Review Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Endosc. May 16, 2015; 7(5): 438-445
Published online May 16, 2015. doi: 10.4253/wjge.v7.i5.438
Risk factors affecting the Barrett's metaplasia-dysplasia-neoplasia sequence
Craig S Brown, Pritzker School of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, United States
Craig S Brown, Michael B Ujiki, Department of Surgery, North Shore University Health Systems, Evanston, IL 60201, United States
Author contributions: Brown CS and Ujiki MB solely contributed to this paper.
Conflict-of-interest: Dr. Michael B Ujiki has received consultant fees from Olympus and Covidien, as well as speaker honoraria from Covidien, Apollo Endo, and GORE. Craig S Brown has no conflicts of interest to disclose.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Michael B Ujiki, MD, Department of Surgery, North Shore University Health Systems, 2650 Ridge Avenue, Evanston, IL 60201, United States. mujiki@northshore.org
Telephone: +1-847-5701700
Received: August 29, 2014
Peer-review started: August 30, 2014
First decision: October 14, 2014
Revised: November 25, 2014
Accepted: January 18, 2015
Article in press: January 20, 2015
Published online: May 16, 2015

Abstract

Esophageal adenocarcinoma has the fastest growing incidence rate of any cancer in the United States, and currently carries a very poor prognosis with 5 years relative survival rates of less than 15%. Current curative treatment options are limited to esophagectomy, a procedure that suffers from high complication rates and high mortality rates. Metaplasia of the esophageal epithelium, a condition known as Barrett’s esophagus (BE), is widely accepted as the precursor lesion for adenocarcinoma of the esophagus. Recently, radio-frequency ablation has been shown to be an effective method to treat BE, although there is disagreement as to whether radio-frequency ablation should be used to treat all patients with BE or whether treatment should be reserved for those at high risk for progressing to esophageal adenocarcinoma while continuing to endoscopically survey those with low risk. Recent research has been targeted towards identifying those at greater risk for progression to esophageal adenocarcinoma so that radio-frequency ablation therapy can be used in a more targeted manner, decreasing the total health care cost as well as improving patient outcomes. This review discusses the current state of the literature regarding risk factors for progression from BE through dysplasia to esophageal adenocarcinoma, as well as the current need for an integrated scoring tool or risk stratification system capable of differentiating those patients at highest risk of progression in order to target these endoluminal therapies.

Key Words: Barrett’s esophagus, Esophageal adenocarcinoma, Endoscopy, Risk factors, Radiofrequency ablation, Antireflux surgery

Core tip: The transformation of Barrett’s esophagus to dysplasia and finally to esophageal adenocarcinoma is a multifactorial process encompassing effects from multiple known and unknown risk factors. Previously, radiofrequency ablation was reserved for use in high risk patients with high-grade dysplasia, but recent evidence supports the expansion of this technique to be potentially used to treat additional patients at moderate risk of progression, such as those with long segments, long duration of symptoms, and those patients who are unable or unwilling to take proton-pump inhibitors’s.



INTRODUCTION

Gastroesophageal reflux disease (GERD) has been estimated to affect nearly 20% of the United States population at any given time[1]. Of this group suffering from GERD, roughly 15% are estimated to have Barrett’s esophagus (BE), a condition characterized by columnar-lined epithelium in the esophagus[2]. It is well established that BE is the link between GERD and esophageal adenocarcinoma (EAC), a neoplastic lesion with an extremely poor prognosis with 5-year survival rates of less than 15% and which currently has the fastest rising incidence rate of any cancer with approximately a 10-fold increased incidence rate among men aged 15-74 in the last 40 years[3-6]. Unfortunately, little progress has been made in treating this extremely aggressive cancer, with median survival time increasing only 3.2 mo over the last 30 years[7]. BE has been shown to be a paradigmatic model for progression from metaplastic disease through dysplasia to neoplasia[8]. In this review, we summarize the current literature regarding the etiology and pathophysiology of BE and EAC.

RESEARCH

We performed a literature review in the PubMed/Medline database using MeSH term “Barrett’s Esophagus” combined with subheadings “etiology”, “physiopathology”, “therapy”, “diagnosis” and “epidemiology” as well as MeSH term “Esophageal Neoplasms” with selected subheadings “diagnosis”, “etiology”, “physiopathology”, “epidemiology” and “therapy” combined by Boolean operator AND with MeSH term “Adenocarcinoma” with selected subheadings “diagnosis”, “epidemiology”, “etiology”, “pathophysiology” and “therapy”. We reviewed abstracts published between 1980 and April of 2013 in English and selected articles relevant to topics discussed herein.

EPIDEMIOLOGY

Columnar lined epithelium has been shown to be present in almost 25% of individuals with GERD symptoms, and columnar lined epithelium with intestinal metaplasia is reported as affecting almost 15% of those with GERD symptoms. The probability of progressing to EAC from BE has been estimated to be approximately 0.5%/year[9,10], with the most convincing evidence provided in a meta-analysis of 47 studies by Yousef et al[11] showing a pooled cancer incidence of 6.4/1000 person-years for the 13 studies conducted in the United States and 6.1/1000 person-years for all 47 studies pooled. EAC incidence has increased roughly 10-fold in select demographics over the last 40 years, with only a small fraction being attributed to increasing obesity rates[12]. Recent data suggests this increase is slowing but still substantial, with average annual percentage increase in incidence rising 6.1% in men and 5.9% in women[13]. Other causes for this rapid increase in incidence have yet to be elucidated, but this continues to be a highly active area of research.

