Abstract
As the incidence and mortality of esophageal adenocarcinoma continue to increase, strategies to counter this need to be explored. Screening for Barrett’s esophagus, which is the known precursor of a large majority of adenocarcinomas, has been debated without a firm consensus. Given evidence for and against perceived benefits of screening, the multitude of challenges in the implementation of such a strategy and in the downstream management of subjects with Barrett’s esophagus who could be diagnosed by screening, support for screening has been modest. Recent advances in form of development and initial accuracy of non-invasive tools for screening, risk assessment tools and biomarker panels to risk stratify subjects with BE, have spurred renewed interest in the early detection of Barrett’s esophagus and related neoplasia, particularly with the advent of effective endoscopic therapy. In this review, we explore in depth, the potential rationale for screening for Barrett’s esophagus, recent advances which have the potential of making screening feasible and also highlight some of the challenges which will have to be overcome to develop an effective approach to improve the outcomes of subjects with esophageal adenocarcinoma.
Keywords: Screening, Barrett’s Esophagus, Esophageal adenocarcinoma, screening tools, risk stratification
Introduction
Esophageal adenocarcinoma (EAC) is the dominant histological subtype of esophageal cancer in the West. The incidence of EAC has risen by almost 6-fold over the last three and a half decades in Europe and North America1, along with a dismal 5-year survival rate of less than 20% in cases diagnosed after onset of symptoms2. Up to 50% of patients may have incurable disease at symptomatic presentation requiring palliative measures3. On the other hand, 5-year survival rates of subjects diagnosed with early asymptomatic cancers are substantially better than those with symptomatic disease4–6. The high prevalence of Barrett’s Esophagus (BE) in subjects with EAC has been reported in several studies and there is consensus that it is the precursor for the majority of EACs7–9. These statistics have provided some impetus to develop strategies for early detection and diagnosis followed by curative therapy as a possible approach to reducing mortality from this lethal cancer.
Recent reports of the feasibility and safety of novel non-endoscopic (sponge based)10 and additional data on established modified unsedated endoscopic (transnasal endoscopy)11–13 techniques have spurred renewed interest in early detection of EAC and its precursors. Recent guidelines from gastroenterology societies from the United States and the United Kingdom have provided more support to screening individuals for BE and EAC, particularly those with “multiple” risk factors, such as chronic reflux symptoms, central obesity and a family history of EAC or BE4, 15, though the number of risk factors which may spur a decision to screen and the tools to screen remain unclear. The aims of this review are to: 1. Provide a summary of the evidence regarding the rationale for and against pursuing select population screening as a possible approach to reduce mortality from EAC. 2. Detail the challenges and recent advances in terms of available tools, identification of a target population and methods for cancer risk stratification, and 3. Identify continuing challenges and areas for future research.
Rationale and Challenges for Screening
It is recognized that there are two main complementary methods to reduce cancer mortality: first, primary prevention (such as smoking cessation) and second, by early detection and treatment16. There are examples for which screening has already reduced cancer deaths, such as cervical smears to detect cervical intraepithelial neoplasia and more recently, colorectal cancer screening to detect adenomas17, 18. However, the benefits and risks have to be carefully weighed, in particular, the economical and psychological consequences of early detection, as the majority of premalignant lesions in select organs may not progress to cancer even if left untreated16.
Surveillance for BE is recommended by medical societies and indeed has been practiced for the last two decades even though cancer progression rates in BE without dysplasia are as low as 0.33% per year and only 5%–8% of esophageal adenocarcinomas are diagnosed during surveillance19. Indeed a recent case control study found no evidence of a survival benefit with surveillance endoscopy in a large population based cohort20. The overall survival of patients with non-dysplastic BE is not different from age and gender matched individuals21. In fact, the majority die from cardiovascular disease and pulmonary disease, while only 7% of BE “surveillance” patients die of EAC in the population22. On the other hand, approximately 66.5% of EACs occur within a year from the initial endoscopic diagnosis of BE9, suggesting a high rate of “prevalent” cancers in BE which are excluded from analysis when reporting cancer progression risk in epidemiological studies and may reflect the yield from a screening procedure.
Evidence from several retrospective studies has suggested that a previous diagnosis of BE, before the diagnosis of EAC is associated with early stage disease, lower rates of metastatic lymphadenopathy and improved survival23. A prior diagnosis of gastroesophageal reflux disease (GERD) and prior endoscopy are also associated with early stage EAC and better survival from EAC, respectively24. While the possibility of lead time and length time bias exists, these data suggest that early detection may reduce mortality from EAC, particularly given data from several studies showing substantially improved survival of subjects with early stage (T1a and T1b) EAC compared to those with locally advanced disease5, 25.
Hence, these data present a dilemma: a continuing rise in the incidence of a lethal cancer uncommonly diagnosed at a curable stage, in the face of surveillance programs that target a population of low risk or “benign” BE and appear to have minimal impact on overall survival.
A potential explanation maybe that current strategies are not identifying a majority of subjects with prevalent BE in the community, leading to limiting clinical attention (by surveillance) to the minority of clinically diagnosed BE in the population. This hypothesis is supported, at least in part, by data from autopsy and population based epidemiological studies estimating that only a third of patients with BE in the population are clinically detected, with the remainder remaining undiagnosed, despite the substantial increase in the use of endoscopy21. Indeed, more than 90% of patients who present with EAC do not have a previous diagnosis of BE7, an indication of the underlying challenge.
Several potential hurdles to the adoption of screening as a widespread strategy exist. These include the lack of an acceptable, accurate and cost effective tool for screening, the lack of a well characterized screening population, absence of real data on the costs of non-invasive screening, the potential downstream costs of performing surveillance as currently recommended (without consideration of an individual’s risk of progression) and lack of prospective evidence that screening will lead to improved outcomes. An additional hurdle to the expansion of screening in BE had been the lack of therapeutic options for subjects diagnosed with high grade dysplasia (HGD) or intramucosal carcinoma, other than esophagectomy which is a morbid procedure with associated mortality and substantial morbidity. With the advent of effective minimally invasive Barrett’s endotherapy techniques, such as endoscopic mucosal resection and ablation, which are cost-effective and safe alternatives to surgery, there is greater impetus to develop and study techniques which can detect precursors to EAC in an acceptable, accurate and cost effective manner26, 27.
In the next few sections, we will review some recent advances, which may partially mitigate and address some of these limitations.
Advances in tools for screening
Standard endoscopy is not ideal for use in large scale screening, given that it has to be performed in an office or hospital based endoscopy suite, with a small but significant risk of morbidity and mortality related to complications from conscious sedation and the procedure itself28. It is also associated with significant direct (sedation, monitoring, staff, recovery) and indirect (time off work and reduced productivity) costs. All these factors have limited the widespread use of standard endoscopy for screening purposes, since screening tests should ideally be simple and safe to perform, acceptable to the population and cost-effective29. As a result, several alternatives to standard endoscopy have been evaluated.
Non-endoscopic techniques
Balloon cytology sampling devices that collect esophageal samples by swallowing and the withdrawing a sizable balloon with a mesh cover have been utilized with limited success. This technique has been compared with standard endoscopy and brush cytology. Adequate samples were obtained in only 83% of patients with balloon cytology compared to 97% with brush cytology. Sensitivity of the former for HGD and EAC was 80% but only 25% for LGD30. A non-endoscopic flexible mesh balloon catheter was also assessed but adequate specimens were obtained in only 73% of patients. The sensitivity of identifying goblet cells in those patients was 87.5%31. These techniques were hence limited by the inadequacy of sampling and poor accuracy.
