Targeting care in Barrett s oesophagus

Similar documents
Barrett s Esophagus: Old Dog, New Tricks

Current Management of Low-Grade Dysplasia in Barrett Esophagus

Barrett s Esophagus. Abdul Sami Khan, M.D. Gastroenterologist Aurora Healthcare Burlington, Elkhorn, Lake Geneva, WI

Citation for published version (APA): Phoa, K. Y. N. (2014). Endoscopic management of Barrett s esophagus with dysplasia

New Developments in the Endoscopic Diagnosis and Management of Barrett s Esophagus

MANAGEMENT OF BARRETT S RELATED NEOPLASIA IN 2018

Barrett esophagus. Bible class Inselspital

Ablation for Barrett s Esophagus: Burn or Freeze

History. Prevalence at Endoscopy. Prevalence and Reflux Sx. Prevalence at Endoscopy. Barrett s Esophagus: Controversy and Management

Disclosure Information

Screening Patients for Barrett s Esophagus with Cytosponge Coupled with Trefoil Factor 3 Expression Compared to Endoscopy

ACG Clinical Guideline: Diagnosis and Management of Barrett s Esophagus

Barrett s Esophagus: What to Do for No Dysplasia, LGD, and HGD?

Page 1. Is the Risk This High? Dysplasia in the IBD Patient. Dysplasia in the Non IBD Patient. Increased Risk of CRC in Ulcerative Colitis

Everything Esophagus: Barrett s Esophagus. Nicholas Shaheen, MD, MPH Center for Esophageal Diseases and Swallowing University of North Carolina

Definition of GERD American College of Gastroenterology

Barrett s Esophagus: Ablate Everyone?

Histopathology of Endoscopic Resection Specimens from Barrett's Esophagus

In 1998, the American College of Gastroenterology issued ALIMENTARY TRACT

Barrett s esophagus. Barrett s neoplasia treatment trends

The Pathologist s Role in the Diagnosis and Management of Neoplasia in Barrett s Oesophagus Cian Muldoon, St. James s Hospital, Dublin

Editorial: Advanced endoscopic therapeutics in Barrett s neoplasia; where are we now and where are we heading?

Screening of Barrett: Is it cost-effective? Is there a high-risk population? T Ponchon Ed. Herriot Hospital Lyon, France

Present Day Management of Barrett s Esophagus

SAM PROVIDER TOOLKIT

Changes to the diagnosis and management of Barrett s Oesophagus

Barrett s Esophagus. Radiofrequency Ablation with the HALO Technology A Reference Book

Sixteen-year follow-up of Barrett s esophagus, endoscopically treated with argon plasma coagulation

Joel A. Ricci, MD SUNY Downstate Medical Center Department of Surgery

Is Radiofrequency Ablation Effective In Treating Barrett s Esophagus Patients with High-Grade Dysplasia?

Barrett's Esophagus: Sorting Out the Controversy

Learning Objectives:

Alberta Colorectal Cancer Screening Program (ACRCSP) Post Polypectomy Surveillance Guidelines

Current Management: Role of Radiofrequency Ablation

Oesophagus and Stomach update dysplasia and early cancer

Original article INTRODUCTION

Barrett s esophagus: lessons from recent clinical trials

Chromoendoscopy and Endomicroscopy for detecting colonic dysplasia

Endoscopic Management of Barrett s Esophagus

Greater Manchester & Cheshire Guidelines for Pathology Reporting for Oesophageal and Gastric Malignancy

Accepted Manuscript. CGH Editorial: Sound the Alarm for Barrett s Screening! Tarek Sawas, M.D., M.P.H., David A. Katzka, M.D

Barrett s Oesophagus Information Leaflet THE DIGESTIVE SYSTEM. gutscharity.org.

Management of Barrett s: From Imaging to Resection

Greater Manchester & Cheshire Guidelines for Pathology Reporting of Oesophageal and Gastric Malignancy

Paris classification (2003) 삼성의료원내과이준행

Management of Barrett s Esophagus. Case Presentation

Stage 4 gastric adenocarcinoma icd 10

Gregory G. Ginsberg, M.D.

