.; Mahbubani, K.T.; Devonshire, G.; Katz-Summercorn, A.; Eling, N.; Wilbrey-Clark, A.; Madissoon, E.; et al. Molecular phenotyping reveals the identity of Barrett’s esophagus and its malignant transition. Science 2021, 373, 760–767. [Google Scholar] [CrossRef] [PubMed]
  44. Straub, D.; Elferink, R.P.J.O.; Jansen, P.L.M.; Bergman, J.J.G.H.M.; Parikh, K.; Krishnadath, K.K. Glyco-conjugated bile acids drive the initial metaplastic gland formation from multi-layered glands through crypt-fission in a murine model. PLoS ONE 2019, 14, e0220050. [Google Scholar] [CrossRef] [PubMed]
  45. Minacapelli, C.D.; Bajpai, M.; Geng, X.; Cheng, C.L.; Chouthai, A.A.; Souza, R.; Spechler, S.J.; Das, K.M. Barrett’s metaplasia develops from cellular reprograming of esophageal squamous epithelium due to gastroesophageal reflux. Am. J. Physiol-Gastr. Liver Physiol. 2017, 312, G615–G622. [Google Scholar] [CrossRef] [PubMed]
  46. Jiang, M.; Li, H.Y.; Zhang, Y.C.; Yang, Y.; Lu, R.; Liu, K.C.; Lin, S.J.; Lan, X.P.; Wang, H.K.; Wu, H.; et al. Transitional basal cells at the squamous-columnar junction generate Barrett’s oesophagus. Nature 2017, 550, 529–533. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, X.; Ouyang, H.; Yamamoto, Y.; Kumar, P.A.; Wei, T.S.; Dagher, R.; Vincent, M.; Lu, X.; Bellizzi, A.M.; Ho, K.Y.; et al. Residual embryonic cells as precursors of a Barrett’s-like metaplasia. Cell 2011, 145, 1023–1035. [Google Scholar] [CrossRef]
  48. Leedham, S.J.; Preston, S.L.; McDonald, S.A.; Elia, G.; Bhandari, P.; Poller, D.; Harrison, R.; Novelli, M.R.; Jankowski, J.A.; Wright, N.A. Individual crypt genetic heterogeneity and the origin of metaplastic glandular epithelium in human Barrett’s oesophagus. Gut 2008, 57, 1041–1048. [Google Scholar] [CrossRef]
  49. Desai, T.K.; Krishnan, K.; Samala, N.; Singh, J.; Cluley, J.; Perla, S.; Howden, C.W. The incidence of oesophageal adenocarcinoma in non-dysplastic Barrett’s oesophagus: A meta-analysis. Gut 2012, 61, 970–976. [Google Scholar] [CrossRef]
  50. Hvid-Jensen, F.; Pedersen, L.; Drewes, A.M.; Sorensen, H.T.; Funch-Jensen, P. Incidence of Adenocarcinoma among Patients with Barrett’s Esophagus. N. Engl. J. Med. 2011, 365, 1375–1383. [Google Scholar] [CrossRef]
  51. Vennalaganti, P.R.; Kanakadandi, V.N.; Gross, S.A.; Parasa, S.; Wang, K.K.; Gupta, N.; Sharma, P. Inter-Observer Agreement among Pathologists Using Wide-Area Transepithelial Sampling With Computer-Assisted Analysis in Patients With Barrett’s Esophagus. Am. J. Gastroenterol. 2015, 110, 1257–1260. [Google Scholar] [CrossRef] [PubMed]
  52. Curvers, W.L.; ten Kate, F.J.; Krishnadath, K.K.; Visser, M.; Elzer, B.; Baak, L.C.; Bohmer, C.; Mallant-Hent, R.C.; van Oijen, A.; Naber, A.H.; et al. Low-grade dysplasia in Barrett’s esophagus: Overdiagnosed and underestimated. Am. J. Gastroenterol. 2010, 105, 1523–1530. [Google Scholar] [CrossRef] [PubMed]
  53. Wani, S.; Falk, G.W.; Post, J.; Yerian, L.; Hall, M.; Wang, A.; Gupta, N.; Gaddam, S.; Singh, M.; Singh, V.; et al. Risk factors for progression of low-grade dysplasia in patients with Barrett’s esophagus. Gastroenterology 2011, 141, 1179–1186.e1171. [Google Scholar] [CrossRef] [PubMed]
  54. Falk, G.W. Current Management of Low-Grade Dysplasia in Barrett Esophagus. Gastroenterol. Hepatol. 2017, 13, 221–225. [Google Scholar]
  55. Hoefnagel, S.J.M.; Mostafavi, N.; Timmer, M.R.; Lau, C.T.; Meijer, S.L.; Wang, K.K.; Krishnadath, K.K. A genomic biomarker-based model for cancer risk stratification of non-dysplastic Barrett’s esophagus patients after extended follow up; results from Dutch surveillance cohorts. PLoS ONE 2020, 15, e0231419. [Google Scholar] [CrossRef]
  56. Falk, G.W. Barrett’s oesophagus: Frequency and prediction of dysplasia and cancer. Best. Pract. Res. Clin. Gastroenterol. 2015, 29, 125–138. [Google Scholar] [CrossRef]
  57. Chandrasekar, V.T.; Hamade, N.; Desai, M.; Rai, T.; Gorrepati, V.S.; Jegadeesan, R.; Sathyamurthy, A.; Sharma, P. Significantly lower annual rates of neoplastic progression in short- compared to long-segment non-dysplastic Barrett’s esophagus: A systematic review and meta-analysis. Endoscopy 2019, 51, 665–672. [Google Scholar] [CrossRef]
  58. Lam, A.K. Updates on World Health Organization classification and staging of esophageal tumors: Implications for future clinical practice. Hum. Pathol. 2021, 108, 100–112. [Google Scholar] [CrossRef]
  59. Fiocca, R.; Mastracci, L.; Lugaresi, M.; Grillo, F.; D’Errico, A.; Malvi, D.; Spaggiari, P.; Tomezzoli, A.; Albarello, L.; Ristimaki, A.; et al. The Prognostic Impact of Histology in Esophageal and Esophago-Gastric Junction Adenocarcinoma. Cancers 2021, 13, 5211. [Google Scholar] [CrossRef]
  60. Jimenez Fonseca, P.; Carmona-Bayonas, A.; Hernandez, R.; Custodio, A.; Cano, J.M.; Lacalle, A.; Echavarria, I.; Macias, I.; Mangas, M.; Visa, L.; et al. Lauren subtypes of advanced gastric cancer influence survival and response to chemotherapy: Real-world data from the AGAMENON National Cancer Registry. Br. J. Cancer 2017, 117, 775–782. [Google Scholar] [CrossRef]
  61. van der Kaaij, R.T.; Snaebjornsson, P.; Voncken, F.E.; van Dieren, J.M.; Jansen, E.P.; Sikorska, K.; Cats, A.; van Sandick, J.W. The prognostic and potentially predictive value of the Lauren classification in oesophageal adenocarcinoma. Eur. J. Cancer 2017, 76, 27–35. [Google Scholar] [CrossRef] [PubMed]
  62. Lam, A.; Kumarasinghe, M. Adenocarcinoma of the oesophagus and oesophagogastric junction NOS. WHO Classif. Tumours 2019, 38–43. [Google Scholar]
  63. Nagtegaal, I.D.; Odze, R.D.; Klimstra, D.; Paradis, V.; Rugge, M.; Schirmacher, P.; Washington, K.M.; Carneiro, F.; Cree, I.A.; the WHO Classification of Tumours Editorial Board. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020, 76, 182–188. [Google Scholar] [CrossRef] [PubMed]
  64. Huo, X.; Zhang, X.; Yu, C.; Zhang, Q.; Cheng, E.; Wang, D.H.; Pham, T.H.; Spechler, S.J.; Souza, R.F. In oesophageal squamous cells exposed to acidic bile salt medium, omeprazole inhibits IL-8 expression through effects on nuclear factor-kappaB and activator protein-1. Gut 2014, 63, 1042–1052. [Google Scholar] [CrossRef] [PubMed]
  65. Huo, X.F.; Zhang, H.Y.; Zhang, X.I.; Lynch, J.P.; Strauch, E.D.; Wang, J.Y.; Melton, S.D.; Genta, R.M.; Wang, D.H.; Spechler, S.J.; et al. Acid and Bile Salt-Induced CDX2 Expression Differs in Esophageal Squamous Cells From Patients With and Without Barrett’s Esophagus. Gastroenterology 2010, 139, 194–203. [Google Scholar] [CrossRef] [PubMed]
  66. O’Riordan, J.M.; Abdel-latif, M.M.; Ravi, N.; McNamara, D.; Byrne, P.J.; McDonald, G.S.A.; Keeling, P.W.N.; Kelleher, D.; Reynolds, J.V. Proinflammatory cytokine and nuclear factor kappa-B expression along the inflammation-metaplasia-dysplasia-adenocarcinoma sequence in the esophagus. Am. J. Gastroenterol. 2005, 100, 1257–1264. [Google Scholar] [CrossRef] [PubMed]
  67. Xia, L.; Tan, S.; Zhou, Y.; Lin, J.; Wang, H.; Oyang, L.; Tian, Y.; Liu, L.; Su, M.; Wang, H.; et al. Role of the NFkappaB-signaling pathway in cancer. Onco. Targets. Ther. 2018, 11, 2063–2073. [Google Scholar] [CrossRef] [PubMed]
  68. Abdel-Latif, M.M.; O’Riordan, J.; Windle, H.J.; Carton, E.; Ravi, N.; Kelleher, D.; Reynolds, J.V. NF-kappaB activation in esophageal adenocarcinoma: Relationship to Barrett’s metaplasia, survival, and response to neoadjuvant chemoradiotherapy. Ann. Surg. 2004, 239, 491–500. [Google Scholar] [CrossRef]