Over the last few years, researchers from Cambridge University have developed the Cytosponge, a novel cell collection device coupled with a biomarker10. It is a gelatin capsule attached to a string. Once swallowed, the capsule dissolves and releases a mildly abrasive spherical sponge which can be withdrawn orally, with the attached string to collect cytology specimens from the esophagus. The Cytosponge samples are then analyzed for the presence of Trefoil Factor 3 (TFF3), a biomarker of columnar epithelium, using immunohistochemistry. In a large primary care study involving 12 UK general practices, subjects with GERD symptoms were invited to undergo the Cytosponge test followed by standard endoscopy with biopsies at the hospital. 504 patients (18%) agreed to take part. The Cytosponge test was technically feasible in 99% of patients and was well tolerated. The sensitivity and specificity of the test for the detection of Barrett’s epithelium was 73.3% and 93.8% (for Barrett’s circumferential length ≥1 cm). When defining Barrett’s as a circumferential length of ≥2 cm, the sensitivity increased to 90.0% with a specificity of 93.5%10. A recent economic modeling study reported that Cytosponge screening in 50 year-old men with GERD symptoms followed by endotherapy in those with dysplasia or intramucosal carcinoma was cost effective compared to no screening and could reduce the number of cases of incident symptomatic EAC by 19%, compared with 17% for screening by standard endoscopy32.
This device is promising in terms of cost-effectiveness, applicability for community use, and acceptability to patients. Nonetheless, it is a non-endoscopic technique; therefore mucosal visualization is not possible and will still need a confirmatory endoscopy. An important aim of a BE screening program is to identify patients with dysplasia and early EAC, thus further evaluation of the Cytosponge ability to detect these two conditions is needed. Multicenter studies are currently underway to validate these results in other UK and international centers.
Imaging-based techniques
Wireless Capsule Endoscopy (WCE) is a technology which can visualize the esophagus, small bowel and colon. While initial studies were promising with respect to accuracy in detecting BE33, later studies revealed somewhat disappointing results34. Bhardwaj et al35 performed a meta-analysis of 9 studies comprising a total of 618 patients. The pooled sensitivity and specificity of WCE for the diagnosis of BE was 77% and 86%, respectively. While WCE is an attractive tool for screening because it is safe, non-invasive, acceptable, does not require sedation, and can be carried out by a single trained technician13, it is remains limited by its low sensitivity, inability to take biopsies, need for confirmatory endoscopy, and lack of cost-effectiveness compared to standard endoscopy36. It is therefore not a suitable tool for BE screening at the present time.
String capsule endoscopy is a modified technique with a string attached to the capsule device allowing for controlled movement up and down the esophagus. The device is also re-usable which reduces the cost. The sensitivity and specificity is 78.3% and 82.8%, respectively, when compared with standard endoscopy37. While this technology is less costly, its accuracy remains suboptimal for use in screening.
Unsedated ultrathin transnasal endoscopy (TNE) has been evaluated for the diagnosis of BE. A meta-analysis of five studies (439 patients) was presented recently38. All patients underwent both TNE (diameter 4–6 mm) and standard endoscopy (gold standard). The pooled sensitivity and specificity of TNE in detecting BE was 91% and 96%, respectively. The procedure was successful (95% to 100) and well tolerated in the majority of patients. The current available TNE devices on the market unfortunately require dedicated endoscopy suites with specialized equipment and decontamination facilities. This could hamper their use for widespread screening. More recently, The EndoSheath® transnasal esophagoscopy (Vision-Sciences Inc., Orangeburg, New York) has been developed. It utilizes a disposable rubber sheath technology that covers the scope obviating the need for sterilization and utilizes a more compact processing system allowing for easy portability. Initial reports on the acceptability and feasibility of using this device in a clinic based setting have been encouraging11. This device is also currently being evaluated for BE screening in the community (ClinicalTrials.gov NCT01288612). Another promising device is the EG Scan® (Intromedic Ltd., Seoul, South Korea), which incorporates a disposable probe omitting the need for decontamination equipment. It is highly compact and portable therefore can be used in the community. TNE can also be performed successfully by physician extenders reducing operator costs39. These technologies require comprehensive evaluation in the setting of screening for BE and EAC.
Over the last few years, advances in “endomicroscopic” imaging technologies have emerged. Researchers have recently developed a tethered capsule endomicroscopy device which has the opportunity to open up new possibilities for screening and diagnosis in the GI tract. It employs optical frequency domain imaging technology to provide cross-sectional and three-dimensional architectural microscopic images of the esophagus. Image information is spatially correlated with histopathology from corresponding areas enabling the comprehensive assessment of subsurface microstructures that cannot be visualized by endoscopy. The training required to conduct the procedure is minimal. It can be performed in a primary care physician’s office. It can be retrieved and disinfected, therefore reducing the cost, making it feasible for large population screening. Data on its accuracy are awaited40.
Hence in the last few years, several new and promising techniques for BE screening which are potentially acceptable, reasonably accurate, widely applicable and potentially cost effective, have emerged (figure 1), providing a possible solution to the issue of the lack of availability of a screening tool.
Figure 1.
Tools currently undergoing evaluation for use in BE screening. A, Cytosponge; B, EG scan system (disconnected); C, EG Scan system (connected); D, EndoSheath system.
Advances in identification of target population for screening
One of the major challenges to screening for BE remains the absence of a well-defined high risk population. While GERD symptoms remain the strongest risk factor for BE and EAC, raising the risk of BE and EAC by 5 and 8 fold, respectively41, 42, it should also be noted that prevalence rates of 14.9%, 25% and 45% for BE have been reported in patients who were asymptomatic for GERD43–45. Indeed a recent meta-analysis demonstrated the lack of association of symptomatic GERD with short segment BE41. Therefore a screening program that only relies on the presence of GERD as a selection criterion will likely miss a significant proportion of patients with BE. Conversely, the target population will be very large because of the high prevalence of GERD in Western populations (15–20%)46.
Numerous studies have reported additional risk factors for developing BE (table 1) namely: male gender; Caucasian race; age; central obesity, smoking and family history47. The typical median age of diagnosis is 60 years and prevalence increases with age48. Central adiposity (visceral adipose tissue area, increased waist-hip ratio, or waist/abdominal circumference) rather than BMI has also been implicated in the pathogenesis of BE49. Several studies have demonstrated a consistent association between central obesity and risk of BE (OR, 1.98; 95% CI, 1.52–2.57) independent of BMI and symptomatic GERD50. Central obesity is also strongly associated with EAC and the effects may be mediated by reflux-dependent and independent mechanisms50.
Table 1.