Clinical studies of indicative biopsies in opportunistic screening in high risk population of oesophageal cancer.

How to characterize dysplastic lesions in IBD?

Relative risk of dysplasia for patients with intestinal metaplasia in the distal oesophagus and in the gastric cardia

LIST OF ABBREVIATIONS

Barrett s Esophagus: Review of Diagnostic Issues and Pre- Neoplastic Lesions

Vital staining and Barrett s esophagus

AGA SECTION. Gastroenterology 2016;150:

Improved diagnostic stratification of digitised Barrett s oesophagus biopsies by p53 immunohistochemical staining

Identification of gastric atrophic changes: from histopathology to endoscopy

Endoscopic Submucosal Dissection ESD

Endoscopic Radiofrequency Ablation or Cryoablation for Barrett s Esophagus

Volumetric laser endomicroscopy can target neoplasia not detected by conventional endoscopic measures in long segment Barrett s esophagus

Esophageal adenocarcinoma is a cancer with increasing

Pathology in Slovenian CRC screening programme:

Dysplasia 4/19/2017. How do I practice Chromoendoscopy for Surveillance of Colitis? SCENIC: Polypoid Dysplasia in UC. Background

Philip Chiu Associate Professor Department of Surgery, Prince of Wales Hospital The Chinese University of Hong Kong

Populations Interventions Comparators Outcomes Individuals: With Barrett esophagus with high-grade dysplasia

SAMs Guidelines DEVELOPING SELF-ASSESSMENT MODULES TEST QUESTIONS. Ver. #

Gastrointestinal pathology 2018 lecture 2. Dr Heyam Awad FRCPath

Opinion Statement. Esophagus (E Dellon, Section Editor)

Is intestinal metaplasia a necessary precursor lesion for adenocarcinomas of the distal esophagus, gastroesophageal junction and gastric cardia?

RFA and Cyrotherapy for Esophageal Disease

Management of pt1 polyps. Maria Pellise

Endoscopic Radiofrequency Ablation or Cryoablation for Barrett Esophagus

Esophageal cancer: Biology, natural history, staging and therapeutic options

NPQR Quality Payment Program (QPP) Measures 21_18247_LS.

Novel endoscopic observation in Barrett s oesophagus using high resolution magnification endoscopy and narrow band imaging

Current status of gastric ESD in Korea. Jun Haeng Lee. Department of Medicine Sungkyunkwanuniversity School of Medicie, Seoul, Korea

Endoscopic management of Barrett s esophagus:european Society of Gastrointestinal Endoscopy (ESGE) Position Statement

Section: Medicine Effective Date: July 15, 2015 Subsection: Original Policy Date: December 7, 2011 Subject:

EMR, ESD and Beyond. Peter Draganov MD. Professor of Medicine Division of Gastroenterology, Hepatology and Nutrition University of Florida

National Oesophago-Gastric Cancer Audit New Patient Registration sheet Patients with Oesophageal High Grade Glandular Dysplasia

It has been estimated that 5.6% of adults in the United States have

American Journal of Gastroenterology. Volumetric Laser Endomicroscopy Detects Subsquamous Barrett s Adenocarcinoma

Frozen Section Analysis of Esophageal Endoscopic Mucosal Resection Specimens in the Real-Time Management of Barrett s Esophagus

Barrett s Esophagus: State of the Art. Food Getting Stuck

CASE DISCUSSION: The Patient with Dysplasia: Surgery or Active Surveillance? Noa Krugliak Cleveland, MD David T. Rubin, MD

Endoscopic Radiofrequency Ablation or Cryoablation for Barrett`s Esophagus. Original Policy Date

Advanced endoscopic imaging of esophageal neoplasia; old looks and new visions Boerwinkel, David

Morphologic Criteria of Invasive Colonic Adenocarcinoma on Biopsy Specimens

Hiatal Hernias and Barrett s esophagus. Dr Sajida Ahad Mercy General Surgery

Medicare Advantage Medical Policy

Advances in Endoscopic Imaging

Chromoendoscopy or Narrow Band Imaging with Targeted biopsies Should be the Cancer Surveillance Endoscopy Procedure of Choice in Ulcerative Colitis

Burning Issues in the Esophagus

Treat Barrett s, Remove the Risk. HALO System

University Mainz. Early Gastric Cancer. Ralf Kiesslich. Johannes Gutenberg University Mainz, Germany. Early Gastric Cancer 15.6.