  69. Eda, A.; Osawa, H.; Satoh, K.; Yanaka, I.; Kihira, K.; Ishino, Y.; Mutoh, H.; Sugano, K. Aberrant expression of CDX2 in Barrett’s epithelium and inflammatory esophageal mucosa. J. Gastroenterol. 2003, 38, 14–22. [Google Scholar] [CrossRef]
  70. Groisman, G.M.; Amar, M.; Meir, A. Expression of the intestinal marker Cdx2 in the columnar-lined esophagus with and without intestinal (Barrett’s) metaplasia. Mod. Pathol. 2004, 17, 1282–1288. [Google Scholar] [CrossRef]
  71. Phillips, R.W.; Frierson, H.F., Jr.; Moskaluk, C.A. Cdx2 as a marker of epithelial intestinal differentiation in the esophagus. Am. J. Surg. Pathol. 2003, 27, 1442–1447. [Google Scholar] [CrossRef] [PubMed]
  72. Vallbohmer, D.; DeMeester, S.R.; Peters, J.H.; Oh, D.S.; Kuramochi, H.; Shimizu, D.; Hagen, J.A.; Danenberg, K.D.; Danenberg, P.V.; DeMeester, T.R.; et al. Cdx-2 expression in squamous and metaplastic columnar epithelia of the esophagus. Dis. Esophagus 2006, 19, 260–266. [Google Scholar] [CrossRef] [PubMed]
  73. Kazumori, H.; Ishihara, S.; Rumi, M.A.; Kadowaki, Y.; Kinoshita, Y. Bile acids directly augment caudal related homeobox gene Cdx2 expression in oesophageal keratinocytes in Barrett’s epithelium. Gut 2006, 55, 16–25. [Google Scholar] [CrossRef] [PubMed]
  74. Tamagawa, Y.; Ishimura, N.; Uno, G.; Aimi, M.; Oshima, N.; Yuki, T.; Sato, S.; Ishihara, S.; Kinoshita, Y. Bile acids induce Delta-like 1 expression via Cdx2-dependent pathway in the development of Barrett’s esophagus. Lab. Investig. 2016, 96, 325–337. [Google Scholar] [CrossRef] [PubMed]
  75. Mari, L.; Milano, F.; Parikh, K.; Straub, D.; Everts, V.; Hoeben, K.K.; Fockens, P.; Buttar, N.S.; Krishnadath, K.K. A pSMAD/CDX2 Complex Is Essential for the Intestinalization of Epithelial Metaplasia. Cell Rep. 2014, 7, 1197–1210. [Google Scholar] [CrossRef] [PubMed]
  76. Milano, F.; van Baal, J.W.; Buttar, N.S.; Rygiel, A.M.; de Kort, F.; DeMars, C.J.; Rosmolen, W.D.; Bergman, J.J.; van Marle, J.; Wang, K.K.; et al. Bone morphogenetic protein 4 expressed in esophagitis induces a columnar phenotype in esophageal squamous cells. Gastroenterology 2007, 132, 2412–2421. [Google Scholar] [CrossRef]
  77. Li, S.; Hoefnagel, S.J.M.; Read, M.; Meijer, S.; van Berge Henegouwen, M.I.; Gisbertz, S.S.; Bonora, E.; Liu, D.S.H.; Phillips, W.A.; Calpe, S.; et al. Selective targeting BMP2 and 4 in SMAD4 negative esophageal adenocarcinoma inhibits tumor growth and aggressiveness in preclinical models. Cell. Oncol. 2022, 45, 639–658. [Google Scholar] [CrossRef] [PubMed]
  78. Abdel-Latif, M.M.; Kelleher, D.; Reynolds, J.V. Potential role of NF-kappaB in esophageal adenocarcinoma: As an emerging molecular target. J. Surg. Res. 2009, 153, 172–180. [Google Scholar] [CrossRef]
  79. Shirvani, V.N.; Ouatu-Lascar, R.; Kaur, B.S.; Omary, M.B.; Triadafilopoulos, G. Cyclooxygenase 2 expression in Barrett’s esophagus and adenocarcinoma: Ex vivo induction by bile salts and acid exposure. Gastroenterology 2000, 118, 487–496. [Google Scholar] [CrossRef]
  80. Souza, R.F.; Shewmake, K.; Beer, D.G.; Cryer, B.; Spechler, S.J. Selective inhibition of cyclooxygenase-2 suppresses growth and induces apoptosis in human esophageal adenocarcinoma cells. Cancer Res. 2000, 60, 5767–5772. [Google Scholar]
  81. Ferguson, H.R.; Wild, C.P.; Anderson, L.A.; Murphy, S.J.; Johnston, B.T.; Murray, L.J.; Watson, R.G.; McGuigan, J.; Reynolds, J.V.; Hardie, L.J. Cyclooxygenase-2 and inducible nitric oxide synthase gene polymorphisms and risk of reflux esophagitis, Barrett’s esophagus, and esophageal adenocarcinoma. Cancer Epidemiol. Biomark. Prev. 2008, 17, 727–731. [Google Scholar] [CrossRef] [PubMed]
  82. Moons, L.M.G.; Kuipers, E.J.; Rygiel, A.M.; Groothuismink, A.Z.M.; Geldof, H.; Bode, W.A.; Krishnadath, K.K.; Bergman, J.J.G.H.M.; van Vliet, A.H.M.; Siersema, P.D.; et al. COX-2 CA-haplotype is a risk factor for the development of esophageal adenocarcinoma. Am. J. Gastroenterol. 2007, 102, 2373–2379. [Google Scholar] [CrossRef] [PubMed]
  83. Buttar, N.S.; Wang, K.K.; Leontovich, O.; Westcott, J.Y.; Pacifico, R.J.; Anderson, M.A.; Krishnadath, K.K.; Lutzke, L.S.; Burgart, L.J. Chemoprevention of esophageal adenocarcinoma by COX-2 inhibitors in an animal model of Barrett’s esophagus. Gastroenterology 2002, 122, 1101–1112. [Google Scholar] [CrossRef] [PubMed]
  84. Neureiter, D.; Mayr, C.; Winkelmann, P.; Neumayer, B.; Klieser, E.; Wagner, A.; Hufnagl, C.; Emmanuel, K.; Holzinger, J.; Koch, O.; et al. Expression of the microRNA-200 Family, microRNA-205, and Markers of Epithelial-Mesenchymal Transition as Predictors for Endoscopic Submucosal Dissection over Esophagectomy in Esophageal Adenocarcinoma: A Single-Center Experience. Cells 2020, 9, 486. [Google Scholar] [CrossRef] [PubMed]
  85. Fang, H.Y.; Munch, N.S.; Schottelius, M.; Ingermann, J.; Liu, H.B.; Schauer, M.; Stangl, S.; Multhoff, G.; Steiger, K.; Gerngross, C.; et al. CXCR4 Is a Potential Target for Diagnostic PET/CT Imaging in Barrett’s Dysplasia and Esophageal Adenocarcinoma. Clin. Cancer Res. 2018, 24, 1048–1061. [Google Scholar] [CrossRef] [PubMed]
  86. Milano, F.; Jorritsma, T.; Rygiel, A.M.; Bergman, J.J.; Sondermeijer, C.; Ten Brinke, A.; vanHam, S.M.; Krishnadath, K.K. Expression pattern of immune suppressive cytokines and growth factors in oesophageal adenocarcinoma reveal a tumour immune escape-promoting microenvironment. Scand. J. Immunol. 2008, 68, 616–623. [Google Scholar] [CrossRef] [PubMed]
  87. Oh, D.S.; DeMeester, S.R.; Vallbohmer, D.; Mori, R.; Kuramochi, H.; Hagen, J.A.; Lipham, J.; Danenberg, K.D.; Danenberg, P.V.; Chandrasoma, P.; et al. Reduction of interleukin 8 gene expression in reflux esophagitis and Barrett’s esophagus with antireflux surgery. Arch. Surg. 2007, 142, 554–559; discussion 559–560. [Google Scholar] [CrossRef]
  88. Karstens, K.F.; Kempski, J.; Giannou, A.D.; Pelczar, P.; Steglich, B.; Steurer, S.; Freiwald, E.; Woestemeier, A.; Konczalla, L.; Tachezy, M.; et al. Anti-inflammatory microenvironment of esophageal adenocarcinomas negatively impacts survival. Cancer Immunol. Immunother. 2020, 69, 1043–1056. [Google Scholar] [CrossRef]
  89. Ebbing, E.A.; van der Zalm, A.P.; Steins, A.; Creemers, A.; Hermsen, S.; Rentenaar, R.; Klein, M.; Waasdorp, C.; Hooijer, G.K.J.; Meijer, S.L.; et al. Stromal-derived interleukin 6 drives epithelial-to-mesenchymal transition and therapy resistance in esophageal adenocarcinoma. Proc. Natl. Acad. Sci. USA 2019, 116, 2237–2242. [Google Scholar] [CrossRef]
  90. Gockel, I.; Schimanski, C.C.; Heinrich, C.; Wehler, T.; Frerichs, K.; Drescher, D.; von Langsdorff, C.; Domeyer, M.; Biesterfeld, S.; Galle, P.R.; et al. Expression of chemokine receptor CXCR4 in esophageal squamous cell and adenocarcinoma. BMC Cancer 2006, 6, 290. [Google Scholar] [CrossRef]
  91. Goto, M.; Shibahara, Y.; Baciu, C.; Allison, F.; Yeung, J.C.; Darling, G.E.; Liu, M. Prognostic Impact of CXCR7 and CXCL12 Expression in Patients with Esophageal Adenocarcinoma. Ann. Surg. Oncol. 2021, 28, 4943–4951. [Google Scholar] [CrossRef] [PubMed]
  92. Lagisetty, K.H.; McEwen, D.P.; Nancarrow, D.J.; Schiebel, J.G.; Ferrer-Torres, D.; Ray, D.; Frankel, T.L.; Lin, J.; Chang, A.C.; Kresty, L.A.; et al. Immune determinants of Barrett’s progression to esophageal adenocarcinoma. JCI Insight 2021, 6, e143888. [Google Scholar] [CrossRef]