Studies reporting risk factors for BE
| Study author | Year & country |
Sample size | Study design | Risk factors for BE** |
|---|---|---|---|---|
| BE | ||||
|
Eloubeidi MA, et al48 |
2001, USA |
107 BE 104 GERD |
Prospective case-control study Questionnaire based |
Age>40 (p=0.008) Heartburn or acid regurgitation (p=0.03) Heartburn > once per week (p=0.007) |
| Gerson LB, et al70 | 2001, USA |
517 GERD (99 known) |
Prospective cohort 7- symptom questionnaire |
Male gender Heartburn Nocturnal pain Odynophagia. |
| Avidan B, et al71 | 2002, USA |
256 BE 229 GERD |
Prospective case-control study |
Number reflux episodes Hiatus hernia Excess alcohol and smoking |
| Chak A, et al72 | 2002, USA |
58 BE 106 controls |
Case control study |
family history (OR 12.2; 95%CI 3.3, 44.8) |
| Smith KJ, et al73 | 2005, Australia |
167 BE 261 controls |
Population based case-control study, self-completed questionnaires |
Weekly GERD (OR 29.7; 95%CI 12.2, 72.6) Smoking (OR 3.1) BMI not associated |
| Cook MB, et al74* | 2005, UK |
32 studies | Meta-analysis | Pooled Male: female ratio of 1.96:1 (95%CI 1.77, 2.17/1) |
| Corley DA, et al75 | 2007, USA |
320 BE 316 GERD 317 controls |
Case-control study |
Larger waist circumference (OR 2.24; 95%CI 1.21, 4.15); circumference >80 cm vs. <80 cm. No association with BMI |
|
Johansson J, et al76 |
2007, Sweden |
604 undergoing endoscopy 160 controls |
Prospective case-control study self- administered questionnaire |
GERD and smoking indicated a 10.7- and 3.3-fold risk, respectively, for BE (95%CI 3.5, 33.4 and 1.1, 9.9, respectively). BMI was unrelated to risk. |
|
Edelstein ZR, et al77 |
2007, USA |
193 BE 211 controls |
Prospective Case-control study |
Waist-to-hip ratio (OR 2.4; 95%CI 1.4, 3.9 all cases and 4.3; 95%CI 1.9, 9.9 for LSBE) independent of BMI. Association with BMI and smoking weaker |
|
Anderson LA, et al78 |
2007, Northern Ireland |
224 BE 227 EAC 260 controls |
Population based case-control study |
GERD (OR 12.0; 95%CI 7.64, 18.7). BMI and smoking not associated with BE |
| Cook MB, et al79* | 2008, UK |
9 studies (BE/GERD controls). 3 studies (BE/population controls) |
Meta-analysis |
BMI 9 studies pooled OR 0.99 per kg/m2 (95%CI 0.97, 1.01 and 3 studies OR 1.02 per kg/m2 (95%CI 1.01, 1.04 |
|
Edelstein ZR, et al80 |
2009, USA |
197 BE 418 controls |
Case control study |
Older age (OR per decade for SIM=1.3; 95%C 1.1, 1.5; LSBE =1.5; 95%CI 1.2, 1.9) Male gender (OR SIM=1.5; 95%CI 1.1, 2.2; LSBE= 3.9; 95%CI=1.9, 8.1 waist-to-hip ratio (male high= ≥0.9, female high= ≥0.8) SIM (OR 1.3; 95%CI 0.9, 2.1), LSBE (OR 4.1; 95%CI 1.5, 11.4) Smoking (ever) SIM (OR 1.8; 95%CI 1.2–2.6), LSBE (OR 2.6; 95%C I 1.3, 4.9) |
| Kubo A, et al81 | 2009, USA |
320 BE 316 GERD 317 controls |
Case-control study interview questionnaires |
Alcohol not associated. Wine consumption & education status inversely associated |
|
Anderson LA, et al82 |
2009, Northern Ireland |
230 GERD 224 BE 227 EAC 260 controls |
Case-control study using structured Interview |
No association between total alcohol consumption and BE. Wine consumption may reduce risk of BE |
| Taylor JB, et al41* | 2010, USA |
26 studies | Meta-analysis |
GERD (OR in all studies 2.90; 95% CI 1.86, 4.54) Strong association with LSBE (OR 4.92; 95%CI 2.01, 12.0), no association with SSBE |
| Orloff M, et al53 | 2011, USA |
Validation cohort 176 BE/EAC 200 controls |
Model-free linkage analyses |
MSR1, ASCC1, and CTHRC1 genes associated with BE/EAC (P< .001) |
| Su Z, et al52 | 2012, UK |
Discovery cohort 1,852 BE 5,172 controls replication cohort 5,986 cases 12,825 controls |
genome-wide association study (GWAS) |
Chromosome 6p21 locus (OR 1.21; 95%CI 1.13, 1.28) Chromosome 16q24 (OR 1.14; 95% CI 1.10, 1.19) |
| Cook MB, et al54 | 2012, USA |
1059 BE 1332 GERD 1143 controls |
Analyzed data from 5 case-control studies81–84 |
Ever smoked (OR 1.67 and 1.61 compared to controls and GERD controls respectively. Association strengthened with increased exposure until 20 pack-years) |
|
Balasubramanian G, et al47 |
20 12, USA |
1058 patients with GERD |
Prospective cohort using validated GERD questionnaire |
Heartburn >5 years (OR 1.50; 95% CI 1.07, 2.09), Caucasian race (OR 2.40; 95% CI 1.42, 4.03), hiatus hernia (OR: 2.07; 95% CI 1.50, 2.87) |
| Leggett CL, et al85 | 2013, USA |
36 BE 78 OSA 74 BE+OSA 74 no BE or OSA |
Case-control study |
OSA (OR 1.8; 95%CI 1.1, 3.2). Independent of age, sex, BMI, GERD, and smoking history. Risk higher with more severe OSA |
| Thrift AP, et al86 | 2013, Australia |
258 BE 453 primary care controls 1145 endoscopy controls |
Case-control study |
Smoking: no association (status, intensity, age at initiation, duration, pack-years and cessation). Alcohol: not associated, moderate intake inversely associated. |
| Garcia JM, et al87 | 2013, USA |
141 BE 139 controls |
Prospective case-control |
OR 2.62 (95%CI 1.0, 6.8), 5.18 (95%CI 1.7, 15.7), and 8.02 (95% CI 2.79, 23.07) for highest vs. lowest quintile of levels of IL-12p70, IL-8, and leptin, respectively, |
| Singh S, et al50 | 2013, USA |
40 studies | Meta-analysis |
central adiposity had a higher risk of BE (17 studies; OR 1.98; 95% CI 1.52, 2.57) |
where available, meta-analysis of established risk factors (GERD, male gender, obesity) presented instead of individual studies. OR, odds ratio; CI, confidence interval; BMI, body mass index; LSBE, long segment Barrett’s esophagus; SIM, small intestinal metaplasia; OSA, obstructive sleep apnea.
Family history of BE or EAC is also associated with an increased risk of BE and as many as 28% of first degree relatives of patients with EAC or Barrett’s HGD have BE in some reports51. A recent large genome-wide association study (GWAS) study identified genetic variants associated with BE risk at two loci; one at chromosome 6p21 (OR 1.21, 95%CI 1.13–1.28), and another on chromosome 16q24 (OR 1.14, 95%CI 1.10–1.19). The closest protein-coding gene to this chromosome was FOXF1, which is a transcription factor involved in esophageal development and structure52. ASCC1, MSR1, and CTHRC1 genes germline mutations have also been associated with BE and EAC53. Patients with BE are significantly more likely to have ever smoked compared to population-based controls (OR: 1.67; 95% CI: 1.04–2.67) or GERD controls (OR: 1.61; 95% CI: 1.33–1.96). This association was strengthened with increased exposure to smoking until approximately 20 pack-years, when it began to plateau. Smoking also had synergistic effects with heartburn or regurgitation54.
Rubenstein et al55 performed the largest study so far by extracting data from a USA national standardized endoscopic database established by the Clinical Outcomes Research Initiative (CORI). 9% (4,339 out of 48,476) of GERD patients had suspected BE on endoscopy and this was confirmed histologically in 2.8% of patients who had both endoscopy and histology reports available (histological sub-cohort n=25,337). Prevalence of BE in white non-Hispanic male GERD subjects increased sharply from 3.3% in the 4th decade, to 6.3% in the 5th decade, to 9.3% in the 6th decade of life, and then reached a plateau afterwards. Remarkably, white women with GERD symptoms were no more likely to have BE than white men without GERD. However, this may have been biased by the differential reporting of GERD symptoms in the indication for the endoscopies, or differential selection for endoscopy between men and women. Results from a recent modeling study suggest that screening for EAC should be considered in white men age ≥60 with weekly GERD and those aged ≥55 who have daily GERD. Authors estimated the symptom-, age-, and sex-specific incidences of EAC, and compared these figures to other cancers for which screening is endorsed, namely, colorectal and breast cancer (figure 2a and 2b)56. The projected incidence of EAC in women with GERD was estimated to be comparable to that of breast cancer in men. Incidence in women with GERD remained lower than a threshold at which screening for colon cancer in women is endorsed and the utility of screening for BE in women was questioned.