ETIOLOGY AND PATHOPHYSIOLOGY

BE is caused by reflux of gastric contents into the esophagus, which causes damage to the stratified squamous epithelium. Not surprisingly, it has also been shown that GERD symptoms increase odds of EAC by 7.7 fold, odds which increase to 43.5 fold when comparing patients with long-standing and severe GERD symptoms[13]. It is currently contested as to whether gastric acid, bile reflux, or the combination is responsible. Several studies have shown increased intraluminal bilirubin content, a proxy for duodenal juice content, in patients with BE, suggesting that bile acid plays an important role in BE development[14]. Likewise, gallbladder function was shown to be impaired in patients with BE and EAC in a real-time ultrasonography experiment following a 10-h fast leading to increased duodenogastric reflux[15]. Cholecystectomy has also been shown to increase risk of EAC, albeit slightly[16,17]. The body’s compensatory mechanism can but does not always include metaplasia in the form of simple columnar epithelium, which is thought to be more tolerant to the low pH[18,19]. BE is the most predictive risk factor for the development of EAC, with a relative risk for developing esophageal cancer of 11.3 when compared to the general population[20]. Much research recently has been focused on determining what the risk factors are for developing BE. Age has been shown to be correlated with increased risk of developing BE, with a low of 2 diagnoses per 100000 person-years for those aged 21-30 years and peaking at 31 diagnoses per 100000 person-years in those aged 61-70 years[21]. Males also experience BE incidence rates roughly twice that of females, although the reason for this difference remains to be elucidated[21].

Obesity and its related conditions have been shown to be a risk factor for many diseases, and BE is no exception. A meta-analysis by Cook et al[22] suggests that increasing obesity is correlated with an increased risk for BE development but only indirectly due to obesity’s effect on GERD development. This view is contested by El-Serag et al[23], who suggest that increasing visceral adipose tissue to subcutaneous adipose tissue ratio is correlated with the presence of BE [adjusted OR = 1.47 (95%CI: 0.92 to 4.09)] as well as Kendall et al[24], whose data shows a significant correlation between all measures of obesity tracked (waist circumference, waist-hip ratio, sagittal abdominal diameter, and waist-height ratio) and presence of BE in males even after adjusting for GERD symptoms. It has been proposed that the association between obesity and risk of BE is due to several factors including increased intra-abdominal pressure leading to worsening GERD, as well as increased circulating levels of leptin, adiponectin, and other chemicals secreted by adipose tissue, although this link remains to be confirmed. Recently, low birth weight and preterm birth have been implicated as a risk factor for BE, with several studies reporting those born very small for gestational age, < 3rd percentile in one study and < 2000 g in another, having between a three and eleven-fold increase in odds when compared to those born at a normal weight for gestational age[25,26]. Hiatal hernia has been shown to be another risk factor for BE, with size of hiatal hernia correlating with increasing risk of both BE as well EAC[27,28]. Metabolic syndrome, another obesity related factor, has been shown to increase risk for BE by two-fold relative to those without metabolic syndrome[29].

It is being currently debated as to whether Helicobacter pylori (H. pylori) infection leads to increased or decreased risk of developing BE, but two meta-analyses, of 49 studies conducted by Fischbach et al[30] and 19 studies conducted by Islami et al[31], both suggest that, although significant selection and information bias may be present in these studies, H. pylori infection appears to be associated with a decreased risk of BE. Aggressive eradication of H. pylori infection over the last 30 years may provide an explanation for a small portion of the drastic increase in incidence.

Along with being male and older age[32-34], those with low dietary antioxidant intake have also been shown to not only have an increased risk of developing BE, but also have an increased risk of developing EAC[35,36]. Similarly, length of GERD symptoms is a risk factor for both development of BE as well as EAC[36,37]. The reasons for males experiencing high incidence rates is not well understood, but it appears to be due to other reasons than differential exposure to known risk factors[38,39]. Hormonal factors, studied by comparing patients undergoing hormone therapy, do not appear to account for the discrepancy in EAC incidence rates between males and females[40]. Heme iron intake in the diet has been suggested as a risk factor corresponding to EAC development as well[41]. Dietary iron has been shown to be a growth factor for H. pylori, making this association one in need of further investigation.

Many studies recently have elucidated relationships between various risk factors and the development of EAC, a goal that has potential to directly affect patient outcomes and change clinical practice with respect to ablative therapy. Sikkema et al[42] conducted a prospective cohort study in which they found statistically significant associations between many risk factors and progression to high grade dysplasia (HGD) and/or EAC including esophagitis and length of BE segment, with a risk ratio of 1.11 per centimeter increase in length, and known duration of BE of greater than or equal to 10 years with a risk ratio of 3.2. Also, previous partial gastrectomy is linked to EAC development[43]. Patients who underwent esophagectomy for EAC were shown in a case-control study to have a 45% prevalence of colonic polyps when compared to control patients who also underwent screening colonoscopies, of whom only 14% were shown to have colon polyps[44]. Whether there is a predictive relationship between presence of colon polyps and risk of EAC is still a contested topic and deserves further attention. Also, early research shows no evidence of viral genomic sequences present in tumors[45]. The single most predictive clinical factor for progression to HGD and/or EAC found to date is the presence of low grade dysplasia (LGD) found during biopsy with a relative risk of 9.7 (95%CI: 4.4-21.5) according to Sikkema et al and 5.5 (95%CI: 1.1-28.6) according to Oberg et al[46] compared to those without LGD.