  93. Lind, A.; Siersema, P.D.; Kusters, J.G.; Konijn, T.; Mebius, R.E.; Koenderman, L. The Microenvironment in Barrett’s Esophagus Tissue Is Characterized by High FOXP3 and RALDH2 Levels. Front. Immunol. 2018, 9, 1375. [Google Scholar] [CrossRef] [PubMed]
  94. Rubinkiewicz, M.; Migaczewski, M.; Hankus, J.; Major, P.; Pedziwiatr, M.; Budzynski, P.; Okon, K.; Budzynski, A. Foxp3+ lymphocyte count in Barrett’s esophagus tissue is higher than in in amed esophageal tissue. Folia. Med. Cracov. 2016, 56, 51–59. [Google Scholar] [PubMed]
  95. Gokon, Y.; Fujishima, F.; Taniyama, Y.; Ishida, H.; Yamagata, T.; Sawai, T.; Uzuki, M.; Ichikawa, H.; Itakura, Y.; Takahashi, K.; et al. Immune microenvironment in Barrett’s esophagus adjacent to esophageal adenocarcinoma: Possible influence of adjacent mucosa on cancer development and progression. Virchows Arch. 2020, 477, 825–834. [Google Scholar] [CrossRef] [PubMed]
  96. Volkweis, B.S.; Gurski, R.R.; Meurer, L.; Pretto, G.G.; Mazzini Gda, S.; Edelweiss, M.I. Ki-67 Antigen Overexpression Is Associated with the Metaplasia-Adenocarcinoma Sequence in Barrett’s Esophagus. Gastroenterol. Res. Pract. 2012, 2012, 639748. [Google Scholar] [CrossRef] [PubMed]
  97. Derks, S.; Nason, K.S.; Liao, X.; Stachler, M.D.; Liu, K.X.; Liu, J.B.; Sicinska, E.; Goldberg, M.S.; Freeman, G.J.; Rodig, S.J.; et al. Epithelial PD-L2 Expression Marks Barrett’s Esophagus and Esophageal Adenocarcinoma. Cancer Immunol. Res. 2015, 3, 1123–1129. [Google Scholar] [CrossRef]
  98. Moons, L.M.G.; Kusters, J.G.; Bultman, E.; Kuipers, E.J.; van Dekken, H.; Tra, W.M.W.; Kleinjan, A.; Kwekkeboom, J.; van Vliet, A.H.M.; Siersema, P.D. Barrett’s oesophagus is characterized by a predominantly humoral inflammatory response. J. Pathol. 2005, 207, 269–276. [Google Scholar] [CrossRef]
  99. Fitzgerald, R.C.; Onwuegbusi, B.A.; Bajaj-Elliott, M.; Saeed, I.T.; Burnham, W.R.; Farthing, M.J. Diversity in the oesophageal phenotypic response to gastro-oesophageal reflux: Immunological determinants. Gut 2002, 50, 451–459. [Google Scholar] [CrossRef]
  100. Liu, J.Y.; Geng, X.F.; Hou, J.X.; Wu, G.S. New insights into M1/M2 macrophages: Key modulators in cancer progression. Cancer Cell Int. 2021, 21, 389. [Google Scholar] [CrossRef]
  101. Kotsafti, A.; Fassan, M.; Cavallin, F.; Angerilli, V.; Saadeh, L.; Cagol, M.; Alfieri, R.; Pilati, P.; Castoro, C.; Castagliuolo, I.; et al. Tumor immune microenvironment in therapy-naive esophageal adenocarcinoma could predict the nodal status. Cancer Med. 2023, 12, 5526–5535. [Google Scholar] [CrossRef] [PubMed]
  102. Davis, B.P.; Rothenberg, M.E. Eosinophils and cancer. Cancer Immunol. Res. 2014, 2, 1–8. [Google Scholar] [CrossRef] [PubMed]
  103. Croft, W.; Evans, R.P.T.; Pearce, H.; Elshafie, M.; Griffiths, E.A.; Moss, P. The single cell transcriptional landscape of esophageal adenocarcinoma and its modulation by neoadjuvant chemotherapy. Mol. Cancer 2022, 21, 200. [Google Scholar] [CrossRef] [PubMed]
  104. Caspa Gokulan, R.; Garcia-Buitrago, M.T.; Zaika, A.I. From genetics to signaling pathways: Molecular pathogenesis of esophageal adenocarcinoma. Biochim. Biophys. Acta. Rev. Cancer 2019, 1872, 37–48. [Google Scholar] [CrossRef] [PubMed]
  105. Dulak, A.M.; Stojanov, P.; Peng, S.; Lawrence, M.S.; Fox, C.; Stewart, C.; Bandla, S.; Imamura, Y.; Schumacher, S.E.; Shefler, E.; et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat. Genet. 2013, 45, 478–486. [Google Scholar] [CrossRef] [PubMed]
  106. Weaver, J.M.J.; Ross-Innes, C.S.; Shannon, N.; Lynch, A.G.; Forshew, T.; Barbera, M.; Murtaza, M.; Ong, C.J.; Lao-Sirieix, P.; Dunning, M.J.; et al. Ordering of mutations in preinvasive disease stages of esophageal carcinogenesis. Nat. Genet. 2014, 46, 837–843. [Google Scholar] [CrossRef] [PubMed]
  107. Ross-Innes, C.S.; Becq, J.; Warren, A.; Cheetham, R.K.; Northen, H.; O’Donovan, M.; Malhotra, S.; di Pietro, M.; Ivakhno, S.; He, M.; et al. Whole-genome sequencing provides new insights into the clonal architecture of Barrett’s esophagus and esophageal adenocarcinoma. Nat. Genet. 2015, 47, 1038–1046. [Google Scholar] [CrossRef]
  108. Nones, K.; Waddell, N.; Wayte, N.; Patch, A.M.; Bailey, P.; Newell, F.; Holmes, O.; Fink, J.L.; Quinn, M.C.J.; Tang, Y.H.; et al. Genomic catastrophes frequently arise in esophageal adenocarcinoma and drive tumorigenesis. Nat. Commun. 2014, 5, 5224. [Google Scholar] [CrossRef]
  109. Gerlinger, M.; Rowan, A.J.; Horswell, S.; Math, M.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012, 366, 883–892. [Google Scholar] [CrossRef]
  110. The ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium. Pan-cancer analysis of whole genomes. Nature 2020, 578, 82–93. [Google Scholar] [CrossRef]
  111. Cai, H.; Jing, C.; Chang, X.; Ding, D.; Han, T.; Yang, J.; Lu, Z.; Hu, X.; Liu, Z.; Wang, J.; et al. Mutational landscape of gastric cancer and clinical application of genomic profiling based on target next-generation sequencing. J. Transl. Med. 2019, 17, 189. [Google Scholar] [CrossRef] [PubMed]
  112. Waddell, N.; Pajic, M.; Patch, A.M.; Chang, D.K.; Kassahn, K.S.; Bailey, P.; Johns, A.L.; Miller, D.; Nones, K.; Quek, K.; et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015, 518, 495–501. [Google Scholar] [CrossRef] [PubMed]
  113. Stachler, M.D.; Taylor-Weiner, A.; Peng, S.; McKenna, A.; Agoston, A.T.; Odze, R.D.; Davison, J.M.; Nason, K.S.; Loda, M.; Leshchiner, I.; et al. Paired exome analysis of Barrett’s esophagus and adenocarcinoma. Nat. Genet. 2015, 47, 1047–1055. [Google Scholar] [CrossRef]
  114. Maley, C.C.; Galipeau, P.C.; Finley, J.C.; Wongsurawat, V.J.; Li, X.; Sanchez, C.A.; Paulson, T.G.; Blount, P.L.; Risques, R.A.; Rabinovitch, P.S.; et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat. Genet. 2006, 38, 468–473. [Google Scholar] [CrossRef] [PubMed]
  115. Reid, B.J.; Barrett, M.T.; Galipeau, P.C.; Sanchez, C.A.; Neshat, K.; Cowan, D.S.; Levine, D.S. Barrett’s esophagus: Ordering the events that lead to cancer. Eur. J. Cancer Prev. 1996, 5, 57–65. [Google Scholar] [CrossRef] [PubMed]
  116. Martincorena, I.; Fowler, J.C.; Wabik, A.; Lawson, A.R.J.; Abascal, F.; Hall, M.W.J.; Cagan, A.; Murai, K.; Mahbubani, K.; Stratton, M.R.; et al. Somatic mutant clones colonize the human esophagus with age. Science 2018, 362, 911–917. [Google Scholar] [CrossRef] [PubMed]
  117. Martinez, P.; Timmer, M.R.; Lau, C.T.; Calpe, S.; Sancho-Serra Mdel, C.; Straub, D.; Baker, A.M.; Meijer, S.L.; Kate, F.J.; Mallant-Hent, R.C.; et al. Dynamic clonal equilibrium and predetermined cancer risk in Barrett’s oesophagus. Nat. Commun. 2016, 7, 12158. [Google Scholar] [CrossRef]
  118. Gharahkhani, P.; Fitzgerald, R.C.; Vaughan, T.L.; Palles, C.; Gockel, I.; Tomlinson, I.; Buas, M.F.; May, A.; Gerges, C.; Anders, M.; et al. Genome-wide association studies in oesophageal adenocarcinoma and Barrett’s oesophagus: A large-scale meta-analysis. Lancet Oncol. 2016, 17, 1363–1373. [Google Scholar] [CrossRef]
  119. Su, Z.; Gay, L.J.; Strange, A.; Palles, C.; Band, G.; Whiteman, D.C.; Lescai, F.; Langford, C.; Nanji, M.; Edkins, S.; et al. Common variants at the MHC locus and at chromosome 16q24.1 predispose to Barrett’s esophagus. Nat. Genet. 2012, 44, 1131–1136. [Google Scholar] [CrossRef]