Faculty Disclosure. Objectives. State of the Art #3: Referrals for Gastroscopy (focus on common esophagus problems) 24/11/2014

Earlyoesophagealcancer. dr. Nina Zidar Institute of Pathology Faculty ofmedicine University of Ljubljana Slovenia

UK National Screening Committee. Screening for Stomach Cancer. 12 February 2016

CLINICAL EFFECTIVENESS

What s New in the Management of Esophageal Disease

Transcription:

HEPATO-GASTROENTEROLOGY Clinical Medicine 2014 Vol 14, No 6: s78 s83 Targeting care in Barrett s oesophagus Authors: Sebastian Zeki A and Rebecca C Fitzgerald B ABSTRACT Barretts oesophagus represents the most significant risk factor for the development of oesophageal adenocarcinoma (OAC), although the majority of patients will not develop cancer. However, early detection of OAC and its precursors significantly improves outcome and underlines the importance of endoscopic surveillance programmes. Clearly there is a discrepancy between the small number of people who need to undergo surveillance because they are at significant progression risk, and the large number that do. Research is therefore now concentrated on risk stratification. Currently such stratification is currently based on clinical findings, endoscopic diagnosis and histopathological grade. Histopathology can be imperfect and is likely to require molecular confirmation of different grades, thus molecular stratification is becoming more important in this regard and p53 immunohistochemistry is already clinically useful, with other molecular biomarkers likely to prove beneficial in the future. The hope is that non-endoscopic methods, such as the Cytosponge TM may be able to combine molecular biomarkers with histopathology and therefore perhaps benefit a population screening as well as a surveillance programme. KEYWORDS: Barrett s oesophagus, oesophageal adenocarcinoma, molecular stratification Introduction Oesophageal adenocarcinoma (OAC) has a high mortality with a median overall survival of 1 year; even when a curative modality is attempted, for cases without metastases to distant organs, 5-year survival remains poor. 1 There has been a dramatic increase in this disease in the western world over the past 3 decades and, although more recent data have demonstrated that the incidence of OAC might now be plateauing worldwide, in the UK the incidence is increasing and outcome remains poor. 2 One of the most commonly associated risk factors with OAC is the development of Barrett s oesophagus (BO). BO is thought to be the result of a metaplasia from the normal squamous lining of the oesophagus to a columnar, glandular Authors: A postdoctoral research fellow, University of Cambridge, Cambridge, UK; B professor of cancer prevention, University of Cambridge, Cambridge, UK phenotype. Although the cellular origin of BO has not yet been clarified, there is strong evidence from epidemiological and experimental data that the aetiology is related to refluxate. BO is characterised by several cell lineages, including intestinal metaplasia, which is the hallmark and the type that predisposes most to adenocarcinoma. The development of cancer is generally gradual through a metaplasia dysplasia cancer sequence, which is a well-established progression sequence for several different, particularly epithelial, cancers. The sequence is often considered to be temporally linear: metaplasia develops into dysplasia, either of low or high grade (LGD or HGD, respectively), which is the precursor lesion to adenocarcinoma. Importantly, if adenocarcinoma is detected when it is confined to the mucosa (so-called intramucosal carcinoma ), the 5-year survival rate is 95%, whereas invasion into the deeper submucosa increases the risk of lymph node metastases considerably (from 5% to 40% depending on degree of submucosal invasion) and increases the 5-year mortality. 3,4 This is the premise for the importance of early detection in this disease. Cancers take years to evolve understanding precursor lesions and early cancers as targets Sporadic colon cancer, for example, is known to take 5 15 years to develop and is preceded by adenomas comprising LGD or HGD. 5 Prostate, cervical, bladder and breast cancers are all epithelial cancers with precursor lesions that take many years to evolve before invasive cancer occurs (Table 1). Therefore, detectable precursor lesions represent an ideal target for the prevention of cancer development. Such strategies are already integral to several cancer prevention programmes. For example, the UK Bowel Cancer Surveillance Programme screens for the detection of cancer and uses a guideline-based follow-up of adenoma detection and eradication. 6 Another example is the Japanese model of gastric cancer screening. Since the introduction of the national gastric cancer screening programme in Japan, the detection of early gastric cancer has increased significantly with a consistent decrease in patient mortality. 7 Given the importance of early cancer detection and the possibility of treating cancer precursor lesions to prevent cancer development, the question arises concerning how we can best achieve this goal given the low progression rate in BO. At the two ends of the spectrum should we be investigating only patients with upper gastrointestinal (GI) symptoms, those who are considered to be high risk based on clinicopathological features, or should we consider population screening? s78 Royal College of Physicians 2014. All rights reserved.