Figure 2.
a: Estimated incidence of EAC in populations with weekly GERD symptoms. Incidences of colorectal cancer (CRC) and breast cancer (Bra) are displayed for context. The benchmark incidence of cancer for endorsed screening is demonstrated by the green horizontal line. Thick lines represent EAC, thin lines represent CRC, and dotted lines represent Bra. Thin dashed lines represent the upper and lower confidence limits of the incidence of EAC in men with weekly GERD, and the upper confidence limit of EAC in women with weekly GERD. The lower confidence limit for women with GERD overlap the point estimate incidence of EAC in women and are not visible on the figure. (Reproduced with permission from Rubenstein JH, et al56. Copyright Nature Publishing Group).
b: Estimated incidence of EAC in populations with daily GERD symptoms. (Reproduced with permission from Rubenstein JH, et al56. Copyright Nature Publishing Group).
Most recently, BE risk prediction models have been developed. They incorporate multiple established risk factors and have the potential for use an inexpensive preselection tool for screening. So far, three models with different set of predictors have been published57–59. Thrift et al57, initially evaluated a large number of putative risk factors in a case-control study with external validation. The final risk model included age, sex, smoking status, BMI, highest level of education and frequency of use of acid suppressant medications. The area under the receiver operating characteristic curve (AUROC) was 0.70 and 0.61 in the derivation and validation groups, respectively. Another modestly accurate model is the online Michigan Barrett's Esophagus pREdiction Tool (M-BERET) (http://mberet.umms.med.umich.edu/). This model is based on a population of colonoscopy screenees who consented to a study endoscopy and included four variables (weekly GERD, age, waist-to-hip ratio and pack-years of cigarette smoking). It performed better (AUROC = 0.72) than a model with reflux parameters alone (AUROC = 0.61, p<0.001)58.
Thrift et al59, reported a significant association between a multi-biomarker risk score model (based on serum levels of interleukin (IL) 12p70, IL6, IL8, IL10 and leptin) and risk of BE. Subjects with a score of 3 or more had a greater than 10-fold increase risk of BE compared to those with a score of 0 (OR 11.9; 95%CI 4.06, 34.9). The combination of the multi-biomarker risk score model with a demographic and clinical features model (based on GERD frequency and duration, age, sex, race, waist-to-hip ratio, and H. Pylori status) was significantly more accurate at predicting the presence of BE compared to a combination of the risk score with a model based on GERD frequency and duration alone (AUROC 0.85 vs. 0.74; p = 0.01). Data from this case-control study was based on a predominantly white male Veterans population. This prediction model represents a significant improvement in accuracy compared to previous methods. Despite the progress made in attempting to define the target population for screening, several challenges to screening remain.
Methods and recent developments in BE cancer risk stratification
In order to render any future population screening and surveillance program feasible and cost effective, a cancer risk prediction algorithm that incorporates demographic, clinical, histological and molecular biomarkers would be vital. This will help to riskstratify patients with BE and focus surveillance and therapy on the high risk group. Several independent risk factors for increased risk of EAC in BE subjects have been identified.
Demographic and clinical risk factors
Men with BE are at increased risk for development of EAC compared to women. The overall risk is around 0.28% per year in men and 0.13% per year in women60. Data on the association of male gender and risk of progression are somewhat scarce and inconsistent; therefore recommendations for surveillance in both men and women are currently the same. Increasing age is another risk factor reported in some studies, but these findings were not replicated in larger cohort studies61. Active tobacco smoking has been associated with an increased risk of progression (hazard ratio = 2.03; 95% confidence interval, 1.29–3.17) across all classes of smoking intensity in some, but not all studies62, while no link has been found with alcohol consumption. Increased waist to hip ratio, but not BMI, was associated with risk of aneuploidy, 9p loss of heterozygosity (LOH) and 17p LOH, suggesting that visceral adipose tissue may be a risk factor for malignant progression61.
There is substantial evidence that the length of BE segment is a significant risk factor although in few studies the association was not statistically significant14. Besides segment length, the presence of mucosal nodularity, ulceration and strictures have been associated with risk of EAC; however, this may indicate prevalent neoplasia rather than risk of future cancer63. Risk of progression to HGD and cancer are higher in the presence of hiatus hernia and esophagitis, likely as a result of esophageal inflammation driving this process through increased risk of mutations64. Ideally, a future risk prediction model should take into account all these factors; however as of yet, a definitive model has not been developed and validated, hence at present time, segment length appears to be the only factor which can be used to suggest surveillance intervals in patients with NDBE14.
Histological and molecular risk factors
While the grade of dysplasia remains the most widely used risk assessment tool14, poor interobserver agreement among pathologists and sampling error remain major limitations in the use of dysplasia as a risk stratification tool65. Hence attempts have been made to develop biomarkers as complementary methods to predict risk of EAC in BE.
The use of biomarkers in combination with clinical markers is appealing. This was evaluated in one study by analyzing data from an age-, sex-, and year of BE diagnosis-matched case-control cohort. A panel of established (abnormal DNA content, p53 and cyclin A expression) and new biomarkers (levels of sialyl Lewisa, Lewisx, and Aspergillus oryzae lectin and binding of wheat germ agglutinin) was assessed. A combination of low grade dysplasia, abnormal DNA ploidy, and Aspergillus oryzae lectin was most accurate at predicting histologic progression. For each additional factor of these three, BE patients with LGD had an approximately 4-fold increased odds of progressing to develop EAC or HGD (OR, 3.74; 95% CI, 2.43 – 5.79), whereas the risk increased by a 3-fold (OR, 3.31; 95% CI, 1.81– 6.05) for each additional factor in patients with no dysplasia66. Prospective randomized controlled trial evidence needs to be available before these markers can be recommended for use in clinical practice.
Immunohistochemistry for nuclear p53, a tumor suppressor gene, is perhaps currently the most promising potential biomarkers for risk stratification in clinical practice. It is easily applicable; reproducible; and serves as a good predictor of malignant progression65. In a prospective cohort study of 720 patients with BE, p53 overexpression was associated with an increased risk of neoplastic progression (HGD or EAC) independent of age, gender, BE segment length and esophagitis (adjusted relative risk 5.6; 95%CI 3.1, 10.3), and the risk was even higher with loss of p53 expression (adjusted relative risk 14.0; 95%CI 5.3, 37.2). Moreover, the positive predictive value for neoplastic progression increased from 15% with LGD to 33% with LGD and aberrant p53 expression67. While the presence of dysplasia confirmed by two expert histopathologists remains the best tissue biomarker available for assessment of cancer risk in patients with BE currently14, the addition of biomarkers such as p53 to enhance the prediction of progression appears to be on the horizon. This has been recently endorsed by some gastroenterology guidelines, as a complement to histology14.