  120. Frankell, A.M.; Jammula, S.; Li, X.D.; Contino, G.; Killcoyne, S.; Abbas, S.; Perner, J.; Bower, L.; Devonshire, G.; Cocks, E.; et al. The landscape of selection in 551 esophageal adenocarcinomas defines genomic biomarkers for the clinic. Nat. Genet. 2019, 51, 506–516. [Google Scholar] [CrossRef]
  121. Lin, D.C.; Dinh, H.Q.; Xie, J.J.; Mayakonda, A.; Silva, T.C.; Jiang, Y.Y.; Ding, L.W.; He, J.Z.; Xu, X.E.; Hao, J.J.; et al. Identification of distinct mutational patterns and new driver genes in oesophageal squamous cell carcinomas and adenocarcinomas. Gut 2018, 67, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
  122. Orsini, A.; Mastracci, L.; Bozzarelli, I.; Ferrari, A.; Isidori, F.; Fiocca, R.; Lugaresi, M.; D’Errico, A.; Malvi, D.; Cataldi-Stagetti, E.; et al. Correlations between Molecular Alterations, Histopathological Characteristics, and Poor Prognosis in Esophageal Adenocarcinoma. Cancers 2023, 15, 1408. [Google Scholar] [CrossRef] [PubMed]
  123. Secrier, M.; Li, X.; de Silva, N.; Eldridge, M.D.; Contino, G.; Bornschein, J.; MacRae, S.; Grehan, N.; O’Donovan, M.; Miremadi, A.; et al. Mutational signatures in esophageal adenocarcinoma define etiologically distinct subgroups with therapeutic relevance. Nat. Genet. 2016, 48, 1131–1141. [Google Scholar] [CrossRef] [PubMed]
  124. Sherr, C.J. Principles of tumor suppression. Cell 2004, 116, 235–246. [Google Scholar] [CrossRef] [PubMed]
  125. Madani, K.; Zhao, R.; Lim, H.J.; Casson, A.G. Prognostic value of p53 mutations in oesophageal adenocarcinoma: Final results of a 15-year prospective study. Eur. J. Cardiothorac. Surg. 2010, 37, 1427–1432. [Google Scholar] [CrossRef] [PubMed]
  126. Singhi, A.D.; Foxwell, T.J.; Nason, K.; Cressman, K.L.; McGrath, K.M.; Sun, W.J.; Bahary, N.; Zeh, H.J.; Levy, R.M.; Luketich, J.D.; et al. Smad4 Loss in Esophageal Adenocarcinoma Is Associated With an Increased Propensity for Disease Recurrence and Poor Survival. Am. J. Surg. Pathol. 2015, 39, 487–495. [Google Scholar] [CrossRef] [PubMed]
  127. Zack, T.I.; Schumacher, S.E.; Carter, S.L.; Cherniack, A.D.; Saksena, G.; Tabak, B.; Lawrence, M.S.; Zhsng, C.Z.; Wala, J.; Mermel, C.H.; et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 2013, 45, 1134–1140. [Google Scholar] [CrossRef] [PubMed]
  128. Pasello, G.; Agata, S.; Bonaldi, L.; Corradin, A.; Montagna, M.; Zamarchi, R.; Parenti, A.; Cagol, M.; Zaninotto, G.; Ruol, A.; et al. DNA copy number alterations correlate with survival of esophageal adenocarcinoma patients. Mod. Pathol. 2009, 22, 58–65. [Google Scholar] [CrossRef]
  129. Sihag, S.; Nussenzweig, S.C.; Walch, H.S.; Hsu, M.; Tan, K.S.; Sanchez-Vega, F.; Chatila, W.K.; De La Torre, S.A.; Patel, A.; Janjigian, Y.Y.; et al. Next-Generation Sequencing of 487 Esophageal Adenocarcinomas Reveals Independently Prognostic Genomic Driver Alterations and Pathways. Clin. Cancer Res. 2021, 27, 3491–3498. [Google Scholar] [CrossRef]
  130. Paulson, T.G.; Maley, C.C.; Li, X.; Li, H.; Sanchez, C.A.; Chao, D.L.; Odze, R.D.; Vaughan, T.L.; Blount, P.L.; Reid, B.J. Chromosomal instability and copy number alterations in Barrett’s esophagus and esophageal adenocarcinoma. Clin. Cancer Res. 2009, 15, 3305–3314. [Google Scholar] [CrossRef]
  131. Davis, A.; Gao, R.; Navin, N. Tumor evolution: Linear, branching, neutral or punctuated? Biochim. Biophys. Acta. Rev. Cancer 2017, 1867, 151–161. [Google Scholar] [CrossRef] [PubMed]
  132. Navin, N.; Kendall, J.; Troge, J.; Andrews, P.; Rodgers, L.; McIndoo, J.; Cook, K.; Stepansky, A.; Levy, D.; Esposito, D.; et al. Tumour evolution inferred by single-cell sequencing. Nature 2011, 472, 90–94. [Google Scholar] [CrossRef] [PubMed]
  133. Reid, B.J.; Prevo, L.J.; Galipeau, P.C.; Sanchez, C.A.; Longton, G.; Levine, D.S.; Blount, P.L.; Rabinovitch, P.S. Predictors of progression in Barrett’s esophagus II: Baseline 17p (p53) loss of heterozygosity identifies a patient subset at increased risk for neoplastic progression. Am. J. Gastroenterol. 2001, 96, 2839–2848. [Google Scholar] [CrossRef] [PubMed]
  134. Timmer, M.R.; Martinez, P.; Lau, C.T.; Westra, W.M.; Calpe, S.; Rygiel, A.M.; Rosmolen, W.D.; Meijer, S.L.; Ten Kate, F.J.; Dijkgraaf, M.G.; et al. Derivation of genetic biomarkers for cancer risk stratification in Barrett’s oesophagus: A prospective cohort study. Gut 2016, 65, 1602–1610. [Google Scholar] [CrossRef] [PubMed]
  135. Galipeau, P.C.; Li, X.; Blount, P.L.; Maley, C.C.; Sanchez, C.A.; Odze, R.D.; Ayub, K.; Rabinovitch, P.S.; Vaughan, T.L.; Reid, B.J. NSAIDs modulate CDKN2A, TP53, and DNA content risk for progression to esophageal adenocarcinoma. PLoS Med. 2007, 4, e67. [Google Scholar] [CrossRef] [PubMed]
  136. Dulak, A.M.; Schumacher, S.E.; van Lieshout, J.; Imamura, Y.; Fox, C.; Shim, B.; Ramos, A.H.; Saksena, G.; Baca, S.C.; Baselga, J.; et al. Gastrointestinal adenocarcinomas of the esophagus, stomach, and colon exhibit distinct patterns of genome instability and oncogenesis. Cancer Res. 2012, 72, 4383–4393. [Google Scholar] [CrossRef] [PubMed]
  137. Prins, M.J.; Ruurda, J.P.; van Diest, P.J.; van Hillegersberg, R.; Ten Kate, F.J. The significance of the HER-2 status in esophageal adenocarcinoma for survival: An immunohistochemical and an in situ hybridization study. Ann. Oncol. 2013, 24, 1290–1297. [Google Scholar] [CrossRef]
  138. Finley, J.C.; Reid, B.J.; Odze, R.D.; Sanchez, C.A.; Galipeau, P.; Li, X.H.; Self, S.G.; Gollahon, K.A.; Blount, P.L.; Rabinovitch, P.S. Chromosomal instability in Barrett’s esophagus is related to telomere shortening. Cancer Epidem. Biomar. 2006, 15, 1451–1457. [Google Scholar] [CrossRef]
  139. Risques, R.A.; Vaughan, T.L.; Li, X.; Odze, R.D.; Blount, P.L.; Ayub, K.; Gallaher, J.L.; Reid, B.J.; Rabinovitch, P.S. Leukocyte telomere length predicts cancer risk in Barrett’s esophagus. Cancer Epidemiol. Biomark. Prev. 2007, 16, 2649–2655. [Google Scholar] [CrossRef]
  140. Xing, J.; Ajani, J.A.; Chen, M.; Izzo, J.; Lin, J.; Chen, Z.; Gu, J.; Wu, X. Constitutive short telomere length of chromosome 17p and 12q but not 11q and 2p is associated with an increased risk for esophageal cancer. Cancer Prev. Res. 2009, 2, 459–465. [Google Scholar] [CrossRef]
  141. Demissie, S.; Levy, D.; Benjamin, E.J.; Cupples, L.A.; Gardner, J.P.; Herbert, A.; Kimura, M.; Larson, M.G.; Meigs, J.B.; Keaney, J.F.; et al. Insulin resistance, oxidative stress, hypertension, and leukocyte telomere length in men from the Framingham Heart Study. Aging Cell 2006, 5, 325–330. [Google Scholar] [CrossRef] [PubMed]
  142. Valdes, A.M.; Andrew, T.; Gardner, J.P.; Kimura, M.; Oelsner, E.; Cherkas, L.F.; Aviv, A.; Spector, T.D. Obesity, cigarette smoking, and telomere length in women. Lancet 2005, 366, 662–664. [Google Scholar] [CrossRef] [PubMed]
  143. Hoefnagel, S.J.M.; Koemans, W.J.; Khan, H.N.; Koster, J.; Meijer, S.L.; van Dieren, J.M.; Kodach, L.L.; van Sandick, J.W.; Calpe, S.; Del Sancho-Serra, C.M.; et al. Identification of Novel Molecular Subgroups in Esophageal Adenocarcinoma to Predict Response to Neo-Adjuvant Therapies. Cancers 2022, 14, 4498. [Google Scholar] [CrossRef] [PubMed]
  144. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef] [PubMed]
  145. Xu, E.P. Genome-wide methylation analysis shows similar patterns in Barrett’s esophagus and esophageal adenocarcinoma (vol 34, pg 2750, 2013). Carcinogenesis 2014, 35, 738. [Google Scholar]