Targeting care in Barrett s oesophagus Table 1. Examples of cancers and their associated precursor lesions. Site Precursor Time to cancer (years) Colon Adenoma 5 15 Head and neck Oral leukoplakia 6 8 Oesophagus Barrett s 10 20 Cervix Cervical intra-epithelial neoplasia 10 30 Lung Dysplasia 5 20 Skin Actinic keratosis 30 50 Breast Ductal carcinoma in situ 10 20 Prostate Prostatic intraepithelial neoplasia 5 15 Bladder Papillary transitional carcinoma in situ 20 25 The evidence for surveillance Several studies have demonstrated that the overall risk of developing OAC in patients with non-dysplastic BO is small, with two large population-based cohort studies concluding that the per patient risk of OAC is between 0.12% and 0.16% per year. Patients were at greater risk if goblet cells were present (intestinal metaplasia) and with longer Barrett s segments (>3 cm). 8,9 Given the low rate of OAC development and that surveillance programmes exist in most western countries, it is worth considering what the impact of surveillance regimes has been on the development of OAC or its mortality. Although several studies found a positive impact on survival, 10,11 the most recent study from Kaiser Permanente in the USA argues against the need for surveillance. 12 This case-control study by Corley et al matched 38 OAC deaths by age, year and place of diagnosis, sex and race, with 101 controls (patients with Barrett s but who did not die from oesophageal adenocarcinoma by the end of the study, although patients in this group could still have oesophageal cancer) from a large database kept between 1995 and 2007. The authors concluded that a surveillance gastroscopy within 3 years of the diagnosis of cancer did not decrease the risk of death from OAC. This study highlights some of the challenges related to determining surveillance impact in an area where endoscopic technologies and treatments change and where surveillance might not be standardised. Of the patients in this study who developed cancer, 47.4% had previously been found to have dysplasia before cancer development and, in those who had developed cancer, approximately 50% had cancer diagnosed at an advanced stage, indicating that the quality of the surveillance was poor. Therefore, their study underlines the need for high-quality surveillance according to strict protocol. Furthermore, whereas treatment by oesophagectomy would Squamous Non-dysplas c Barre s oesophgus Low grade dysplasia High grade dysplasia Adenocarcinoma Fig 1. An endoscopic (top panels) and histological (bottom panels) description of the development of oesophageal adenocarcinoma in patients with Barrett s oesophagus. The initial lining of the oesophagus comprises squamous cells. These are then replaced by non-dysplastic columnar cells with or without goblet cells. In a few patients, further molecular and genetic aberrations result in low- and then high-grade dysplasia. The latter is the immediate precursor of, and often coexists with, adenocarcinoma. Royal College of Physicians 2014. All rights reserved. s79