Biomarkers have the potential to drastically improve our ability to risk stratify. p53 as well as KI-67, both proteins involved in cell cycle progression, have been shown to be expressed at higher levels in BE samples that progress to EAC[47-51]. Likewise, it has also been shown that cell-free circulating DNA methylation patterns correlate extremely closely (r = 0.92) with aberrant DNA methylation patterns in matched tumor tissue in patients with EAC and also that 911 loci for DNA methylation could perfectly discriminate between EAC and controls, suggesting that cell-free DNA methylation patterns could be used as a non-invasive method to screen premalignant lesions[52]. Promoter hypermethylation of p16 and APC is also strongly correlated with progression to EAC, with one study reporting hypermethylation of p16 and APC, either separately or together, in over 50% of HGD/EAC samples with hypermethylation of the same promoters totally absent in samples from patients with normal esophagus[53]. In a similar way, Mcm2 expression in BE is directly correlated with degree of dysplasia, with 91% of patients diagnosed with dysplasia or EAC in one prospective cohort showing Mcm-2-positive cytological brushings, while brushings from controls without BE showed no signs of Mcm-2 expression on the luminal surface[54]. COX-2 expression is upregulated in BE patients and degree of overexpression is correlated with risk of malignant transformation, suggesting that COX-2 expression could be used as a potential marker as well[55]. This increase in COX-2 expression has been shown to be strongly induced by deoxycholic acid incubation in vitro using OE-19 cells as a Barrett’s model, suggesting a potential mechanism for this phenomenon[56]. Several bile acids have also been shown to induce the expression of other proteins important in cancer progression such as CDX2 as well as induce NF-κB signaling[57]. Other notable biomarkers include increased DNA damage detected by Comet Assay, decreased Beclin-1 expression, increased cyclin A, cyclin B1, and cyclin D1 expression, and abnormal DNA content[49,58-65]. Notably, abnormal DNA content, measured by the number of chromosomes arms with loss, has been shown to be directly correlated with the progression from metaplasia, through low and high grade dysplasia, and finally to neoplasia[66]. Likewise, telomerase reverse transcriptase has been shown to be overexpressed in increasing levels along the metaplasia-dyplasia-neoplasia sequence of BE[67]. Whether these two markers can be used to differentiate between BE patients who will progress and those who will not remains to be studied. The field would benefit from further research into how these biomarkers can be integrated and utilized in a clinical setting as well as which can be used cost effectively to better predict risk of progression to EAC.

Interestingly, high serum leptin levels were associated with increased risk of EA, whereas increased levels of high molecular weight adiponectin conferred a protective effect, with a hazard ratio (HR) of 0.34 (95%CI: 0.14-0.82)[68]. The mechanism for this association might be due to leptin’s effect on proliferation of adenocarcinoma cells independent of apoptosis or necrosis, as has been shown in BIC-1 and SEG-1 cells in vitro[69]. Type 2 diabetes mellitus has been shown to be more prevalent in those diagnosed with EAC, although the effect was attenuated after controlling for differences in BMI[70].

The consumption of several substances have shown to confer protective effects, with use of a multivitamin pill showing a HR of 0.38 (95%CI: 0.15-0.99) when compared to those not taking a multivitamin[71]. Vitamin D intake, however, was found to increase the risk of EAC, showing an OR of 1.99 (95%CI: 1.03-3.86), although vitamin D intake was not associated with BE or reflux esophagitis[72]. Taking proton-pump inhibitors (PPIs) has been shown to confer a protective affect against progressing from BE to EAC, with a hazard ratio of 0.41 (95%CI: 0.18-0.93) and 0.21 (95%CI: 0.07-0.66) for those using proton pump inhibitors at inclusion of the study or during the follow-up period, respectively; a finding supported by several other studies[73,74]. In addition to the use of proton-pump inhibitors, several studies recently have shown decreased rates of progression to EAC from BE when taking aspirin and/or statins, although the mechanism for this protection remains to be elucidated fully[75]. Sadaria et al[76] found that simvastatin attenuated growth and increased apoptosis in human esophageal adenocarcinoma (FLO-1) cells in tissue culture, providing one potential mechanism by which statins reduce risk of progression to EAC. One meta-analysis investigating this protective effect found a number needed to treat of 389 patients with statins to prevent one case of EAC[77]. ACE inhibitors could potentially provide a protective effect, although studies regarding this question were underpowered[78]. Medications that have relaxing effects on the lower-esophageal sphincter, specifically anticholinergics and theophyllines, have been associated with a roughly 1.5-2.5 fold increased risk of EAC, a relationship not seen for other types of cancers of the upper digestive tract[79,80].

As is expected, tobacco smoking has been shown repeatedly to increase the probability of progression to EAC. Interestingly, one study from the NIH Barrett’s Esophagus and Esophageal Adenocarcinoma Consortium found an increased risk of progression to EAC with smoking and even showed a dose-response effect when considering pack-years, but there was a weaker association when considering cigarettes/day[81]. This study corroborates several other studies showing deleterious effects of smoking on risk of progression to EAC, estimating the risk at roughly double for those who smoke relative to those who do not smoke[82-85]. There appears to be no association between alcohol intake and risk of EAC according to several recent studies including meta-analysis, although this has been contested according to a matched case-control study out of North China[81,83,86-88].

Currently, no definitive genetic cause of BE or EAC has been identified. Several case reports, however, have found a remarkable history of BE and EAC among members of the same family, providing evidence that a subset of the population may be genetically susceptible to BE and potential progression to EAC[89-92]. Additionally, a single nucleotide polymorphism in the gene coding for epidermal growth factor (EGF) has been shown to be associated with decreased levels of EGF expression and has also been shown to be more prevalent in patients with BE and EAC[93]. Further research in this area could help identify specific genotypes that would allow clinicians additional tools when risk stratifying patients and making decisions regarding the management of patients with BE.