  146. Smith, E.; De Young, N.J.; Pavey, S.J.; Hayward, N.K.; Nancarrow, D.J.; Whiteman, D.C.; Smithers, B.M.; Ruszkiewicz, A.R.; Clouston, A.D.; Gotley, D.C.; et al. Similarity of aberrant DNA methylation in Barrett’s esophagus and esophageal adenocarcinoma. Mol. Cancer 2008, 7, 75. [Google Scholar] [CrossRef] [PubMed]
  147. Klump, B.; Hsieh, C.J.; Holzmann, K.; Gregor, M.; Porschen, R. Hypermethylation of the CDKN2/p16 promoter during neoplastic progression in Barrett’s esophagus. Gastroenterology 1998, 115, 1381–1386. [Google Scholar] [CrossRef]
  148. Wong, D.J.; Barrett, M.T.; Stoger, R.; Emond, M.J.; Reid, B.J. p16INK4a promoter is hypermethylated at a high frequency in esophageal adenocarcinomas. Cancer Res. 1997, 57, 2619–2622. [Google Scholar]
  149. Peng, D.; Hu, T.L.; Jiang, A.; Washington, M.K.; Moskaluk, C.A.; Schneider-Stock, R.; El-Rifai, W. Location-specific epigenetic regulation of the metallothionein 3 gene in esophageal adenocarcinomas. PLoS ONE 2011, 6, e22009. [Google Scholar] [CrossRef]
  150. Alvi, M.A.; Liu, X.X.; O’Donovan, M.; Newton, R.; Wernisch, L.; Shannon, N.B.; Shariff, K.; di Pietro, M.; Bergman, J.J.G.H.M.; Ragunath, K.; et al. DNA Methylation as an Adjunct to Histopathology to Detect Prevalent, Inconspicuous Dysplasia and Early-Stage Neoplasia in Barrett’s Esophagus. Clin. Cancer Res. 2013, 19, 878–888. [Google Scholar] [CrossRef]
  151. Fassan, M.; Volinia, S.; Palatini, J.; Pizzi, M.; Baffa, R.; De Bernard, M.; Battaglia, G.; Parente, P.; Croce, C.M.; Zaninotto, G.; et al. MicroRNA expression profiling in human Barrett’s carcinogenesis. Int. J. Cancer 2011, 129, 1661–1670. [Google Scholar] [CrossRef] [PubMed]
  152. Huang, J.; Zhang, S.Y.; Gao, Y.M.; Liu, Y.F.; Liu, Y.B.; Zhao, Z.G.; Yang, K. MicroRNAs as oncogenes or tumour suppressors in oesophageal cancer: Potential biomarkers and therapeutic targets. Cell Proliferat. 2014, 47, 277–286. [Google Scholar] [CrossRef] [PubMed]
  153. Kailasam, A.; Mittal, S.K.; Agrawal, D.K. Epigenetics in the Pathogenesis of Esophageal Adenocarcinoma. Clin. Transl. Sci. 2015, 8, 394–402. [Google Scholar] [CrossRef] [PubMed]
  154. Saad, R.; Chen, Z.; Zhu, S.; Jia, P.; Zhao, Z.; Washington, M.K.; Belkhiri, A.; El-Rifai, W. Deciphering the unique microRNA signature in human esophageal adenocarcinoma. PLoS ONE 2013, 8, e64463. [Google Scholar] [CrossRef] [PubMed]
  155. Chen, Z.; Saad, R.; Jia, P.; Peng, D.; Zhu, S.; Washington, M.K.; Zhao, Z.; Xu, Z.; El-Rifai, W. Gastric adenocarcinoma has a unique microRNA signature not present in esophageal adenocarcinoma. Cancer 2013, 119, 1985–1993. [Google Scholar] [CrossRef] [PubMed]
  156. Hu, G.; Chen, D.; Li, X.; Yang, K.; Wang, H.; Wu, W. miR-133b regulates the MET proto-oncogene and inhibits the growth of colorectal cancer cells in vitro and in vivo. Cancer Biol. Ther. 2010, 10, 190–197. [Google Scholar] [CrossRef] [PubMed]
  157. Wu, Y.; Crawford, M.; Yu, B.; Mao, Y.; Nana-Sinkam, S.P.; Lee, L.J. MicroRNA delivery by cationic lipoplexes for lung cancer therapy. Mol. Pharm. 2011, 8, 1381–1389. [Google Scholar] [CrossRef] [PubMed]
  158. Hatley, M.E.; Patrick, D.M.; Garcia, M.R.; Richardson, J.A.; Bassel-Duby, R.; van Rooij, E.; Olson, E.N. Modulation of K-Ras-dependent lung tumorigenesis by MicroRNA-21. Cancer Cell 2010, 18, 282–293. [Google Scholar] [CrossRef]
  159. Iliopoulos, D.; Jaeger, S.A.; Hirsch, H.A.; Bulyk, M.L.; Struhl, K. STAT3 activation of miR-21 and miR-181b-1 via PTEN and CYLD are part of the epigenetic switch linking inflammation to cancer. Mol. Cell 2010, 39, 493–506. [Google Scholar] [CrossRef]
  160. Meng, F.; Henson, R.; Wehbe-Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef]
  161. Mari, L.; Hoefnagel, S.J.M.; Zito, D.; van de Meent, M.; van Endert, P.; Calpe, S.; Sancho Serra, M.D.C.; Heemskerk, M.H.M.; van Laarhoven, H.W.M.; Hulshof, M.; et al. microRNA 125a Regulates MHC-I Expression on Esophageal Adenocarcinoma Cells, Associated With Suppression of Antitumor Immune Response and Poor Outcomes of Patients. Gastroenterology 2018, 155, 784–798. [Google Scholar] [CrossRef]
  162. Arnold, M.; Abnet, C.C.; Neale, R.E.; Vignat, J.; Giovannucci, E.L.; McGlynn, K.A.; Bray, F. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology 2020, 159, 335–349.e315. [Google Scholar] [CrossRef] [PubMed]
  163. Fitzgerald, R. Screening for Barrett Esophagus With a Nonendoscopic Sponge Capsule. Gastroenterol. Hepatol. 2015, 11, 623–625. [Google Scholar]
  164. Peters, Y.; Schrauwen, R.W.M.; Tan, A.C.; Bogers, S.K.; de Jong, B.; Siersema, P.D. Detection of Barrett’s oesophagus through exhaled breath using an electronic nose device. Gut 2020, 69, 1169–1172. [Google Scholar] [CrossRef] [PubMed]
  165. Codipilly, D.C.; Chandar, A.K.; Singh, S.; Wani, S.; Shaheen, N.J.; Inadomi, J.M.; Chak, A.; Iyer, P.G. The Effect of Endoscopic Surveillance in Patients With Barrett’s Esophagus: A Systematic Review and Meta-analysis. Gastroenterology 2018, 154, 2068–2086.e2065. [Google Scholar] [CrossRef] [PubMed]
  166. Cooper, G.S.; Kou, T.D.; Chak, A. Receipt of previous diagnoses and endoscopy and outcome from esophageal adenocarcinoma: A population-based study with temporal trends. Am. J. Gastroenterol. 2009, 104, 1356–1362. [Google Scholar] [CrossRef] [PubMed]
  167. Fountoulakis, A.; Zafirellis, K.D.; Dolan, K.; Dexter, S.P.; Martin, I.G.; Sue-Ling, H.M. Effect of surveillance of Barrett’s oesophagus on the clinical outcome of oesophageal cancer. Br. J. Surg. 2004, 91, 997–1003. [Google Scholar] [CrossRef] [PubMed]
  168. Rubenstein, J.H.; Sonnenberg, A.; Davis, J.; McMahon, L.; Inadomi, J.M. Effect of a prior endoscopy on outcomes of esophageal adenocarcinoma among United States veterans. Gastrointest. Endosc. 2008, 68, 849–855. [Google Scholar] [CrossRef] [PubMed]
  169. Hillman, L.C.; Chiragakis, L.; Shadbolt, B.; Kaye, G.L.; Clarke, A.C. Proton-pump inhibitor therapy and the development of dysplasia in patients with Barrett’s oesophagus. Med. J. Aust. 2004, 180, 387–391. [Google Scholar] [CrossRef]
  170. Kastelein, F.; Spaander, M.C.; Steyerberg, E.W.; Biermann, K.; Valkhoff, V.E.; Kuipers, E.J.; Bruno, M.J.; ProBar Study, G. Proton pump inhibitors reduce the risk of neoplastic progression in patients with Barrett’s esophagus. Clin. Gastroenterol. Hepatol. 2013, 11, 382–388. [Google Scholar] [CrossRef]
  171. Nguyen, D.M.; El-Serag, H.B.; Henderson, L.; Stein, D.; Bhattacharyya, A.; Sampliner, R.E. Medication usage and the risk of neoplasia in patients with Barrett’s esophagus. Clin. Gastroenterol. Hepatol. 2009, 7, 1299–1304. [Google Scholar] [CrossRef] [PubMed]
  172. Corley, D.A.; Kerlikowske, K.; Verma, R.; Buffler, P. Protective association of aspirin/NSAIDs and esophageal cancer: A systematic review and meta-analysis. Gastroenterology 2003, 124, 47–56. [Google Scholar] [CrossRef] [PubMed]
  173. Liao, L.M.; Vaughan, T.L.; Corley, D.A.; Cook, M.B.; Casson, A.G.; Kamangar, F.; Abnet, C.C.; Risch, H.A.; Giffen, C.; Freedman, N.D.; et al. Nonsteroidal anti-inflammatory drug use reduces risk of adenocarcinomas of the esophagus and esophagogastric junction in a pooled analysis. Gastroenterology 2012, 142, 442–452. [Google Scholar] [CrossRef] [PubMed]