Sebastian Zeki and Rebecca C Fitzgerald Endoscopic diagnosis Endoscopic evidence of columnar lined oesophagus according to Prague criteria Quadran c oesophageal biopsies every 2 cm Histological diagnosis Squamous epithelium Glandular metaplasia Barre s oesophagus Sampling error Review endoscopic findings Consider repeat OGD Clinical review of pa ent fitness and preference Endoscopic-histological correla on Maximum length <3 cm Gastric metaplasia Repeat OGD* Length <3 cm Gastric metaplasia Consider discharging Maximum length <3 cm Intes nal metaplasia Repeat OGD every 3 to 5 years Maximum length 3cm Repeat OGD every 2 to 3 years Fig 2. Algorithm for the endoscopic evaluation and follow-up for patients with non-dysplastic Barrett s oesophagus. *Interval depends on the degree of clinical confidence about diagnosis (accuracy of endoscopic report and number of biopsies). Reproduced with permission from Gut. 13 OGD = oesophago-gastro-duodenoscopy. have been available for HGD in this historical cohort, the more successful endoscopic therapies, such as radiofrequency ablation, which are now preferred for early disease, would have only been available after this cohort was collected. Thus, this interesting study not only highlights the difficulty of assessing historical data and retrospective studies, but also underlines the importance of stratifying patients according to their risk of developing cancer, especially given the ability of current endoscopic therapy to eradicate earlier oesophageal precursor lesions. The growing emphasis on targeting the right patients for surveillance Therefore, the standardisation of clinical care for BO is an important part of assessing the true impact of surveillance programmes. The UK guidelines for the diagnosis and treatment of Barrett s have recently been updated. 13 Important evidence-based changes have been introduced that start to clarify how patients can be risk stratified according to clinicopathological features (Fig 2). Clinical stratification The Prague classification for endoscopic reporting of Barrett s segments, which reports the circumferential extent (denoted C for circumferential and the length in cm) as well as total extent (taking tongues of Barrett s into account (M)) from the gastrooesophageal junction, is now a standard endoscopic reporting descriptor. This should be combined with the Paris classification, which standardises the description of visible oesophageal lesions. As regards risk stratification, several studies have demonstrated that patients with longer Barrett s sections (>3 cm) at endoscopy are considered to be at higher risk 14 16 of OAC progression and, therefore, the intensity of endoscopic surveillance is modified between 2 and 5 years accordingly. The need for high-quality, high-resolution endoscopy The need for accurate and high-quality endoscopy has also increased in importance as treatment for early OAC becomes more accurate and targeted. Much progress has been made s80 Royal College of Physicians 2014. All rights reserved.