Surgical management of GERD has been shown to decrease odds of progression to EAC compared to no therapy, however a 2007 systematic review found that, in controlled studies, there was no statistically significant difference in EAC incidence rates between patients treated surgically and those treated medically. If data from uncontrolled case-series are included, the difference becomes significant. Interestingly, surgical management increased the probability of regression of BE and/or dysplasia by almost 15%[94]. This study shows puzzling results given the data from previous studies showing that fundoplication can reduce or even eliminate the reflux of bile acids into the esophagus, compared to medical therapy which only treats the reflux of hydrochloric acid[95]. One possible answer to this question could come from recent case-control data showing that, among patients who’ve undergone antireflux surgery, those with recurrent reflux symptoms are three times more likely to develop EAC than those without, underscoring the importance of addressing continuing reflux symptoms after antireflux surgery[96]. Randomized trials to date have only compared antireflux surgery to medical therapy in patients who were complete responders to medical therapy. This, unfortunately, is not the comparison of interest given the current role for surgery in GERD management. Patients selected for antireflux surgery in practice almost exclusively have failed medical therapy as their indication for surgical management. This suggests that there is some fundamental difference between patients who are responders and those who are not, and limits the usefulness of the comparison in these studies. The question of antireflux surgery vs continued medical management remains unanswered conclusively, but continues to be an active area of research and could benefit heavily from a randomized controlled trial comparing antireflux surgery to continued medical management in a population of patients who have continued reflux symptoms despite full dose medical therapy. Current data suggests that there is a role for antireflux surgery in the management of patients with BE, but the question of exactly which patients should be receiving these procedures remains to be answered.

CONCLUSION

As can be seen from the wealth of information outlined above, the risks associated with progression from BE to esophageal adenocarcinoma are multifactorial, with many different risk factors each contributing a relatively small portion to the overall risk of progression. This suggests that a single intervention aimed at reducing exposure to individual risk factors other than refluxed gastric contents is unlikely to have a drastic impact on increasing adenocarcinoma rates or to affect the risk for individual patients with Barrett’s. Currently, no biomarkers have shown to be clinically useful in BE, but this continues to be an active area of research. More work is necessary to investigate the many risk factors at play and the populations that they apply to, in order to better understand the contributions to risk for any given clinical situation.Recent advancements in knowledge of risk factors and their contributions to progression have made clinical risk stratification models possible in order to target endoluminal therapies capable of eradicating Barrett’s tissue and drastically decreasing risk of progression to adenocarcinoma. Currently, these tools are not widely available. Additional work is required to further develop and validate these tools in order to target patients at the highest risk of progression with either therapeutic intervention or endoscopic surveillance. One risk factor, the presence of LGD, has been very clearly shown to drastically increase the risk of progression to EAC by multiple studies. Given this information along with the known safety and efficacy of radiofrequency ablation and other endoluminal therapies, we believe that there is sufficient data to support the use of RFA in all Barrett’s patients with LGD, even in the absence of additional risk factors. Additional stratification tools are required in order to dictate exactly which patients without LGD should receive RFA/endoluminal therapy and which should not, but given the evidence outlined above, patients with very long segment, patients who have had reflux symptoms for time periods of 10 years or greater, or patients who are unable or unwilling to take PPI’s or are not antireflux surgery candidates should be considered carefully as potential candidates for endoscopic ablation.

Footnotes

P- Reviewer: Alsolaiman M, Amornyotin S, Goral V S- Editor: Song XX L- Editor: A E- Editor: Wu HL