  174. Nguyen, D.M.; Richardson, P.; El-Serag, H.B. Medications (NSAIDs, Statins, Proton Pump Inhibitors) and the Risk of Esophageal Adenocarcinoma in Patients With Barrett’s Esophagus. Gastroenterology 2010, 138, 2260–2266. [Google Scholar] [CrossRef] [PubMed]
  175. Zhang, S.; Zhang, X.Q.; Ding, X.W.; Yang, R.K.; Huang, S.L.; Kastelein, F.; Bruno, M.; Yu, X.J.; Zhou, D.; Zou, X.P. Cyclooxygenase inhibitors use is associated with reduced risk of esophageal adenocarcinoma in patients with Barrett’s esophagus: A meta-analysis. Br. J. Cancer 2014, 110, 2378–2388. [Google Scholar] [CrossRef] [PubMed]
  176. Molina, M.A.; Sitja-Arnau, M.; Lemoine, M.G.; Frazier, M.L.; Sinicrope, F.A. Increased cyclooxygenase-2 expression in human pancreatic carcinomas and cell lines: Growth inhibition by nonsteroidal anti-inflammatory drugs. Cancer Res. 1999, 59, 4356–4362. [Google Scholar] [PubMed]
  177. Yin, M.J.; Yamamoto, Y.; Gaynor, R.B. The anti-inflammatory agents aspirin and salicylate inhibit the activity of I(kappa)B kinase-beta. Nature 1998, 396, 77–80. [Google Scholar] [CrossRef] [PubMed]
  178. Overholt, B.F.; Wang, K.K.; Burdick, J.S.; Lightdale, C.J.; Kimmey, M.; Nava, H.R.; Sivak, M.V., Jr.; Nishioka, N.; Barr, H.; Marcon, N.; et al. Five-year efficacy and safety of photodynamic therapy with Photofrin in Barrett’s high-grade dysplasia. Gastrointest. Endosc. 2007, 66, 460–468. [Google Scholar] [CrossRef]
  179. Genere, J.R.; Visrodia, K.; Zakko, L.; Hoefnagel, S.J.M.; Wang, K.K. Spray cryotherapy versus continued radiofrequency ablation in persistent Barrett’s esophagus. Dis. Esophagus 2022, 35, doab084. [Google Scholar] [CrossRef]
  180. Mansour, N.M.; Groth, S.S.; Anandasabapathy, S. Esophageal Adenocarcinoma: Screening, Surveillance, and Management. Annu. Rev. Med. 2017, 68, 213–227. [Google Scholar] [CrossRef]
  181. Shaheen, N.J.; Sharma, P.; Overholt, B.F.; Wolfsen, H.C.; Sampliner, R.E.; Wang, K.K.; Galanko, J.A.; Bronner, M.P.; Goldblum, J.R.; Bennett, A.E.; et al. Radiofrequency ablation in Barrett’s esophagus with dysplasia. N. Engl. J. Med. 2009, 360, 2277–2288. [Google Scholar] [CrossRef] [PubMed]
  182. Liu, K.; Aadam, A.A. T1b esophageal cancer: Is it time for endoscopic submucosal dissection to enter the stage? Gastrointest. Endosc. 2022, 96, 454–456. [Google Scholar] [CrossRef] [PubMed]
  183. Obermannova, R.; Alsina, M.; Cervantes, A.; Leong, T.; Lordick, F.; Nilsson, M.; van Grieken, N.C.T.; Vogel, A.; Smyth, E.C. Oesophageal cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2022, 33, 992–1004. [Google Scholar] [CrossRef] [PubMed]
  184. van Hagen, P.; Hulshof, M.C.; van Lanschot, J.J.; Steyerberg, E.W.; van Berge Henegouwen, M.I.; Wijnhoven, B.P.; Richel, D.J.; Nieuwenhuijzen, G.A.; Hospers, G.A.; Bonenkamp, J.J.; et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N. Engl. J. Med. 2012, 366, 2074–2084. [Google Scholar] [CrossRef] [PubMed]
  185. Al-Batran, S.E.; Homann, N.; Pauligk, C.; Goetze, T.O.; Meiler, J.; Kasper, S.; Kopp, H.G.; Mayer, F.; Haag, G.M.; Luley, K.; et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): A randomised, phase 2/3 trial. Lancet 2019, 393, 1948–1957. [Google Scholar] [PubMed]
  186. Moussa, O.; Bhogal, R.H.; Malietzis, G.; Fribbens, C.; Starling, N.; Gerlinger, M.; Watkins, D.; Chau, I.; Rao, S.; Cunningham, D.; et al. Effect of perioperative FLOT versus ECF/ECX on short-term outcomes after surgery for resectable oesophagogastric adenocarcinoma: Propensity score-matched study. BJS Open 2022, 6, zrac003. [Google Scholar] [CrossRef] [PubMed]
  187. Reynolds, J.V.; Preston, S.R.; O’Neill, B.; Lowery, M.A.; Baeksgaard, L.; Crosby, T.; Cunningham, M.; Cuffe, S.; Griffiths, G.O.; Roy, R.; et al. Neo-AEGIS (Neoadjuvant trial in Adenocarcinoma of the Esophagus and Esophago-Gastric Junction International Study): Preliminary results of phase III RCT of CROSS versus perioperative chemotherapy (Modified MAGIC or FLOT protocol). (NCT01726452). J. Clin. Oncol. 2021, 39, 4004. [Google Scholar] [CrossRef]
  188. Hoeppner, J.; Lordick, F.; Brunner, T.; Glatz, T.; Bronsert, P.; Rothling, N.; Schmoor, C.; Lorenz, D.; Ell, C.; Hopt, U.T.; et al. ESOPEC: Prospective randomized controlled multicenter phase III trial comparing perioperative chemotherapy (FLOT protocol) to neoadjuvant chemoradiation (CROSS protocol) in patients with adenocarcinoma of the esophagus (NCT02509286). BMC Cancer 2016, 16, 503. [Google Scholar] [CrossRef]
  189. Mariette, C.; Markar, S.R.; Dabakuyo-Yonli, T.S.; Meunier, B.; Pezet, D.; Collet, D.; D’Journo, X.B.; Brigand, C.; Perniceni, T.; Carrere, N.; et al. Hybrid Minimally Invasive Esophagectomy for Esophageal Cancer. N. Engl. J. Med. 2019, 380, 152–162. [Google Scholar] [CrossRef]
  190. van der Sluis, P.C.; van der Horst, S.; May, A.M.; Schippers, C.; Brosens, L.A.A.; Joore, H.C.A.; Kroese, C.C.; Haj Mohammad, N.; Mook, S.; Vleggaar, F.P.; et al. Robot-assisted Minimally Invasive Thoracolaparoscopic Esophagectomy Versus Open Transthoracic Esophagectomy for Resectable Esophageal Cancer: A Randomized Controlled Trial. Ann. Surg. 2019, 269, 621–630. [Google Scholar] [CrossRef]
  191. Crosby, T.; Hurt, C.N.; Falk, S.; Gollins, S.; Staffurth, J.; Ray, R.; Bridgewater, J.A.; Geh, J.I.; Cunningham, D.; Blazeby, J.; et al. Long-term results and recurrence patterns from SCOPE-1: A phase II/III randomised trial of definitive chemoradiotherapy +/− cetuximab in oesophageal cancer. Br. J. Cancer 2017, 116, 709–716. [Google Scholar] [CrossRef] [PubMed]
  192. Conroy, T.; Galais, M.P.; Raoul, J.L.; Bouche, O.; Gourgou-Bourgade, S.; Douillard, J.Y.; Etienne, P.L.; Boige, V.; Martel-Lafay, I.; Michel, P.; et al. Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): Final results of a randomised, phase 2/3 trial. Lancet Oncol. 2014, 15, 305–314. [Google Scholar] [CrossRef] [PubMed]
  193. Ajani, J.A.; D’Amico, T.A.; Bentrem, D.J.; Chao, J.; Corvera, C.; Das, P.; Denlinger, C.S.; Enzinger, P.C.; Fanta, P.; Farjah, F.; et al. Esophageal and Esophagogastric Junction Cancers, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2019, 17, 855–883. [Google Scholar] [CrossRef] [PubMed]
  194. Sun, J.M.; Shen, L.; Shah, M.A.; Enzinger, P.; Adenis, A.; Doi, T.; Kojima, T.; Metges, J.P.; Li, Z.; Kim, S.B.; et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): A randomised, placebo-controlled, phase 3 study. Lancet 2021, 398, 759–771. [Google Scholar] [CrossRef] [PubMed]
  195. Shitara, K.; Doi, T.; Dvorkin, M.; Mansoor, W.; Arkenau, H.T.; Prokharau, A.; Alsina, M.; Ghidini, M.; Faustino, C.; Gorbunova, V.; et al. Trifluridine/tipiracil versus placebo in patients with heavily pretreated metastatic gastric cancer (TAGS): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2018, 19, 1437–1448. [Google Scholar] [CrossRef] [PubMed]
  196. Al-Batran, S.E.; Hartmann, J.T.; Probst, S.; Schmalenberg, H.; Hollerbach, S.; Hofheinz, R.; Rethwisch, V.; Seipelt, G.; Homann, N.; Wilhelm, G.; et al. Phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or cisplatin: A study of the Arbeitsgemeinschaft Internistische Onkologie. J. Clin. Oncol. 2008, 26, 1435–1442. [Google Scholar] [CrossRef] [PubMed]
  197. Yamaguchi, K.; Bang, Y.J.; Iwasa, S.; Sugimoto, N.; Ryu, M.H.; Sakai, D.; Chung, H.C.C.; Kawakami, H.; Yabusaki, H.; Lee, J.; et al. Trastuzumab deruxtecan (T-DXd; DS-8201) in patients with HER2-positive advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma: Final overall survival (OS) results from a randomized, multicenter, open-label, phase 2 study (DESTINY-Gastric01). J. Clin. Oncol. 2021, 40, 4048. [Google Scholar] [CrossRef]