Targeting care in Barrett s oesophagus High-resolu on endoscopy (HRE) Autofluorescence imaging (AFI) Narrowband imaging-zoom (NBI-Z) Fig 3. Different endoscopic modalities for the examination of a Barrett s segment. AFI = autofluorescence imaging; HRE = high-resolution endoscopy; NBI-Z = narrowband imaging-zoom. with endoscopic modalities for early OAC and HGD treatment, but these treatments rely on the recognition of lesions, which can be subtle. The endoscopist s tool-kit contains several modalities to enhance lesion recognition, including autofluorescence, narrowband imaging, confocal microscopy and chromoendoscopy (Fig 3). Although still experimental, the ability to target suspicious areas at endoscopy reduces unnecessary sampling and can increase the yield of potentially treatable areas. 17 Given that modalities such as radiofrequency ablation with or without endoscopic or submucosal mucosal resection are now curative in most cases, including early OAC, such targeting is of paramount importance. Histological stratification Histologically, although the presence of intestinal metaplasia is not a prerequisite for the diagnosis of BO, as it is in North America, its presence might also indicate a higher risk of OAC development 9,18,19 and, therefore, endoscopic follow-up of these patients is also stratified accordingly. A key emphasis is placed on the fact that, if the patient has gastric metaplasia without goblet cells, and the Barrett s segment is less than 3 cm, then discharge from surveillance should be considered because the risk of endoscopy might outweigh the benefit of surveillance. Given the importance of a histological diagnosis, the guidelines also stress the need for histopathological quality control, particularly in relation to dysplasia grade, because this can trigger a therapeutic intervention with endoscopic therapy. The detection of LGD carries a hazard ratio of 5.67 for the development of HGD and OAC combined. 9 Historically there have also been different treatment pathways based on the detection of LGD and HGD; thus, the lack of consensus in diagnosing dysplasia subtypes between histopathologists has been of concern. Patients with HGD will all need to be discussed at the local multidisciplinary team meeting after consensus review and, depending on endoscopic and histopathological features, might undergo radiofrequency ablation, endoscopic mucosal resection, surgery or a combination thereof. However, patients with LGD were advised to undergo a gastroscopy every 6 months until two consecutive endoscopies demonstrated non-dysplastic Barrett s or the patient developed HGD. Several studies have demonstrated kappa values for interobserver agreement in the diagnosis of LGD as low as 0.18. 20 In fact, although LGD without consensus is reported to have an incidence rate of later OAC development of 0.49%, at least one study has demonstrated that, with consensus between at least two histopathologists, the incidence rate increases to 13.4%. 21 A recent randomised controlled trial has demonstrated that with consensus between expert GI pathologists, patients who undergo ablation for LGD are significantly less likely to develop adenocarcinoma and therefore the ablation of Barrett's containing confirmed LGD is now recommended. 22 Can molecular biology help histopathological diagnosis? Molecular stratification Based on the above discussion, strategies to increase the consensus around a diagnosis of LGD would be helpful. The protein product, p53 of the tumour suppressor gene TP53, which is often mutated in the progression to OAC, is now validated as an adjunct to help clarify the diagnosis of LGD. An abnormal pattern is associated with an increased risk of OAC progression in patients with BO (Fig 4). It is also a better predictor of neoplastic progression compared with the histological diagnosis of LGD and has the benefit of good interobserver agreement. 