References
1.  Sobieraj DM, Coleman SM, Coleman CI. US prevalence of upper gastrointestinal symptoms: a systematic literature review. Am J Manag Care. 2011;17:e449-e458.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Balasubramanian G, Singh M, Gupta N, Gaddam S, Giacchino M, Wani SB, Moloney B, Higbee AD, Rastogi A, Bansal A. Prevalence and predictors of columnar lined esophagus in gastroesophageal reflux disease (GERD) patients undergoing upper endoscopy. Am J Gastroenterol. 2012;107:1655-1661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
3.  Devesa SS, Blot WJ, Fraumeni JF. Changing patterns in the incidence of esophageal and gastric carcinoma in the United States. Cancer. 1998;83:2049-2053.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Lepage C, Rachet B, Jooste V, Faivre J, Coleman MP. Continuing rapid increase in esophageal adenocarcinoma in England and Wales. Am J Gastroenterol. 2008;103:2694-2699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 314]  [Article Influence: 19.6]  [Reference Citation Analysis (1)]
5.  Sihvo EI, Luostarinen ME, Salo JA. Fate of patients with adenocarcinoma of the esophagus and the esophagogastric junction: a population-based analysis. Am J Gastroenterol. 2004;99:419-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 56]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
6.  Polednak AP. Trends in survival for both histologic types of esophageal cancer in US surveillance, epidemiology and end results areas. Int J Cancer. 2003;105:98-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 134]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
7.  Crane SJ, Locke GR, Harmsen WS, Zinsmeister AR, Romero Y, Talley NJ. Survival trends in patients with gastric and esophageal adenocarcinomas: a population-based study. Mayo Clin Proc. 2008;83:1087-1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
8.  Miros M, Kerlin P, Walker N. Only patients with dysplasia progress to adenocarcinoma in Barrett’s oesophagus. Gut. 1991;32:1441-1446.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Dulai GS, Shekelle PG, Jensen DM, Spiegel BM, Chen J, Oh D, Kahn KL. Dysplasia and risk of further neoplastic progression in a regional Veterans Administration Barrett’s cohort. Am J Gastroenterol. 2005;100:775-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 77]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
10.  Bhat S, Coleman HG, Yousef F, Johnston BT, McManus DT, Gavin AT, Murray LJ. Risk of malignant progression in Barrett’s esophagus patients: results from a large population-based study. J Natl Cancer Inst. 2011;103:1049-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 500]  [Cited by in F6Publishing: 489]  [Article Influence: 37.6]  [Reference Citation Analysis (0)]
11.  Yousef F, Cardwell C, Cantwell MM, Galway K, Johnston BT, Murray L. The incidence of esophageal cancer and high-grade dysplasia in Barrett’s esophagus: a systematic review and meta-analysis. Am J Epidemiol. 2008;168:237-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 287]  [Cited by in F6Publishing: 307]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
12.  Kong CY, Nattinger KJ, Hayeck TJ, Omer ZB, Wang YC, Spechler SJ, McMahon PM, Gazelle GS, Hur C. The impact of obesity on the rise in esophageal adenocarcinoma incidence: estimates from a disease simulation model. Cancer Epidemiol Biomarkers Prev. 2011;20:2450-2456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
13.  Hur C, Miller M, Kong CY, Dowling EC, Nattinger KJ, Dunn M, Feuer EJ. Trends in esophageal adenocarcinoma incidence and mortality. Cancer. 2013;119:1149-1158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 359]  [Cited by in F6Publishing: 374]  [Article Influence: 31.2]  [Reference Citation Analysis (0)]
14.  Kauer WK, Peters JH, DeMeester TR, Ireland AP, Bremner CG, Hagen JA. Mixed reflux of gastric and duodenal juices is more harmful to the esophagus than gastric juice alone. The need for surgical therapy re-emphasized. Ann Surg. 1995;222:525-531; discussion 531-533.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Nassr AO, Gilani SN, Atie M, Abdelhafiz T, Connolly V, Hickey N, Walsh TN. Does impaired gallbladder function contribute to the development of Barrett’s esophagus and esophageal adenocarcinoma? J Gastrointest Surg. 2011;15:908-914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
16.  Lagergren J, Mattsson F. Cholecystectomy as a risk factor for oesophageal adenocarcinoma. Br J Surg. 2011;98:1133-1137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
17.  Freedman J, Ye W, Näslund E, Lagergren J. Association between cholecystectomy and adenocarcinoma of the esophagus. Gastroenterology. 2001;121:548-553.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Spechler SJ. Clinical practice. Barrett’s Esophagus. N Engl J Med. 2002;346:836-842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 432]  [Cited by in F6Publishing: 381]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
19.  Fein M, Peters JH, Chandrasoma P, Ireland AP, Oberg S, Ritter MP, Bremner CG, Hagen JA, DeMeester TR. Duodenoesophageal reflux induces esophageal adenocarcinoma without exogenous carcinogen. J Gastrointest Surg. 1998;2:260-268.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Hvid-Jensen F, Pedersen L, Drewes AM, Sørensen HT, Funch-Jensen P. Incidence of adenocarcinoma among patients with Barrett’s esophagus. N Engl J Med. 2011;365:1375-1383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 985]  [Cited by in F6Publishing: 918]  [Article Influence: 70.6]  [Reference Citation Analysis (1)]
21.  Corley DA, Kubo A, Levin TR, Block G, Habel L, Rumore G, Quesenberry C, Buffler P. Race, ethnicity, sex and temporal differences in Barrett’s oesophagus diagnosis: a large community-based study, 1994-2006. Gut. 2009;58:182-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 122]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
22.  Cook MB, Greenwood DC, Hardie LJ, Wild CP, Forman D. A systematic review and meta-analysis of the risk of increasing adiposity on Barrett’s esophagus. Am J Gastroenterol. 2008;103:292-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 98]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
23.  El-Serag HB, Hashmi A, Garcia J, Richardson P, Alsarraj A, Fitzgerald S, Vela M, Shaib Y, Abraham NS, Velez M. Visceral abdominal obesity measured by CT scan is associated with an increased risk of Barrett’s oesophagus: a case-control study. Gut. 2014;63:220-229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 85]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
24.  Kendall BJ, Macdonald GA, Hayward NK, Prins JB, O’Brien S, Whiteman DC. The risk of Barrett’s esophagus associated with abdominal obesity in males and females. Int J Cancer. 2013;132:2192-2199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