  198. Fuchs, C.S.; Tomasek, J.; Yong, C.J.; Dumitru, F.; Passalacqua, R.; Goswami, C.; Safran, H.; Dos Santos, L.V.; Aprile, G.; Ferry, D.R.; et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): An international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2014, 383, 31–39. [Google Scholar] [CrossRef]
  199. Wilke, H.; Muro, K.; Van Cutsem, E.; Oh, S.C.; Bodoky, G.; Shimada, Y.; Hironaka, S.; Sugimoto, N.; Lipatov, O.; Kim, T.Y.; et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): A double-blind, randomised phase 3 trial. Lancet Oncol. 2014, 15, 1224–1235. [Google Scholar] [CrossRef]
  200. Kelly, R.J.; Ajani, J.A.; Kuzdzal, J.; Zander, T.; Van Cutsem, E.; Piessen, G.; Mendez, G.; Feliciano, J.; Motoyama, S.; Lièvre, A.; et al. Adjuvant Nivolumab in Resected Esophageal or Gastroesophageal Junction Cancer. N. Engl. J. Med. 2021, 384, 1191–1203. [Google Scholar] [CrossRef]
  201. Janjigian, Y.Y.; Shitara, K.; Moehler, M.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Campos Bragagnoli, A.; et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): A randomised, open-label, phase 3 trial. Lancet 2021, 398, 27–40. [Google Scholar] [CrossRef] [PubMed]
  202. Janjigian, Y.Y.; Kawazoe, A.; Yanez, P.E.; Luo, S.X.; Lonardi, S.; Kolesnik, O.; Barajas, O.; Bai, Y.X.; Shen, L.; Tang, Y.; et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2+metastatic gastric or gastroesophageal junction (G/GEJ) cancer: Initial findings of the global phase 3 KEYNOTE-811 study. J. Clin. Oncol. 2021, 39, iv25. [Google Scholar] [CrossRef]
  203. Li, S.; Hoefnagel, S.J.M.; Krishnadath, K.K. Single domain Camelid antibody fragments for molecular imaging and therapy of cancer. Front. Oncol. 2023, 13, 1257175. [Google Scholar] [CrossRef] [PubMed]
  204. Hazelton, W.D.; Curtius, K.; Inadomi, J.M.; Vaughan, T.L.; Meza, R.; Rubenstein, J.H.; Hur, C.; Luebeck, E.G. The Role of Gastroesophageal Reflux and Other Factors during Progression to Esophageal Adenocarcinoma. Cancer Epidemiol. Biomark. Prev. 2015, 24, 1012–1023. [Google Scholar] [CrossRef] [PubMed]
  205. Ansari, A.; Miyashita, T.; Lay, F.; Ahmed, A.K.; Matsangos, A.E.; Born, L.; Ng, C.; Cohen, R.M.; Stricker-Krongrad, A.H.; Salimian, K.; et al. Targeting the hedgehog pathway in esophageal adenocarcinoma (EAC) using Itraconazole. J. Clin. Oncol. 2018, 36, e13552. [Google Scholar] [CrossRef]
  206. Vaninetti, N.; Williams, L.; Geldenhuys, L.; Porter, G.A.; Guernsey, D.L.; Casson, A.G. Regulation of CDX2 expression in esophageal adenocarcinoma. Mol. Carcinog. 2009, 48, 965–974. [Google Scholar] [CrossRef] [PubMed]
  207. Revenko, A.; Carnevalli, L.S.; Sinclair, C.; Johnson, B.; Peter, A.; Taylor, M.; Hettrick, L.; Chapman, M.; Klein, S.; Solanki, A.; et al. Direct targeting of FOXP3 in Tregs with AZD8701, a novel antisense oligonucleotide to relieve immunosuppression in cancer. J. Immunother. Cancer 2022, 10, e003892. [Google Scholar] [CrossRef]
  208. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef]
  209. Mantovani, A.; Allavena, P.; Marchesi, F.; Garlanda, C. Macrophages as tools and targets in cancer therapy. Nat. Rev. Drug Discov. 2022, 21, 799–820. [Google Scholar] [CrossRef]
  210. Geboes, K.; Hoorens, A. The cell of origin for Barrett’s esophagus. Science 2021, 373, 737–738. [Google Scholar] [CrossRef]
  211. Correia, A.C.P.; Straub, D.; Calpe, S.; Krishnadath, K.K. Novel In Vivo Mouse Cryoablation Model to Explore Unique Therapeutic Approaches for Premalignant Columnar Lesions. Methods Protoc. 2021, 4, 6. [Google Scholar] [CrossRef]
  212. Sihag, S.; Nussenzweig, S.C.; Walch, H.S.; Hsu, M.; Tan, K.S.; De La Torre, S.; Janjigian, Y.Y.; Maron, S.B.; Ku, G.Y.; Tang, L.H.; et al. The Role of the TP53 Pathway in Predicting Response to Neoadjuvant Therapy in Esophageal Adenocarcinoma. Clin. Cancer Res. 2022, 28, 2669–2678. [Google Scholar] [CrossRef]
Figure 1. Schematic overview of important molecular pathways driving metaplasia in esophageal squamous epithelium as a result of GERD.
Figure 1. Schematic overview of important molecular pathways driving metaplasia in esophageal squamous epithelium as a result of GERD.
Cancers 15 05410 g001
Table 1. The existent theories about the origin of Barrett’s esophagus as suggested by original research.
Table 1. The existent theories about the origin of Barrett’s esophagus as suggested by original research.
ReferenceAuthorsYearOriginDrivers
[43]K. Nowicki-osuch et al.2021Gastric cardiac-MYC- and HNF4A-driven transcriptional programs
[44]D. Straub et al.2019Non-squamous cells residing in submucosal gland ductsGlycine-conjugated bile acids
[45]C. Minacapelli et al.2018Normal esophageal squamous epithelial cellsAcid and bile
[46]M. Jiang2017p63+KRT5+KRT7+ basal cells in the upper gastrointestinal tractEctopic expression of CDX2
[40]M. Quante2012Gastric cardia stem cellsBile acids and/or nitrosamines
[47]X. Wang et al.2011Embryonic cells at the squamocolumnar junctionCompetitive interactions between cell lineages
[48]S. Leedham2008Squamous gland ducts situated throughout the esophagusGene mutations
Table 2. Mutated genes in EAC as suggested by original research.
Table 2. Mutated genes in EAC as suggested by original research.
ReferenceAuthorsYearMutated GenesDetecting Methods
[122]A. Orsini et al.2023TP53, ATM, MSH6, APC, PIK3CA, SMAD4, CDKN2A, SMARCA4, ERBB2, HNF1A, CHEK2, FLT3, PTEN, IDH2, CTNNB1, MET, STK11, ALK, KRAS, RET, EGFR, ARID2, CDK6, TSSC1, MAP2K1, SRCTargeted sequencing
[120]A. Frankell et al.2019TP53, CDKN2A, KRAS, MYC, ERBB2, GATA4, CCND1, GATA6, SMAD4, CDK6, ARID1A, EGFR, CCNE1, CCND3, MUC6, MDM2, KCNQ3, APC, SMARCA4, PIK3CA, ABCB1, PTEN, MET, RNF43, DNAH7, TSHZ3, LRRK2, TRPA1, NAV3, ARID2, SLIT2, EPHA3, SCN3A, CRISPLD1, AXIN1, FBXW7, PPM1D, ACVR2A, RASA1, CD1A, CCDC102B, CHL1, LIN7A, COIL, MAP2K7, EPHA2, PBRM1, POLQ, ARID1B, CTNNB1, SIN3A, RPL22, PIK3R1, MAP3K1, NIPBL, B2M, FAM196B, HIST1H3B, TGFBR2, MBD6, BRAF, MSH3, CHD4, CDH1, GATAD1, KDM6A, CDKN1B, ACVR1B, STK11, NOTCH1, ZFHX3, JAK1, PCDH17, ELF3, GPATCH8, C3orf62Whole-genome sequencing
[121]D. Lin et al.2018PIK3CA, PBRM1, SMARCA4, CTNNB1, PCDH18, C6orf114, CHRNB1, EPHA2, SEMA5A, PGCP, DOCK2, CDKN2A, ARID1A, SMAD4, FBXW7, KRAS, TP53Whole-genome sequencing
[1]TCGA Research Network2017TP53, CDKN2A, ARID1A, SMAD4, ERBB2, VEGFA, GATA6, CCNE1, KRAS, EGFR, IGF1R, VEGFA, GATA4, ARID1A, SMARCA4, PBRM1Whole-exome sequencing
[123]M. Secrier et al.2016SMYD3, RUNX1, CTNNA3, RBFOX1, AGBL4, INK4/ARF, SAMD5, CDK14, KIF26B, THADA, SASH1, MECOM, JUP, IKZF3, FHIT, WWOX, MACROD2, IMMP2L, CCSER1, PDE4D, NAALADL2, PARK2, PARD3B, PRKG1, TP53, SMAD4, ARID1A, CDKN2A, KCNQ3, CCDC102B, CYP7B1Whole-genome sequencing
[106]J. Weaver et al.2014ABCB1, ARID1A, CCDC102B, CCDC153, CDKN2A, CNTNAP5, FGF10, MMP16, MYD88, MYF6, MYO18B, PCDH9, PNLIPRP3, SEMA5A, SMAD4, SMARCA4, SSTR4, TLR1, TLR4, TLR7, TLR9, TP53, TRAF3, TRAF6, TRIM58, UNC13CWhole-genome sequencing
[105]A. Dulak et al.2013TP53, CDKN2A, EYS, ARID1A, SMAD4, PIK3CA, SLC39A12, SPG20, DOCK2, AKAP6, TLL1, TLR4, ARID2, HECW1, ELMO1, SYNE1, SMARCA4, AJAP1, C6orf118, ACTL7B, F5, KCNU1, NUAK1, MYST3, SCN10A, CNTNAP5Whole-exome sequencing
Table 3. Genes with CNVs in EAC as suggested by original research.