23,24 The future considering population screening Cytosponge for population screening Most of the work in BO has been undertaken in patients who have already been found to have the condition and, usually, they have presented with upper GI symptoms of reflux. However, given that the population prevalence of BO is estimated to be around 2% of the total population in the western world, over 80% of patients with BO will remain undiagnosed. 25,26 To reduce the incidence of OAC, it follows that we should also concentrate on diagnosing the population at risk through screening as well as risk stratifying those with diagnosed BO. Endoscopic population screening would be unfeasible and, therefore, recent trials have assessed novel technologies for Royal College of Physicians 2014. All rights reserved. s81

Sebastian Zeki and Rebecca C Fitzgerald (a) (b) Fig 4. Immunohistochemistry for p53. (a) Overexpression of p53. (b) Absent p53 pattern. Both are pathological. this purpose, including ultrathin transnasal endoscopy and non-endoscopic technologies, such as the Cytosponge. The Cytosponge is a capsule on a string, which the patient swallows. Once in the stomach, the capsule dissolves to release a spherical sponge mesh that can then be retrieved using the string; during its retrieval, approximately 500,000 oesophageal cells accumulate within the mesh. The administration and retrieval of the Cytopsonge takes 5 minutes and can be performed in the GP surgery. These cells can then be prepared for histological and molecular analyses with immunohistochemical analysis of a Barrett s marker, trifoil factor 3 (TFF3), proving crucial for the success of this test. The Cytosponge test combined with TFF3 detection was compared with endoscopy and biopsy in a primary care cohort study of just over 500 patients. 27 The sensitivity and specificity of Cytosponge and TFF3 detection for the detection of BO was 73.3% and 93.8%, respectively for short-segment BO (<1 cm circumferential length) and increased to 90% and 93.5%, respectively for longer segments >3 cm. This compares well with tests used in other national cancer screening programmes. A second study of over 1,000 patients is now being completed to evaluate this approach further and the results are awaited (in a Cancer Research UK-funded and National Institute of Health Research portfolio study called BEST2 ). The Cytosponge raises the possibility of population screening for BO while uncoupling endoscopy units from the burden of carrying out what would necessarily be a large screening programme. An obvious next step is to ask whether the Cytosponge test could be taken further so that, once the BO has been detected, further analyses might enable patients with BO to be stratified as high or low risk for progression to OAC. This possibility will be evaluated in the recently completed BEST2 study. Using non-endoscopic means, such as the Cytosponge, to both diagnose and risk stratify those with BO might not only reduce the burden on patients having to undergo endoscopic surveillance for BO, but also reduce the healthcare cost burden by reducing the overall number of endoscopies needed. 28 A future paradigm is demonstrated in Fig 6, whereby symptomatic patients with heartburn would undergo Cytosponge assessment and, if found to contain Barrett s Heartburn sufferers: 10% of adults aged 50 70 years Cytosponge TM 90% 9% 1% Nega ve Low risk posi ve High risk posi ve Repeat 3 5 years Fig 5. The Cytosponge. The Cytosponge is swallowed as a capsule. The soft mesh expands once in the stomach and, on withdrawal, the mesh traps a sample of oesophageal cells, which can then be analysed. Discharge Cytosponge TM Endoscopy and treatment if confirmed Fig 6. A possible future algorithm for the stratification of patients with Barrett s oesophagus using the Cytosponge. s82 Royal College of Physicians 2014. All rights reserved.