25.  Forssell L, Cnattingius S, Bottai M, Edstedt Bonamy AK, Lagergren J, Agréus L, Akre O. Increased risk of Barrett’s esophagus among individuals born preterm or small for gestational age. Clin Gastroenterol Hepatol. 2013;11:790-794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
26.  Kaijser M, Akre O, Cnattingius S, Ekbom A. Preterm birth, low birth weight, and risk for esophageal adenocarcinoma. Gastroenterology. 2005;128:607-609.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Andrici J, Tio M, Cox MR, Eslick GD. Hiatal hernia and the risk of Barrett’s esophagus. J Gastroenterol Hepatol. 2013;28:415-431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 54]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
28.  Avidan B, Sonnenberg A, Schnell TG, Chejfec G, Metz A, Sontag SJ. Hiatal hernia size, Barrett’s length, and severity of acid reflux are all risk factors for esophageal adenocarcinoma. Am J Gastroenterol. 2002;97:1930-1936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 201]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
29.  Leggett CL, Nelsen EM, Tian J, Schleck CB, Zinsmeister AR, Dunagan KT, Locke GR, Wang KK, Talley NJ, Iyer PG. Metabolic syndrome as a risk factor for Barrett esophagus: a population-based case-control study. Mayo Clin Proc. 2013;88:157-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 31]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
30.  Fischbach LA, Nordenstedt H, Kramer JR, Gandhi S, Dick-Onuoha S, Lewis A, El-Serag HB. The association between Barrett’s esophagus and Helicobacter pylori infection: a meta-analysis. Helicobacter. 2012;17:163-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
31.  Islami F, Kamangar F. Helicobacter pylori and esophageal cancer risk: a meta-analysis. Cancer Prev Res (Phila). 2008;1:329-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 249]  [Article Influence: 16.6]  [Reference Citation Analysis (1)]
32.  Löfdahl HE, Lu Y, Lagergren J. Sex-specific risk factor profile in oesophageal adenocarcinoma. Br J Cancer. 2008;99:1506-1510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
33.  Vizcaino AP, Moreno V, Lambert R, Parkin DM. Time trends incidence of both major histologic types of esophageal carcinomas in selected countries, 1973-1995. Int J Cancer. 2002;99:860-868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 304]  [Cited by in F6Publishing: 295]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
34.  de Jonge PJ, van Blankenstein M, Looman CW, Casparie MK, Meijer GA, Kuipers EJ. Risk of malignant progression in patients with Barrett’s oesophagus: a Dutch nationwide cohort study. Gut. 2010;59:1030-1036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 203]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
35.  Ibiebele TI, Hughes MC, Nagle CM, Bain CJ, Whiteman DC, Webb PM. Dietary antioxidants and risk of Barrett’s esophagus and adenocarcinoma of the esophagus in an Australian population. Int J Cancer. 2013;133:214-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
36.  Pohl H, Wrobel K, Bojarski C, Voderholzer W, Sonnenberg A, Rösch T, Baumgart DC. Risk factors in the development of esophageal adenocarcinoma. Am J Gastroenterol. 2013;108:200-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 107]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
37.  Rubenstein JH, Taylor JB. Meta-analysis: the association of oesophageal adenocarcinoma with symptoms of gastro-oesophageal reflux. Aliment Pharmacol Ther. 2010;32:1222-1227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 133]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
38.  Rutegård M, Nordenstedt H, Lu Y, Lagergren J, Lagergren P. Sex-specific exposure prevalence of established risk factors for oesophageal adenocarcinoma. Br J Cancer. 2010;103:735-740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
39.  Freedman ND, Derakhshan MH, Abnet CC, Schatzkin A, Hollenbeck AR, McColl KE. Male predominance of upper gastrointestinal adenocarcinoma cannot be explained by differences in tobacco smoking in men versus women. Eur J Cancer. 2010;46:2473-2478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
40.  Bodelon C, Anderson GL, Rossing MA, Chlebowski RT, Ochs-Balcom HM, Vaughan TL. Hormonal factors and risks of esophageal squamous cell carcinoma and adenocarcinoma in postmenopausal women. Cancer Prev Res (Phila). 2011;4:840-850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 46]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
41.  Ward MH, Cross AJ, Abnet CC, Sinha R, Markin RS, Weisenburger DD. Heme iron from meat and risk of adenocarcinoma of the esophagus and stomach. Eur J Cancer Prev. 2012;21:134-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
42.  Sikkema M, Looman CW, Steyerberg EW, Kerkhof M, Kastelein F, van Dekken H, van Vuuren AJ, Bode WA, van der Valk H, Ouwendijk RJ. Predictors for neoplastic progression in patients with Barrett’s Esophagus: a prospective cohort study. Am J Gastroenterol. 2011;106:1231-1238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 128]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
43.  Tsibouris P, Hendrickse MT, Kalantzis C, Isaacs PE. Patients with partial gastrectomy and Barrett esophagus are in higher risk to develop esophageal adenocarcinoma than those with Barretts without gastrectomy. Hepatogastroenterology. 2012;59:1118-1122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
44.  Bollschweiler E, Schloesser T, Leers J, Vallböhmer D, Schäfer H, Hölscher AH. High prevalence of colonic polyps in white males with esophageal adenocarcinoma. Dis Colon Rectum. 2009;52:299-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
45.  Morgan RJ, Perry AC, Newcomb PV, Hardwick RH, Alderson D. Investigation of oesophageal adenocarcinoma for viral genomic sequences. Eur J Surg Oncol. 1997;23:24-29.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Oberg S, Wenner J, Johansson J, Walther B, Willén R. Barrett esophagus: risk factors for progression to dysplasia and adenocarcinoma. Ann Surg. 2005;242:49-54.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Murray L, Sedo A, Scott M, McManus D, Sloan JM, Hardie LJ, Forman D, Wild CP. TP53 and progression from Barrett’s metaplasia to oesophageal adenocarcinoma in a UK population cohort. Gut. 2006;55:1390-1397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 96]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
48.  Binato M, Gurski RR, Fagundes RB, Meurer L, Edelweiss MI. P53 and Ki-67 overexpression in gastroesophageal reflux disease--Barrett’s esophagus and adenocarcinoma sequence. Dis Esophagus. 2009;22:588-595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
49.  Shi XY, Bhagwandeen B, Leong AS. p16, cyclin D1, Ki-67, and AMACR as markers for dysplasia in Barrett esophagus. Appl Immunohistochem Mol Morphol. 2008;16:447-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 24]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