Thrift et al68 recently developed a logistic regression model using epidemiological data from 364 patients with incident EAC and 1580 population controls. The final risk model included highest level of education, BMI, smoking status, frequency of GERD and/or use of acid-suppressant medications, and frequency of non-steroidal anti-inflammatory drugs use. The absolute risk of developing EAC was estimated with an AUROC of 0.75 indicating good discrimination. External validation of this model and other biomarker panels may allow more targeted surveillance and therapy in the future.
Continuing challenges to screening and future directions
Despite these advances, several challenges to widespread BE screening remain. Non-endoscopic methods for BE remain to be validated in large populations in terms of acceptability, accuracy, safety and cost effectiveness. Despite encouraging data on the accuracy and acceptability of TNE11, 13, acceptance by physicians remains low69. Novel non-invasive markers identifiable in blood or stool would be ideal for widespread application, but are currently not available.
BE risk prediction models with adequate sensitivity can significantly reduce the number of “missed” patients in a screening program and be used as an inexpensive pre-selection tool for screening. While encouraging, the current accuracy of BE prediction models likely is insufficient for widespread clinical use (AUROC 0.6–0.85). The MBERET and other scores require external validation in unselected populations, testing in females, and likely addition of other circulating biomarkers. Iterative refinements of such models combined with a simple diagnostic test may potentially make screening for BE more cost-effective. As a result, it is likely that the use of prediction models based on multiple risk factors, will emerge as the means of identifying at risk populations for EAC. Limiting screening to higher risk subgroups (table 2) such as men with chronic reflux (daily or weekly) reflux symptoms as advocated by some studies may have to be considered.
Table 2.
| Demographic profile to be considered for screening: |
|---|
|
Any screening program is likely to result in an increased workload and costs to the healthcare system which are challenging to justify. Management of screening-diagnosed patients with NDBE remains unclear, particularly with the majority not likely to progress to cancer. In these subjects performing routine endoscopic surveillance may potentially expose patients to unnecessary harm and lead to escalating costs without substantial benefit. Therefore, methods to predict individuals at increased risk of EAC merit serious and careful consideration. Further validation, refinement and cost effectiveness of models of predictors of progression, likely combining clinical and biomarker variables, will be crucial in allowing the identification of low and high risk subsets. Subjects with a low risk of progression could likely be discharged from surveillance while those at a higher risk could be placed in intensive surveillance with advanced imaging or offered ablation to decrease the risk of developing EAC. A conceptual flow diagram outlining an approach to decrease the incidence of and mortality from EAC is presented in Figure 3. This represents a future vision of how this process could potentially be effective in reducing EAC incidence and mortality..
Figure 3. Proposed future approach to reducing mortality from EAC by utilizing screening and targeted surveillance coupled with endotherapy.
BE, Barrett’s Esophagus; EAC, esophageal adenocarcinoma; GERD, gastroesophageal reflux disease; HGD, high grade dysplasia; LGD, low grade dysplasia; LSBE, long segment Barrett’s Esopahgus; TNE, transnasal endoscopy
In summary, while significant progress had been made in the early detection and treatment of esophageal Neoplasia, several challenges to the widespread application to screening remain. Nevertheless, this area is ripe for research, particularly focusing on the comprehensive evaluation of emerging minimally-invasive and novel screening tools as well as deriving and validating BE and EAC risk prediction models.
Acknowledgments
Grant Support:
Funded in part by The Mayo Foundation and NIDDK (grant RC4DK090413). Dr. S. S. Sami is funded by an Olypmpus-Core National Endoscopy Fellowship grant, Core charity, United Kingdom.
Abbreviations
- AUROC
receiver operating characteristic curve
- BE
Barrett’s Esophagus
- BMI
body mass index
- Bra
breast cancer
- CI
confidence interval
- CRC
colorectal cancer
- EAC
esophageal adenocarcinoma
- GERD
gastroesophageal reflux disease
- GWAS
genome-wide association study
- HGD
high grade dysplasia
- LGD
low grade dysplasia
- LOH
loss of heterozygosity
- LSBE
long segment Barrett’s Esophagus
- M-BERET
Michigan Barrett’s Esophagus pREdiction Tool
- NDBE
nondysplastic Barrett’s Esophagus
- OSA
obstructive sleep apnea
- OR
odds ratio
- SIM
small intestinal metaplasia
- TFF3
Trefoil Factor 3
- TNE
transnasal endoscopy
- WCE
wireless capsule endoscopy
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosures:
Dr. Ragunath received research grant funding from Intromedic Ltd (Seoul, South Korea) and Olympus Keymed (United Kingdom).
Author Contributions:
Prasad G. Iyer is the guarantor of this article, contributed to the design of the study and critical review of the subsequent drafts of the manuscript.
Sarmed S. Sami contributed to the design, literature search, data collection, and written the first draft of manuscript.
Krish Ragunath contributed to the design, and critically reviewed the manuscript. All authors approved the final version of the manuscript.
References
- 1.Pohl H, Sirovich B, Welch HG. Esophageal adenocarcinoma incidence: are we reaching the peak? Cancer Epidemiology Biomarkers and Prevention. 2010;19:1468–1470. doi: 10.1158/1055-9965.EPI-10-0012. [DOI] [PubMed] [Google Scholar]
- 2.Pohl H, Welch HG. The role of overdiagnosis and reclassification in the marked increase of esophageal adenocarcinoma incidence. J Natl Cancer Inst. 2005;97:142–146. doi: 10.1093/jnci/dji024. [DOI] [PubMed] [Google Scholar]
- 3.Auvinen MI, Sihvo EI, Ruohtula T, et al. Incipient angiogenesis in Barrett's epithelium and lymphangiogenesis in Barrett's adenocarcinoma. J Clin Oncol. 2002;20:2971–2979. doi: 10.1200/JCO.2002.09.011. [DOI] [PubMed] [Google Scholar]
- 4.Prasad GA, Wu TT, Wigle DA, et al. Endoscopic and surgical treatment of mucosal (T1a) esophageal adenocarcinoma in Barrett's esophagus. Gastroenterology. 2009;137:815–823. doi: 10.1053/j.gastro.2009.05.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Wani S, Drahos J, Cook MB, et al. Comparison of endoscopic therapies and surgical resection in patients with early esophageal cancer: a population-based study. Gastrointest Endosc. 2014;79:224 e1–232 e1. doi: 10.1016/j.gie.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pech O, May A, Manner H, et al. Long-term Efficacy and Safety of Endoscopic Resection for Patients With Mucosal Adenocarcinoma of the Esophagus. Gastroenterology. 2013 Nov 20; doi: 10.1053/j.gastro.2013.11.006. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 7.Dulai GS, Guha S, Kahn KL, et al. Preoperative prevalence of Barrett's esophagus in esophageal adenocarcinoma: A systematic review. Gastroenterology. 2002;122:26–33. doi: 10.1053/gast.2002.30297. [DOI] [PubMed] [Google Scholar]