Table 3. Genes with CNVs in EAC as suggested by original research.
ReferenceAuthorsYearsGenes with CNVsDetected Method
[129]Sihag et al.2021ERBB2, KRAS, CCNE1, MYC, CCND1, MDM2, VEGFA, EGFR, CDK6, CCND3, CDKN2A/B, SMAD4, ARID1A, PIK3CA, APC, TP53 NGSNext-generation sequencing
[120]Frankell et al.2019ERBB2, KRAS, SMAD4Whole-genome sequencing
[1]TCGA Research Network2017VEGFA, ERBB2, SMAD4, GATA6, CCNE1Whole-genome sequencing
[123]Secrier et al.201650 genes: CCND1, EGFR, ERBB2, VEGFA, KRAS etc.Whole-genome sequencing
[107]Ross-Innes et al.201535 genes: GATA4, KLF5, MYB, PRKCI, CCND1, FGF3, FGF4, FGF19, VEGFA, A2BP1, CDKN2A, PDE4D, PTPRD, PARK2 etc. Whole-genome sequencing
[108]Nones et al.2014210 genes: CCNE1, ERBB2, FRS2, GATA4, KRAS, MTMR9, MDM2, CDKN2A, FHIT, RUNX1 etc.Whole-genome sequencing
[130]Paulson et al.200947 genes: EGFR, MYC, EGFR, MTAP, CDKN2A, CDKN2B, SMAD2, SMAD4, SMAD7 etc.BAC array CGH
[128]Pasello et al.200997 genes: VEGF, PTK2, ING1, SCYA3, ABCG2, DCC etc.MLPA
CNVs: copy number variations; BAC: bacterial artificial chromosome; CGH: comparative genomic hybridization; MLPA: multiplex ligation-dependent probe amplification.
Table 4. Genes with DNA methylation in BE and EAC as suggested by original research.
Table 4. Genes with DNA methylation in BE and EAC as suggested by original research.
ReferenceAuthorsYearGenesStatusType of Lesion
[145]Xu et al.201320 genes: SH3GL3, GBX2, SLC18A3, SLC6A2 etc.hypermethylatedBE (vs. NE)
20 genes: ZNF625, PTPRT, ST6GAL2, SLC18A3 etc. hypermethylatedEAC (vs. NE)
[150]Alvi et al.2012SLC22A18, PIGR, GJA12, RIN2, RGN, TCEAL7hypermethylatedEAC (vs. BE)
[149]Peng et al.2011MT3hypermethylatedEAC (vs. NE)
[146]Smith et al.2008APC, CDKN2A, ID4, MGMT, RBP1, RUNX3, SFRP1, TIMP3, TMEFF2hypermethylatedEAC (vs. NE)
APC, ID4, MGMT, RUNX3, SFRP1, TIMP3, TMEFF2hypermethylatedBE (vs. NE)
[147]Klump et al.1998p16hypermethylatedBE (vs. NE)
[148]Wong et al.1997p16hypermethylatedEAC/BE (vs. NE)
BE: Barrett’s esophagus; EAC: esophageal adenocarcinoma; NE: normal esophagus.
Table 5. Altered microRNA expression during the development and progression of EAC.
Table 5. Altered microRNA expression during the development and progression of EAC.
ReferenceAuthorsYearMicroRNAStatusType of Lesion
[84]Neureiter et al.2020miR-205upregulatedLE-EAC (vs. RI-EAC)
[161]Mari et al.2018miR125a-5pdownregulated EAC (vs. NE)
[153]Kailasam et al.2015miR-28, miR-30a-5p, miR-126, miR-143, miR-145, miR-181a/b, miR-199aupregulatedEAC (vs. NE)
miR-27b, miR-99a, miR-149, miR-193a/b, miR-210, miR-345, miR-494, miR-513, miR-617, let-7a/b/cdownregulatedEAC (vs. NE)
[152]Huang et al.2014miR-126, miR-143, miR-145, miR-181a, miR-181b, miR-199a, miR-28, miR-30a-5pupregulatedEAC (vs. HGD)
miR-149, miR-203, miR-210, miR-27b, miR-513, miR-617, miR-99a let-7a/b/c, miR-193a, miR-345, miR-494downregulatedEAC (vs. HGD)
miR-25, miR-93, miR-106b, miR-192upregulatedEAC (vs. BE)
miR-203, let-7downregulatedEAC (vs. BE)
miR-200a, miR-513, miR-125b, miR-101, miR-197upregulatedHGD (vs. LGD)
miR-23b, miR-20b, miR-181b, miR-203, miR-193b, miR-636downregulatedHGD (vs. LGD)
[154]Saad et al.2013miR-194, miR-31, miR-192, miR-200aupregulatedEAC (vs. BE)
miR-203, miR-205downregulatedEAC (vs. BE)
miR-194, miR-192, miR-21, miR-31upregulatedHGD (vs. BE)
[155]Chen et al.2013miRNA-21, miRNA-133b, miR-200aupregulatedEAC (vs. NE)
[151]Fassan et al.2011miR-215, miR-560, miR-615-3p, miR-192, miR-326, miR-147upregulatedBE (vs. NE)
miR-100, miR-23a, miR-605, miR-99a, miR-205, let-7c, miR-203downregulatedBE (vs. NE)
BE: Barrett’s esophagus; EAC: esophageal adenocarcinoma; NE: normal esophagus; LGD: low-grade dysplasia; HGD: high-grade dysplasia; LE-EAC: localized and early esophageal adenocarcinoma; RI-EAC: regional and invasive esophageal adenocarcinoma.
Table 6. Currently employed targeted therapies and immunotherapies for EAC.
Table 6. Currently employed targeted therapies and immunotherapies for EAC.
TherapyTargetPatientsTreatmentApprovalClinical TrialsCurrent Status
Targeted therapyHER2HER2 positive metastatic EACTrastuzumab plus chemotherapyFDAPhase III (NCT01041404) [15]In clinic
Targeted therapyHER2HER2 positive metastatic EACFam-Trastuzumab Deruxtecan-nxkiFDAPhase II (NCT03329690) [197]In clinic
Monotherapy/
Combination therapy
VEGFR2advanced or metastatic EACRamucirumab (plus paclitaxel)FDAPhase III (NCT00917384; NCT01170663) [198,199]In clinic
ImmunotherapyPD-1resectable EACNivolumab following nCRT plus radical resectionFDA; EMAPhase III (NCT02743494) [200]In clinic
ImmunotherapyPD-1unresectable HER2 negative metastatic EACNivolumab plus chemotherapyFDA; EMAPhase III (NCT02872116) [201]In clinic
ImmunotherapyPD-1unresectable HER2 positive metastatic EACPembrolizumab plus Trastuzumab plus chemotherapyFDAPhase III (NCT03615326) [202]In clinic
ImmunotherapyPD-1locally advanced or metastatic EACPembrolizumab plus chemotherapyFDAPhase III (NCT03189719) [194]In clinic
HER2: human epidermal growth factor receptor 2; VEGFR2: vascular endothelial growth factor receptor 2; PD-1: programmed cell death protein 1; nCRT: neoadjuvant chemoradiotherapy; FDA: Food and Drug Administration; EMA: European Medicines Agency.
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

Li, S.; Hoefnagel, S.J.M.; Krishnadath, K.K. Molecular Biology and Clinical Management of Esophageal Adenocarcinoma. Cancers 2023, 15, 5410. https://doi.org/10.3390/cancers15225410

AMA Style

Li S, Hoefnagel SJM, Krishnadath KK. Molecular Biology and Clinical Management of Esophageal Adenocarcinoma. Cancers. 2023; 15(22):5410. https://doi.org/10.3390/cancers15225410

Chicago/Turabian Style

Li, Shulin, Sanne Johanna Maria Hoefnagel, and Kausilia Krishnawatie Krishnadath. 2023. "Molecular Biology and Clinical Management of Esophageal Adenocarcinoma" Cancers 15, no. 22: 5410. https://doi.org/10.3390/cancers15225410

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