50.  Kerkhof M, Steyerberg EW, Kusters JG, van Dekken H, van Vuuren AJ, Kuipers EJ, Siersema PD. Aneuploidy and high expression of p53 and Ki67 is associated with neoplastic progression in Barrett esophagus. Cancer Biomark. 2008;4:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Polkowski W, van Lanschot JJ, Ten Kate FJ, Baak JP, Tytgat GN, Obertop H, Voorn WJ, Offerhaus GJ. The value of p53 and Ki67 as markers for tumour progression in the Barrett’s dysplasia-carcinoma sequence. Surg Oncol. 1995;4:163-171.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Zhai R, Zhao Y, Su L, Cassidy L, Liu G, Christiani DC. Genome-wide DNA methylation profiling of cell-free serum DNA in esophageal adenocarcinoma and Barrett esophagus. Neoplasia. 2012;14:29-33.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Wang JS, Guo M, Montgomery EA, Thompson RE, Cosby H, Hicks L, Wang S, Herman JG, Canto MI. DNA promoter hypermethylation of p16 and APC predicts neoplastic progression in Barrett’s esophagus. Am J Gastroenterol. 2009;104:2153-2160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 88]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
54.  Sirieix PS, O’Donovan M, Brown J, Save V, Coleman N, Fitzgerald RC. Surface expression of minichromosome maintenance proteins provides a novel method for detecting patients at risk for developing adenocarcinoma in Barrett’s esophagus. Clin Cancer Res. 2003;9:2560-2566.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Majka J, Rembiasz K, Migaczewski M, Budzynski A, Ptak-Belowska A, Pabianczyk R, Urbanczyk K, Zub-Pokrowiecka A, Matlok M, Brzozowski T. Cyclooxygenase-2 (COX-2) is the key event in pathophysiology of Barrett’s esophagus. Lesson from experimental animal model and human subjects. J Physiol Pharmacol. 2010;61:409-418.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Burnat G, Majka J, Konturek PC. Bile acids are multifunctional modulators of the Barrett’s carcinogenesis. J Physiol Pharmacol. 2010;61:185-192.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Debruyne PR, Witek M, Gong L, Birbe R, Chervoneva I, Jin T, Domon-Cell C, Palazzo JP, Freund JN, Li P. Bile acids induce ectopic expression of intestinal guanylyl cyclase C Through nuclear factor-kappaB and Cdx2 in human esophageal cells. Gastroenterology. 2006;130:1191-1206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
58.  Olliver JR, Hardie LJ, Gong Y, Dexter S, Chalmers D, Harris KM, Wild CP. Risk factors, DNA damage, and disease progression in Barrett’s esophagus. Cancer Epidemiol Biomarkers Prev. 2005;14:620-625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 66]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
59.  Bird-Lieberman EL, Dunn JM, Coleman HG, Lao-Sirieix P, Oukrif D, Moore CE, Varghese S, Johnston BT, Arthur K, McManus DT. Population-based study reveals new risk-stratification biomarker panel for Barrett’s esophagus. Gastroenterology. 2012;143:927-935.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 134]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
60.  Reid BJ, Levine DS, Longton G, Blount PL, Rabinovitch PS. Predictors of progression to cancer in Barrett’s esophagus: baseline histology and flow cytometry identify low- and high-risk patient subsets. Am J Gastroenterol. 2000;95:1669-1676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 194]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
61.  Roesly HB, Khan MR, Chen HD, Hill KA, Narendran N, Watts GS, Chen X, Dvorak K. The decreased expression of Beclin-1 correlates with progression to esophageal adenocarcinoma: the role of deoxycholic acid. Am J Physiol Gastrointest Liver Physiol. 2012;302:G864-G872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
62.  Lao-Sirieix P, Lovat L, Fitzgerald RC. Cyclin A immunocytology as a risk stratification tool for Barrett’s esophagus surveillance. Clin Cancer Res. 2007;13:659-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 57]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
63.  Bani-Hani K, Martin IG, Hardie LJ, Mapstone N, Briggs JA, Forman D, Wild CP. Prospective study of cyclin D1 overexpression in Barrett’s esophagus: association with increased risk of adenocarcinoma. J Natl Cancer Inst. 2000;92:1316-1321.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Brankley SM, Fritcher EG, Smyrk TC, Keeney ME, Campion MB, Voss JS, Clayton AC, Wang KK, Lutzke LS, Kipp BR. Fluorescence in situ hybridization mapping of esophagectomy specimens from patients with Barrett’s esophagus with high-grade dysplasia or adenocarcinoma. Hum Pathol. 2012;43:172-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
65.  Geddert H, Heep HJ, Gabbert HE, Sarbia M. Expression of cyclin B1 in the metaplasia-dysplasia-carcinoma sequence of Barrett esophagus. Cancer. 2002;94:212-218.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Gu J, Ajani JA, Hawk ET, Ye Y, Lee JH, Bhutani MS, Hofstetter WL, Swisher SG, Wang KK, Wu X. Genome-wide catalogue of chromosomal aberrations in barrett’s esophagus and esophageal adenocarcinoma: a high-density single nucleotide polymorphism array analysis. Cancer Prev Res (Phila). 2010;3:1176-1186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 70]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
67.  Lord RV, Salonga D, Danenberg KD, Peters JH, DeMeester TR, Park JM, Johansson J, Skinner KA, Chandrasoma P, DeMeester SR. Telomerase reverse transcriptase expression is increased early in the Barrett’s metaplasia, dysplasia, adenocarcinoma sequence. J Gastrointest Surg. 2000;4:135-142.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Duggan C, Onstad L, Hardikar S, Blount PL, Reid BJ, Vaughan TL. Association between markers of obesity and progression from Barrett’s esophagus to esophageal adenocarcinoma. Clin Gastroenterol Hepatol. 2013;11:934-943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 106]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
69.  Somasundar P, Riggs D, Jackson B, Vona-Davis L, McFadden DW. Leptin stimulates esophageal adenocarcinoma growth by nonapoptotic mechanisms. Am J Surg. 2003;186:575-578.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Neale RE, Doecke JD, Pandeya N, Sadeghi S, Green AC, Webb PM, Whiteman DC. Does type 2 diabetes influence the risk of oesophageal adenocarcinoma? Br J Cancer. 2009;100:795-798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
71.  Dong LM, Kristal AR, Peters U, Schenk JM, Sanchez CA, Rabinovitch PS, Blount PL, Odze RD, Ayub K, Reid BJ. Dietary supplement use and risk of neoplastic progression in esophageal adenocarcinoma: a prospective study. Nutr Cancer. 2008;60:39-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 26]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
72.  Mulholland HG, Murray LJ, Anderson LA, Cantwell MM. Vitamin D, calcium and dairy intake, and risk of oesophageal adenocarcinoma and its precursor conditions. Br J Nutr. 2011;106:732-741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
73.  El-Serag HB, Aguirre TV, Davis S, Kuebeler M, Bhattacharyya A, Sampliner RE. Proton pump inhibitors are associated with reduced incidence of dysplasia in Barrett’s esophagus. Am J Gastroenterol. 2004;99:1877-1883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 242]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