- 8.Chak A, Faulx A, Eng C, et al. Gastroesophageal reflux symptoms in patients with adenocarcinoma of the esophagus or cardia. Cancer. 2006;107:2160–2166. doi: 10.1002/cncr.22245. [DOI] [PubMed] [Google Scholar]
- 9.Hvid-Jensen F, Pedersen L, Drewes AM, et al. Incidence of adenocarcinoma among patients with Barrett's esophagus. N Engl J Med. 2011;365:1375–1383. doi: 10.1056/NEJMoa1103042. [DOI] [PubMed] [Google Scholar]
- 10.Kadri SR, Lao-Sirieix P, O'Donovan M, et al. Acceptability and accuracy of a non-endoscopic screening test for Barrett's oesophagus in primary care: cohort study. BMJ. 2010;341:c4372. doi: 10.1136/bmj.c4372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Peery AF, Hoppo T, Garman KS, et al. Feasibility, safety, acceptability, and yield of office-based, screening transnasal esophagoscopy (with video) Gastrointestinal Endoscopy. 2012;75:945–953. doi: 10.1016/j.gie.2012.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Shariff MK, Bird-Lieberman EL, O'Donovan M, et al. Randomized crossover study comparing efficacy of transnasal endoscopy with that of standard endoscopy to detect Barrett's esophagus. Gastrointestinal Endoscopy. 2012;75:954–961. doi: 10.1016/j.gie.2012.01.029. [DOI] [PubMed] [Google Scholar]
- 13.Chang JY, Talley NJ, Locke GR, 3rd, et al. Population screening for barrett esophagus: a prospective randomized pilot study. Mayo Clinic Proceedings. 2011;86:1174–1180. doi: 10.4065/mcp.2011.0396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Fitzgerald RC, di Pietro M, Ragunath K, et al. British Society of Gastroenterology guidelines on the diagnosis and management of Barrett's oesophagus. Gut. 2014;63:7–42. doi: 10.1136/gutjnl-2013-305372. [DOI] [PubMed] [Google Scholar]
- 15.Spechler SJ, Sharma P, Souza RF, et al. American gastroenterological association technical review on the management of Barrett's esophagus. Gastroenterology. 2011;140:e18–e52. doi: 10.1053/j.gastro.2011.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lao-Sirieix P, Fitzgerald RC. Screening for oesophageal cancer. Nature Reviews Clinical Oncology. 2012;9:278–287. doi: 10.1038/nrclinonc.2012.35. [DOI] [PubMed] [Google Scholar]
- 17.Machii R, Saito H. Time trends in cervical cancer screening rates in the OECD countries. Jpn J Clin Oncol. 2011;41:731–732. doi: 10.1093/jjco/hyr058. [DOI] [PubMed] [Google Scholar]
- 18.Benson VS, Patnick J, Davies AK, et al. Colorectal cancer screening: a comparison of 35 initiatives in 17 countries. Int J Cancer. 2008;122:1357–1367. doi: 10.1002/ijc.23273. [DOI] [PubMed] [Google Scholar]
- 19.Desai TK, Krishnan K, Samala N, et al. The incidence of oesophageal adenocarcinoma in non-dysplastic Barrett's oesophagus: a meta-analysis. Gut. 2012;61:970–976. doi: 10.1136/gutjnl-2011-300730. [DOI] [PubMed] [Google Scholar]
- 20.Corley DA, Mehtani K, Quesenberry C, et al. Impact of Endoscopic Surveillance on Mortality From Barrett's Esophagus–Associated Esophageal Adenocarcinomas. Gastroenterology. 2013;145:312–319. doi: 10.1053/j.gastro.2013.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Jung KW, Talley NJ, Romero Y, et al. Epidemiology and natural history of intestinal metaplasia of the gastroesophageal junction and Barrett's esophagus: a population-based study. American Journal of Gastroenterology. 2011;106:1447–1455. doi: 10.1038/ajg.2011.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sikkema M, de Jonge PJ, Steyerberg EW, et al. Risk of esophageal adenocarcinoma and mortality in patients with Barrett's esophagus: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2010;8:235–244. doi: 10.1016/j.cgh.2009.10.010. [DOI] [PubMed] [Google Scholar]
- 23.Cooper GS, Yuan Z, Chak A. Association of prediagnosis endoscopy with stage and survival in adenocarcinoma of the esophagus and gastric cardia. Cancer. 2002;95:32–38. doi: 10.1002/cncr.10646. [DOI] [PubMed] [Google Scholar]
- 24.Cooper GS, Kou TD, 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. doi: 10.1038/ajg.2009.159. [DOI] [PubMed] [Google Scholar]
- 25.Das A, Singh V, Fleischer DE, et al. A Comparison of Endoscopic Treatment and Surgery in Early Esophageal Cancer: An Analysis of Surveillance Epidemiology and End Results Data. Am J Gastroenterol. 2008;103:1340–1345. doi: 10.1111/j.1572-0241.2008.01889.x. [DOI] [PubMed] [Google Scholar]
- 26.Bennett C, Vakil N, Bergman J, et al. Consensus statements for management of Barrett's dysplasia and early-stage esophageal adenocarcinoma, based on a Delphi process. Gastroenterology. 2012;143:336–346. doi: 10.1053/j.gastro.2012.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Inadomi JM, Somsouk M, Madanick RD, et al. A cost-utility analysis of ablative therapy for Barrett's esophagus. Gastroenterology. 2009;136:2101–2114. doi: 10.1053/j.gastro.2009.02.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bell GD. Review article: Premedication and intravenous sedation for upper gastrointestinal endoscopy. Aliment Pharmacol Ther. 1990;4:103–122. doi: 10.1111/j.1365-2036.1990.tb00455.x. [DOI] [PubMed] [Google Scholar]
- 29.UK National Screening Commitee, Criteria for appraising the viability, effectiveness and appropriateness of a screening programme. 2009 http://wwwscreeningnhsuk/criteria. [Google Scholar]
- 30.Falk GW, Chittajallu R, Goldblum JR, et al. Surveillance of patients with Barrett's esophagus for dysplasia and cancer with balloon cytology. Gastroenterology. 1997;112:1787–1797. doi: 10.1053/gast.1997.v112.pm9178668. [DOI] [PubMed] [Google Scholar]
- 31.Rader AE, Faigel DO, Ditomasso J, et al. Cytological screening for Barrett's esophagus using a prototype flexible mesh catheter. Digestive Diseases and Sciences. 2001;46:2681–2686. doi: 10.1023/a:1012771328187. [DOI] [PubMed] [Google Scholar]
- 32.Benaglia T, Sharples LD, Fitzgerald RC, et al. Health benefits and cost effectiveness of endoscopic and nonendoscopic cytosponge screening for Barrett's esophagus. Gastroenterology. 2013;144:62–73. doi: 10.1053/j.gastro.2012.09.060. [DOI] [PubMed] [Google Scholar]
- 33.Eliakim R, Sharma VK, Yassin K, et al. A prospective study of the diagnostic accuracy of PillCam ESO esophageal capsule endoscopy versus conventional upper endoscopy in patients with chronic gastroesophageal reflux diseases. Journal of Clinical Gastroenterology. 2005;39:572–578. doi: 10.1097/01.mcg.0000170764.29202.24. [DOI] [PubMed] [Google Scholar]
- 34.Sharma P, Wani S, Rastogi A, et al. The diagnostic accuracy of esophageal capsule endoscopy in patients with gastroesophageal reflux disease and Barrett's esophagus: a blinded, prospective study. American Journal of Gastroenterology. 2008;103:525–532. doi: 10.1111/j.1572-0241.2007.01233.x. [DOI] [PubMed] [Google Scholar]
