51
|
Grady WM, Yu M, Markowitz SD, Chak A. Barrett's Esophagus and Esophageal Adenocarcinoma Biomarkers. Cancer Epidemiol Biomarkers Prev 2020; 29:2486-2494. [PMID: 33093162 DOI: 10.1158/1055-9965.epi-20-0223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/31/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
Esophageal adenocarcinoma is a major cause of cancer-related morbidity and mortality in Western countries. The incidences of esophageal adenocarcinoma and its precursor Barrett's esophagus have increased substantially in the last four decades. Current care guidelines recommend that endoscopy be used for the early detection and monitoring of patients with Barrett's esophagus; however, the efficacy of this approach is unclear. To prevent the increasing morbidity and mortality from esophageal adenocarcinoma, there is a tremendous need for early detection and surveillance biomarker assays that are accurate, low-cost, and clinically feasible to implement. The last decade has seen remarkable advances in the development of minimally invasive molecular biomarkers, an effort led in large part by the Early Detection Research Network (EDRN). Advances in multi-omics analysis, the development of swallowable cytology collection devices, and emerging technology have led to promising assays that are likely to be implemented into clinical care in the next decade. In this review, an updated overview of the molecular pathology of Barrett's esophagus and esophageal adenocarcinoma and emerging molecular biomarker assays, as well as the role of EDRN in biomarker discovery and validation, will be discussed.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- William M Grady
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington. .,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ming Yu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sanford D Markowitz
- Oncology Division, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Amitabh Chak
- Gastroenterology Division, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
52
|
Maitra I, Date RS, Martin FL. Towards screening Barrett's oesophagus: current guidelines, imaging modalities and future developments. Clin J Gastroenterol 2020; 13:635-649. [PMID: 32495144 PMCID: PMC7519897 DOI: 10.1007/s12328-020-01135-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
Barrett's oesophagus is the only known precursor to oesophageal adenocarcinoma (OAC). Although guidelines on the screening and surveillance exist in Barrett's oesophagus, the current strategies are inadequate. Oesophagogastroduodenoscopy (OGD) is the gold standard method in screening for Barrett's oesophagus. This invasive method is expensive with associated risks negating its use as a current screening tool for Barrett's oesophagus. This review explores current definitions, epidemiology, biomarkers, surveillance, and screening in Barrett's oesophagus. Imaging modalities applicable to this condition are discussed, in addition to future developments. There is an urgent need for an alternative non-invasive method of screening and/or surveillance which could be highly beneficial towards reducing waiting times, alleviating patient fears and reducing future costs in current healthcare services. Vibrational spectroscopy has been shown to be promising in categorising Barrett's oesophagus through to high-grade dysplasia (HGD) and OAC. These techniques need further validation through multicentre trials.
Collapse
Affiliation(s)
- Ishaan Maitra
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE UK
| | | | | |
Collapse
|
53
|
di Pietro M, Modolell I, O'Donovan M, Price C, Pilonis ND, Debiram-Beecham I, Fitzgerald RC. Use of Cytosponge as a triaging tool to upper gastrointestinal endoscopy during the COVID-19 pandemic. Lancet Gastroenterol Hepatol 2020; 5:805-806. [PMID: 32738939 PMCID: PMC7392598 DOI: 10.1016/s2468-1253(20)30242-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Massimiliano di Pietro
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, CB2 0XZ, UK; Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, UK
| | - Ines Modolell
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, CB2 0XZ, UK; Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, UK
| | - Maria O'Donovan
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, CB2 0XZ, UK; Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, UK
| | - Catherine Price
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, UK
| | - Nastazja D Pilonis
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, CB2 0XZ, UK
| | - Irene Debiram-Beecham
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, CB2 0XZ, UK
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, CB2 0XZ, UK; Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, UK.
| |
Collapse
|
54
|
Dam AN, Klapman J. A narrative review of Barrett's esophagus in 2020, molecular and clinical update. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1107. [PMID: 33145326 PMCID: PMC7575938 DOI: 10.21037/atm-20-4406] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Barrett’s esophagus (BE) is a condition resulting from an acquired metaplastic epithelial change in the esophagus in response to gastroesophageal reflux. BE is the only known precursor lesion to esophageal adenocarcinoma, and can progress from non-dysplastic BE (NDBE) to low grade dysplasia (LGD) and high grade dysplasia (HGD), and ultimately invasive carcinoma. Although the risk of developing esophageal adenocarcinoma (EAC) in NBDE is less than 0.5% per year, there has been a rising incidence of EAC in Western countries, which continue to drive efforts to optimize screening and surveillance methods. The current gold standard for diagnosis is esophagogastroduodenoscopy (EGD), and there has been significant interest in alternative, minimally invasive methods for screening which would be more readily accessible in the primary care setting. Surveillance endoscopy in 3–5 years is recommended for NDBE given the low progression to EAC. The mainstay of treatment for LGD and HGD is endoscopic eradication therapy (EET). Visible lesions are treated with endoscopic mucosal resection (EMR) or endoscopic submucosal dissection (ESD). Radiofrequency ablation (RFA) is considered first line therapy for flat dysplastic BE and cryotherapy has shown promising results as an alternate form of treatment for of dysplasia. The molecular progression of BE to EAC is a complex process involving multiple pathways involving genetic and epigenetic modifications. Genomic studies have further led to the understanding of the complex molecular landscape that occurs early and late in the disease process. Promising biomarker panels have been investigated to help with the diagnosis of BE as well as aid in the risk stratification of BE during surveillance. In addition, clinical prediction models have been developed to categorize BE patients in low, intermediate, and high risk for progression to HGD and EAC. Further clinical and translational research is needed to help refine markers and techniques in diagnosis, screening, and surveillance.