74.  Kastelein F, Spaander MC, Steyerberg EW, Biermann K, Valkhoff VE, Kuipers EJ, Bruno MJ. Proton pump inhibitors reduce the risk of neoplastic progression in patients with Barrett’s esophagus. Clin Gastroenterol Hepatol. 2013;11:382-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 159]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
75.  Beales IL, Vardi I, Dearman L. Regular statin and aspirin use in patients with Barrett’s oesophagus is associated with a reduced incidence of oesophageal adenocarcinoma. Eur J Gastroenterol Hepatol. 2012;24:917-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
76.  Sadaria MR, Reppert AE, Yu JA, Meng X, Fullerton DA, Reece TB, Weyant MJ. Statin therapy attenuates growth and malignant potential of human esophageal adenocarcinoma cells. J Thorac Cardiovasc Surg. 2011;142:1152-1160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
77.  Singh S, Singh AG, Singh PP, Murad MH, Iyer PG. Statins are associated with reduced risk of esophageal cancer, particularly in patients with Barrett’s esophagus: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2013;11:620-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 138]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
78.  Sjöberg T, García Rodríguez LA, Lindblad M. Angiotensin-converting enzyme inhibitors and risk of esophageal and gastric cancer: a nested case-control study. Clin Gastroenterol Hepatol. 2007;5:1160-1166.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 39]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
79.  Alexandre L, Broughton T, Loke Y, Beales IL. Meta-analysis: risk of esophageal adenocarcinoma with medications which relax the lower esophageal sphincter. Dis Esophagus. 2012;25:535-544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
80.  Vaughan TL, Farrow DC, Hansten PD, Chow WH, Gammon MD, Risch HA, Stanford JL, Schoenberg JB, Mayne ST, Rotterdam H. Risk of esophageal and gastric adenocarcinomas in relation to use of calcium channel blockers, asthma drugs, and other medications that promote gastroesophageal reflux. Cancer Epidemiol Biomarkers Prev. 1998;7:749-756.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Lubin JH, Cook MB, Pandeya N, Vaughan TL, Abnet CC, Giffen C, Webb PM, Murray LJ, Casson AG, Risch HA. The importance of exposure rate on odds ratios by cigarette smoking and alcohol consumption for esophageal adenocarcinoma and squamous cell carcinoma in the Barrett’s Esophagus and Esophageal Adenocarcinoma Consortium. Cancer Epidemiol. 2012;36:306-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
82.  Coleman HG, Bhat S, Johnston BT, McManus D, Gavin AT, Murray LJ. Tobacco smoking increases the risk of high-grade dysplasia and cancer among patients with Barrett’s esophagus. Gastroenterology. 2012;142:233-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 84]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
83.  Hardikar S, Onstad L, Blount PL, Odze RD, Reid BJ, Vaughan TL. The role of tobacco, alcohol, and obesity in neoplastic progression to esophageal adenocarcinoma: a prospective study of Barrett’s esophagus. PLoS One. 2013;8:e52192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 73]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
84.  Cook MB, Shaheen NJ, Anderson LA, Giffen C, Chow WH, Vaughan TL, Whiteman DC, Corley DA. Cigarette smoking increases risk of Barrett’s esophagus: an analysis of the Barrett’s and Esophageal Adenocarcinoma Consortium. Gastroenterology. 2012;142:744-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 127]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
85.  Tramacere I, La Vecchia C, Negri E. Tobacco smoking and esophageal and gastric cardia adenocarcinoma: a meta-analysis. Epidemiology. 2011;22:344-349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 71]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
86.  Tramacere I, Pelucchi C, Bagnardi V, Rota M, Scotti L, Islami F, Corrao G, Boffetta P, La Vecchia C, Negri E. A meta-analysis on alcohol drinking and esophageal and gastric cardia adenocarcinoma risk. Ann Oncol. 2012;23:287-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 72]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
87.  Chen J, Zhang N, Ling Y, Wakai T, He Y, Wei L, Wang S, Akazawa K. Alcohol consumption as a risk factor for esophageal adenocarcinoma in North China. Tohoku J Exp Med. 2011;224:21-27.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Freedman ND, Murray LJ, Kamangar F, Abnet CC, Cook MB, Nyrén O, Ye W, Wu AH, Bernstein L, Brown LM. Alcohol intake and risk of oesophageal adenocarcinoma: a pooled analysis from the BEACON Consortium. Gut. 2011;60:1029-1037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 84]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
89.  Groves C, Jankowski J, Barker F, Holdstock G. A family history of Barrett’s oesophagus: another risk factor? Scand J Gastroenterol. 2005;40:1127-1128.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Munitiz V, Parrilla P, Ortiz A, Martinez-de-Haro LF, Yelamos J, Molina J. High risk of malignancy in familial Barrett’s esophagus: presentation of one family. J Clin Gastroenterol. 2008;42:806-809.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
91.  Sappati Biyyani RS, Chessler L, McCain E, Nelson K, Fahmy N, King J. Familial trends of inheritance in gastro esophageal reflux disease, Barrett’s esophagus and Barrett’s adenocarcinoma: 20 families. Dis Esophagus. 2007;20:53-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
92.  Jochem VJ, Fuerst PA, Fromkes JJ. Familial Barrett’s esophagus associated with adenocarcinoma. Gastroenterology. 1992;102:1400-1402.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Menke V, Pot RG, Moons LM, van Zoest KP, Hansen B, van Dekken H, Siersema PD, Kusters JG, Kuipers EJ. Functional single-nucleotide polymorphism of epidermal growth factor is associated with the development of Barrett’s esophagus and esophageal adenocarcinoma. J Hum Genet. 2012;57:26-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
94.  Chang EY, Morris CD, Seltman AK, O’Rourke RW, Chan BK, Hunter JG, Jobe BA. The effect of antireflux surgery on esophageal carcinogenesis in patients with barrett esophagus: a systematic review. Ann Surg. 2007;246:11-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 151]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
95.  Stein HJ, Kauer WK, Feussner H, Siewert JR. Bile reflux in benign and malignant Barrett’s esophagus: effect of medical acid suppression and nissen fundoplication. J Gastrointest Surg. 1998;2:333-341.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Löfdahl HE, Lu Y, Lagergren P, Lagergren J. Risk factors for esophageal adenocarcinoma after antireflux surgery. Ann Surg. 2013;257:579-582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]