- 35.Bhardwaj A, Hollenbeak CS, Pooran N, et al. A meta-analysis of the diagnostic accuracy of esophageal capsule endoscopy for Barrett's esophagus in patients with gastroesophageal reflux disease. American Journal of Gastroenterology. 2009;104:1533–1539. doi: 10.1038/ajg.2009.86. [DOI] [PubMed] [Google Scholar]
- 36.Rubenstein JH, Inadomi JM, Brill JV, et al. Cost utility of screening for Barrett's esophagus with esophageal capsule endoscopy versus conventional upper endoscopy. Clinical Gastroenterology & Hepatology. 2007;5:312–318. doi: 10.1016/j.cgh.2006.12.008. [DOI] [PubMed] [Google Scholar]
- 37.Ramirez FC, Akins R, Shaukat M. Screening of Barrett's esophagus with string-capsule endoscopy: a prospective blinded study of 100 consecutive patients using histology as the criterion standard. Gastrointestinal Endoscopy. 2008;68:25–31. doi: 10.1016/j.gie.2007.10.040. [DOI] [PubMed] [Google Scholar]
- 38.Sami SS, Subramanian V, Ortiz-Fernández-Sordó J, et al. The utility of ultrathin endoscopy as a diagnostic tool for barrett's oesophagus (BO). Systematic review and meta-analysis United European Gastroenterology Week. Volume UEG13-ABS-2547. Berlin. 2013 [Google Scholar]
- 39.Alashkar B, Faulx AL, Hepner A, et al. Development of a Program to train Physician Extenders to Perform Transnasal Esophagoscopy and Screen for Barrett's Esophagus. Clin Gastroenterol Hepatol. 2013 Oct 23; doi: 10.1016/j.cgh.2013.10.014. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gora MJ, Sauk JS, Carruth RW, et al. Tethered capsule endomicroscopy enables less invasive imaging of gastrointestinal tract microstructure. Nat Med. 2013;19:238–240. doi: 10.1038/nm.3052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Taylor JB, Rubenstein JH. Meta-analyses of the effect of symptoms of gastroesophageal reflux on the risk of Barrett's esophagus. The American journal of gastroenterology. 2010;105:1729–1737. doi: 10.1038/ajg.2010.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lagergren J, Bergstrom R, Lindgren A, et al. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N Engl J Med. 1999;340:825–831. doi: 10.1056/NEJM199903183401101. [DOI] [PubMed] [Google Scholar]
- 43.Gerson LB, Shetler K, Triadafilopoulos G. Prevalence of Barrett's esophagus in asymptomatic individuals. Gastroenterology. 2002;123:461–467. doi: 10.1053/gast.2002.34748. [DOI] [PubMed] [Google Scholar]
- 44.Ward EM, Wolfsen HC, Achem SR, et al. Barrett's esophagus is common in older men and women undergoing screening colonoscopy regardless of reflux symptoms. American Journal of Gastroenterology. 2006;101:12–17. doi: 10.1111/j.1572-0241.2006.00379.x. [DOI] [PubMed] [Google Scholar]
- 45.Zagari RM, Fuccio L, Wallander MA, et al. Gastro-oesophageal reflux symptoms, oesophagitis and barrett's oesophagus in the general population: The Loiano-Monghidoro study. Gut. 2008;57:1354–1359. doi: 10.1136/gut.2007.145177. [DOI] [PubMed] [Google Scholar]
- 46.Dent J, El-Serag HB, Wallander MA, et al. Epidemiology of gastro-oesophageal reflux disease: a systematic review. Gut. 2005;54:710–717. doi: 10.1136/gut.2004.051821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Balasubramanian G, Singh M, Gupta N, et al. Prevalence and predictors of columnar lined esophagus in gastroesophageal reflux disease (GERD) patients undergoing upper endoscopy. Am J Gastroenterol. 2012;107:1655–1661. doi: 10.1038/ajg.2012.299. [DOI] [PubMed] [Google Scholar]
- 48.Eloubeidi MA, Provenzale D. Clinical and demographic predictors of Barrett's esophagus among patients with gastroesophageal reflux disease: a multivariable analysis in veterans. Journal of Clinical Gastroenterology. 2001;33:306–309. doi: 10.1097/00004836-200110000-00010. [DOI] [PubMed] [Google Scholar]
- 49.Wong A, Fitzgerald RC. Epidemiologic risk factors for Barrett's esophagus and associated adenocarcinoma. Clin Gastroenterol Hepatol. 2005;3:1–10. doi: 10.1016/s1542-3565(04)00602-0. [DOI] [PubMed] [Google Scholar]
- 50.Singh S, Sharma AN, Murad MH, et al. Central Adiposity Is Associated With Increased Risk of Esophageal Inflammation, Metaplasia, and Adenocarcinoma: A Systematic Review and Metaanalysis. Clin Gastroenterol Hepatol. 2013;11:1399–1412. doi: 10.1016/j.cgh.2013.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Juhasz A, Mittal SK, Lee TH, et al. Prevalence of Barrett esophagus in first-degree relatives of patients with esophageal adenocarcinoma. J Clin Gastroenterol. 2011;45:867–871. doi: 10.1097/MCG.0b013e31821f44a8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Su Z, Gay LJ, Strange A, et al. Common variants at the MHC locus and at chromosome 16q24.1 predispose to Barrett's esophagus. Nat Genet. 2012;44:1131–1136. doi: 10.1038/ng.2408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Orloff M, Peterson C, He X, et al. Germline mutations in msr1, ascc1, and cthrc1 in patients with barrett esophagus and esophageal adenocarcinoma. JAMA. 2011;306:410–419. doi: 10.1001/jama.2011.1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Cook MB, Shaheen NJ, Anderson LA, et al. Cigarette smoking increases risk of Barrett's esophagus: an analysis of the Barrett's and Esophageal Adenocarcinoma Consortium. Gastroenterology. 2012;142:744–753. doi: 10.1053/j.gastro.2011.12.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Rubenstein JH, Mattek N, Eisen G. Age- and sex-specific yield of Barrett's esophagus by endoscopy indication. Gastrointestinal Endoscopy. 2010;71:21–27. doi: 10.1016/j.gie.2009.06.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Rubenstein JH, Scheiman JM, Sadeghi S, et al. Esophageal adenocarcinoma incidence in individuals with gastroesophageal reflux: synthesis and estimates from population studies. Am J Gastroenterol. 2011;106:254–260. doi: 10.1038/ajg.2010.470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Thrift AP, Kendall BJ, Pandeya N, et al. A Clinical Risk Prediction Model for Barrett Esophagus. Cancer Prevention Research. 2012;5:1115–1123. doi: 10.1158/1940-6207.CAPR-12-0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Rubenstein JH, Morgenstern H, Appelman H, et al. Prediction of Barrett's esophagus among men. Am J Gastroenterol. 2013;108:353–362. doi: 10.1038/ajg.2012.446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Thrift AP, Garcia JM, El-Serag HB. A Multibiomarker Risk Score Helps Predict Risk for Barrett's Esophagus. Clin Gastroenterol Hepatol. 2013 Dec 19; doi: 10.1016/j.cgh.2013.12.014. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bhat S, Coleman HG, Yousef F, et al. Risk of malignant progression in Barrett's esophagus patients: results from a large population-based study. J Natl Cancer Inst. 2011;103:1049–1057. doi: 10.1093/jnci/djr203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.de Jonge PJ, van Blankenstein M, Grady WM, et al. Barrett's oesophagus: epidemiology, cancer risk and implications for management. Gut. 2014;63:191–202. doi: 10.1136/gutjnl-2013-305490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Coleman HG, Bhat S, Johnston BT, et al. Tobacco smoking increases the risk of high-grade dysplasia and cancer among patients with Barrett's esophagus. Gastroenterology. 2012;142:233–240. doi: 10.1053/j.gastro.2011.10.034. [DOI] [PubMed] [Google Scholar]