Targeting care in Barrett s oesophagus cells as determined by histology and TFF3, they would undergo further histological and molecular analyses to determine the risk of progression to OAC. On the basis of this stratification, patients might then be offered a further Cytosponge at a later date if they are low risk, or an endoscopy if there is evidence of dysplasia, to treat with the appropriate endoscopic modality. Although work has started to stratify patients according to clinical, histological and molecular risk, there is more work to be done. Factors, such as a patient s age or sex, body mass index or waist:hip ratio and drug intake, have all been implicated in Barrett s progression and, pending further studies, might have a part in deciding the need for surveillance. Similarly, advanced imaging techniques might enable targeted biopsy in patients who otherwise would undergo standardised sampling. Finally, genetic and molecular markers are likely to have an increasingly important role in predicting those who will progress to OAC and also in diagnosing different degrees of dysplasia. References 1 Atkins BZ, Shah AS, Hutcheson KA et al. Reducing hospital morbidity and mortality following esophagectomy. Ann Thorac Surg 2004;78:1170 6. 2 Murray CJL, Richards MA, Newton JN et al. UK health performance: findings of the Global Burden of Disease Study 2010. Lancet 2013;381:997 1020. 3 Ancona E, Rampado S, Cassaro M et al. Prediction of lymph node status in superficial esophageal carcinoma. Ann Surg Oncol 2008;15:3278 88. 4 Rice TW, Zuccaro G, Adelstein DJ et al. Esophageal carcinoma: depth of tumor invasion is predictive of regional lymph node status. Ann Thorac Surg 1998;65:787 92. 5 Jones S, Chen W-D, Parmigiani G et al. Comparative lesion sequencing provides insights into tumor evolution. Proc Natl Acad Sci U S A 2008;105:4283 8. 6 Cairns SR, Scholefield JH, Steele RJ et al. Guidelines for colorectal cancer screening and surveillance in moderate and high risk groups (update from 2002). Gut 2010;59:666 89. 7 Lambert R, Guilloux A, Oshima A et al. Incidence and mortality from stomach cancer in Japan, Slovenia and the USA. Int J Cancer 2002;97:811 8. 8 Hvid-Jensen F, Pedersen L, Drewes AM et al. Incidence of adenocarcinoma among patients with Barrett s esophagus. N Engl J Med 2011;365:375 83. 9 Bhat S, Coleman HG, Yousef F et al. Risk of malignant progression in Barrett s esophagus patients: results from a large populationbased study. J Natl Cancer Inst 2011;103:1049 57. 10 Cooper SC, El-agib A, Dar S et al. Endoscopic surveillance for Barrett s oesophagus: the patients perspective. Eur J Gastroenterol Hepatol 2009;21:850 4. 11 Rubenstein JH, Sonnenberg A, Davis J et al. Effect of a prior endoscopy on outcomes of esophageal adenocarcinoma among United States veterans. Gastrointest Endosc 2008;68:849 55. 12 Corley DA, Mehtani K, Quesenberry C et al. Impact of endoscopic surveillance on mortality from Barrett s esophagus-associated esophageal adenocarcinomas. Gastroenterology 2013;145:312 9. 13 Fitzgerald RC, Pietro M, Ragunath K et al. British Society of Gastroenterology guidelines on the diagnosis and management of Barrett s oesophagus. Gut 2013;0:1 36. 14 Yousef F, Cardwell C, Cantwell MM, Galway K 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;168:237 49. 15 Thomas T, Abrams KR, De Caestecker JS, Robinson RJ. Meta analysis: cancer risk in Barrett s oesophagus. Aliment Pharmacol Ther 2007;26:1465 77. 16 Desai TK, Krishnan K, Samala N et al. The incidence of oesophageal adenocarcinoma in non-dysplastic Barrett s oesophagus: a meta-analysis. Gut 2012;61:970 6. 17 Boerwinkel DF, Swager A-F, Curvers WL, Bergman JJGHM. The clinical consequences of advanced imaging techniques in Barrett s esophagus. Gastroenterology 2014;146:622 9. 18 Smith RR, Hamilton SR, Boitnott JK, Rogers EL. The spectrum of carcinoma arising in Barrett s esophagus. A clinicopathologic study of 26 patients. Am J Surg Pathol 1984;8:563 73. 19 Skinner DB, Walther BC, Riddell RH et al. Barrett s esophagus. Comparison of benign and malignant cases. Ann Surg 1983;198:554 65. 20 Wani S, Falk GW, Post J et al. Risk factors for progression of lowgrade dysplasia in patients with Barrett s esophagus. Gastroenterology 2011;141:1179 86. 21 Curvers WL, ten Kate FJ, Krishnadath KK et al. Low-grade dysplasia in Barrett s esophagus: overdiagnosed and underestimated. Am J Gastroenterol 2010;105:1523 30 22 Phoa KN, van Vilsteren FG, Weusten BL et al. Radiofrequency ablation vs endoscopic surveillance for patients with Barrett s esophagus and low-grade dysplasia: a randomized clinical trial. JAMA 2014;311:1209 17. 23 Skacel M, Petras RE, Rybicki LA et al. p53 expression in low grade dysplasia in Barrett s esophagus: correlation with interobserver agreement and disease progression. Am J Gastroenterol 2002;97:2508 13. 24 Kastelein F, Biermann K, Steyerberg EW et al. Aberrant p53 protein expression is associated with an increased risk of neoplastic progression in patients with Barrett s oesophagus. Gut 2013;62:1676 83. 25 Zagari RM, Fuccio L, Wallander M-A et al. Gastro-oesophageal reflux symptoms, oesophagitis and Barrett s oesophagus in the general population: the Loiano-Monghidoro study. Gut 2008;57:1354 9. 26 Ronkainen J, Aro P, Storskrubb T et al. Prevalence of Barrett s esophagus in the general population: an endoscopic study. Gastroenterology 2005;129:1825 31. 27 Kadri SR, Lao-Sirieix P, O Donovan M et al. Acceptability and accuracy of a non-endoscopic screening test for Barrett s oesophagus in primary care: cohort study. BMJ 2010;341:c4372. 28 Benaglia T, Sharples LD, Fitzgerald RC, Lyratzopoulos G. Health benefits and cost effectiveness of endoscopic and nonendoscopic cytosponge screening for Barrett s esophagus. Gastroenterology 2013;144:62 73. Address for correspondence: Prof R Fitzgerald, MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Hills Road, Cambridge, CB2 0XZ, UK. Email: rcf29@mrc-cu.cam.ac.uk Royal College of Physicians 2014. All rights reserved. s83