- 63.Switzer-Taylor V, Schlup M, Lubcke R, et al. Barrett's esophagus: a retrospective analysis of 13 years surveillance. J Gastroenterol Hepatol. 2008;23:1362–1367. doi: 10.1111/j.1440-1746.2008.05311.x. [DOI] [PubMed] [Google Scholar]
- 64.Moons LM, Kusters JG, Bultman E, et al. Barrett's oesophagus is characterized by a predominantly humoral inflammatory response. J Pathol. 2005;207:269–276. doi: 10.1002/path.1847. [DOI] [PubMed] [Google Scholar]
- 65.Kaye PV, Haider SA, Ilyas M, et al. Barrett's dysplasia and the Vienna classification: reproducibility, prediction of progression and impact of consensus reporting and p53 immunohistochemistry. Histopathology. 2009;54:699–712. doi: 10.1111/j.1365-2559.2009.03288.x. [DOI] [PubMed] [Google Scholar]
- 66.Bird-Lieberman EL, Dunn JM, Coleman HG, et al. Population-based study reveals new risk-stratification biomarker panel for Barrett's esophagus. Gastroenterology. 2012;143:927–935. doi: 10.1053/j.gastro.2012.06.041. [DOI] [PubMed] [Google Scholar]
- 67.Kastelein F, Biermann K, Steyerberg EW, et al. Aberrant p53 protein expression is associated with an increased risk of neoplastic progression in patients with Barrett's oesophagus. Gut. 2013;62:1676–1683. doi: 10.1136/gutjnl-2012-303594. [DOI] [PubMed] [Google Scholar]
- 68.Thrift AP, Kendall BJ, Pandeya N, et al. A model to determine absolute risk for esophageal adenocarcinoma. Clin Gastroenterol Hepatol. 2013;11:138–144. doi: 10.1016/j.cgh.2012.10.026. [DOI] [PubMed] [Google Scholar]
- 69.Atkinson M, Das A, Faulx A, et al. Ultrathin esophagoscopy in screening for Barrett's esophagus at a Veterans Administration Hospital: easy access does not lead to referrals. American Journal of Gastroenterology. 2008;103:92–97. doi: 10.1111/j.1572-0241.2007.01501.x. [DOI] [PubMed] [Google Scholar]
- 70.Gerson LB, Edson R, Lavori PW, et al. Use of a simple symptom questionnaire to predict Barrett's esophagus in patients with symptoms of gastroesophageal reflux. American Journal of Gastroenterology. 2001;96:2005–2012. doi: 10.1111/j.1572-0241.2001.03933.x. [DOI] [PubMed] [Google Scholar]
- 71.Avidan B, Sonnenberg A, Schnell TG, et al. Hiatal hernia and acid reflux frequency predict presence and length of Barrett's esophagus. Digestive Diseases & Sciences. 2002;47:256–264. doi: 10.1023/a:1013797417170. [DOI] [PubMed] [Google Scholar]
- 72.Chak A, Lee T, Kinnard MF, et al. Familial aggregation of Barrett's oesophagus, oesophageal adenocarcinoma, and oesophagogastric junctional adenocarcinoma in Caucasian adults. Gut. 2002;51:323–328. doi: 10.1136/gut.51.3.323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Smith KJ, O'Brien SM, Smithers BM, et al. Interactions among smoking, obesity, and symptoms of acid reflux in Barrett's esophagus. Cancer Epidemiol Biomarkers Prev. 2005;14:2481–2486. doi: 10.1158/1055-9965.EPI-05-0370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cook MB, Wild CP, Forman D. A systematic review and meta-analysis of the sex ratio for Barrett's esophagus, erosive reflux disease, and nonerosive reflux disease. Am J Epidemiol. 2005;162:1050–1061. doi: 10.1093/aje/kwi325. [DOI] [PubMed] [Google Scholar]
- 75.Corley DA, Kubo A, Levin TR, et al. Abdominal obesity and body mass index as risk factors for Barrett's esophagus. Gastroenterology. 2007;133:34–41. doi: 10.1053/j.gastro.2007.04.046. [DOI] [PubMed] [Google Scholar]
- 76.Johansson J, Hakansson HO, Mellblom L, et al. Risk factors for Barrett's oesophagus: a population-based approach. Scandinavian journal of gastroenterology. 2007;42:148–156. doi: 10.1080/00365520600881037. [DOI] [PubMed] [Google Scholar]
- 77.Edelstein ZR, Farrow DC, Bronner MP, et al. Central adiposity and risk of Barrett's esophagus. Gastroenterology. 2007;133:403–411. doi: 10.1053/j.gastro.2007.05.026. [DOI] [PubMed] [Google Scholar]
- 78.Anderson LA, Watson RG, Murphy SJ, et al. Risk factors for Barrett's oesophagus and oesophageal adenocarcinoma: results from the FINBAR study. World J Gastroenterol. 2007;13:1585–1594. doi: 10.3748/wjg.v13.i10.1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Cook MB, Greenwood DC, Hardie LJ, et al. A systematic review and meta-analysis of the risk of increasing adiposity on Barrett's esophagus. Am J Gastroenterol. 2008;103:292–300. doi: 10.1111/j.1572-0241.2007.01621.x. [DOI] [PubMed] [Google Scholar]
- 80.Edelstein ZR, Bronner MP, Rosen SN, et al. Risk factors for Barrett's esophagus among patients with gastroesophageal reflux disease: a community clinic-based case-control study. Am J Gastroenterol. 2009;104:834–842. doi: 10.1038/ajg.2009.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kubo A, Levin TR, Block G, et al. Alcohol types and sociodemographic characteristics as risk factors for Barrett's esophagus. Gastroenterology. 2009;136:806–815. doi: 10.1053/j.gastro.2008.11.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Anderson LA, Cantwell MM, Watson RG, et al. The association between alcohol and reflux esophagitis, Barrett's esophagus, and esophageal adenocarcinoma. Gastroenterology. 2009;136:799–805. doi: 10.1053/j.gastro.2008.12.005. [DOI] [PubMed] [Google Scholar]
- 83.Thompson OM, Beresford SA, Kirk EA, et al. Vegetable and fruit intakes and risk of Barrett's esophagus in men and women. Am J Clin Nutr. 2009;89:890–896. doi: 10.3945/ajcn.2008.26497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Smith KJ, O'Brien SM, Green AC, et al. Current and past smoking significantly increase risk for Barrett's esophagus. Clinical Gastroenterology & Hepatology. 2009;7:840–848. doi: 10.1016/j.cgh.2009.04.018. [DOI] [PubMed] [Google Scholar]
- 85.Leggett CL, Gorospe EC, Calvin AD, et al. Obstructive Sleep Apnea Is a Risk Factor for Barrett's Esophagus. Clin Gastroenterol Hepatol. 2013 Sep 11; doi: 10.1016/j.cgh.2013.08.043. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Thrift AP, Kramer JR, Richardson PA, et al. No Significant Effects of Smoking or Alcohol Consumption on Risk of Barrett's Esophagus. Dig Dis Sci. 2014;59:108–116. doi: 10.1007/s10620-013-2892-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Garcia JM, Splenser AE, Kramer J, et al. Circulating Inflammatory Cytokines and Adipokines Are Associated With Barrett's Esophagus: A Case-Control Study. Clin Gastroenterol Hepatol. 2014;12:229–238. doi: 10.1016/j.cgh.2013.07.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Wang KK, Sampliner RE. Updated guidelines 2008 for the diagnosis, surveillance and therapy of Barrett's esophagus. American Journal of Gastroenterology. 2008;103:788–797. doi: 10.1111/j.1572-0241.2008.01835.x. [DOI] [PubMed] [Google Scholar]
- 89.Hirota WK, Zuckerman MJ, Adler DG, et al. ASGE guideline: the role of endoscopy in the surveillance of premalignant conditions of the upper GI tract. Gastrointest Endosc. 2006;63:570–580. doi: 10.1016/j.gie.2006.02.004. [DOI] [PubMed] [Google Scholar]