Collapse
Affiliation(s)
- Aamir N Dam
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jason Klapman
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
55
|
Krause J, Rosenbaum C, Grimm M, Rump A, Keßler R, Hosten N, Weitschies W. The EsoCap-system - An innovative platform to drug targeting in the esophagus. J Control Release 2020; 327:1-7. [PMID: 32781172 DOI: 10.1016/j.jconrel.2020.08.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
For the therapy of esophageal diseases such as eosinophilic esophagitis, there are no possibilities of local targeted therapy. This publication describes a novel, innovative drug delivery concept, that enables a targeted, long-lasting administration of drug substances to the esophageal mucosa. In addition to a comprehensive in-vitro characterization of the dosage form, this work includes a proof-of-concept study with healthy volunteers, which shows the functionality and acceptance of this novel drug delivery concept. This novel drug delivery technology enables for the first time a targeted, local and long-lasting therapy of the esophagus.
Collapse
Affiliation(s)
- Julius Krause
- University of Greifswald, Institute of Pharmacy, Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, Felix-Hausdorff-Str. 3, 17487 Greifswald, Germany
| | - Christoph Rosenbaum
- University of Greifswald, Institute of Pharmacy, Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, Felix-Hausdorff-Str. 3, 17487 Greifswald, Germany
| | - Michael Grimm
- University of Greifswald, Institute of Pharmacy, Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, Felix-Hausdorff-Str. 3, 17487 Greifswald, Germany
| | - Adrian Rump
- University of Greifswald, Institute of Pharmacy, Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, Felix-Hausdorff-Str. 3, 17487 Greifswald, Germany
| | - Rebecca Keßler
- Department of Diagnostic Radiology and Neuroradiology, University Hospital Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Norbert Hosten
- Department of Diagnostic Radiology and Neuroradiology, University Hospital Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Werner Weitschies
- University of Greifswald, Institute of Pharmacy, Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, Felix-Hausdorff-Str. 3, 17487 Greifswald, Germany.
| |
Collapse
|
56
|
Hanada Y, Wang KK. Screening for Barrett's oesophagus: is now the time? Lancet 2020; 396:292-293. [PMID: 32738941 PMCID: PMC7392563 DOI: 10.1016/s0140-6736(20)31531-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/25/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Yuri Hanada
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kenneth K Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
57
|
Fitzgerald RC, di Pietro M, O'Donovan M, Maroni R, Muldrew B, Debiram-Beecham I, Gehrung M, Offman J, Tripathi M, Smith SG, Aigret B, Walter FM, Rubin G, Sasieni P. Cytosponge-trefoil factor 3 versus usual care to identify Barrett's oesophagus in a primary care setting: a multicentre, pragmatic, randomised controlled trial. Lancet 2020; 396:333-344. [PMID: 32738955 PMCID: PMC7408501 DOI: 10.1016/s0140-6736(20)31099-0] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Treatment of dysplastic Barrett's oesophagus prevents progression to adenocarcinoma; however, the optimal diagnostic strategy for Barrett's oesophagus is unclear. The Cytosponge-trefoil factor 3 (TFF3) is a non-endoscopic test for Barrett's oesophagus. The aim of this study was to investigate whether offering this test to patients on medication for gastro-oesophageal reflux would increase the detection of Barrett's oesophagus compared with standard management. METHODS This multicentre, pragmatic, randomised controlled trial was done in 109 socio-demographically diverse general practice clinics in England. Randomisation was done both at the general practice clinic level (cluster randomisation) and at the individual patient level, and the results for each type of randomisation were analysed separately before being combined. Patients were eligible if they were aged 50 years or older, had been taking acid-suppressants for symptoms of gastro-oesophageal reflux for more than 6 months, and had not undergone an endoscopy procedure within the past 5 years. General practice clinics were selected by the local clinical research network and invited to participate in the trial. For cluster randomisation, clinics were randomly assigned (1:1) by the trial statistician using a computer-generated randomisation sequence; for individual patient-level randomisation, patients were randomly assigned (1:1) by the general practice clinics using a centrally prepared computer-generated randomisation sequence. After randomisation, participants received either standard management of gastro-oesophageal reflux (usual care group), in which participants only received an endoscopy if required by their general practitioner, or usual care plus an offer of the Cytosponge-TFF3 procedure, with a subsequent endoscopy if the procedure identified TFF3-positive cells (intervention group). The primary outcome was the diagnosis of Barrett's oesophagus at 12 months after enrolment, expressed as a rate per 1000 person-years, in all participants in the intervention group (regardless of whether they had accepted the offer of the Cytosponge-TFF3 procedure) compared with all participants in the usual care group. Analyses were intention-to-treat. The trial is registered with the ISRCTN registry, ISRCTN68382401, and is completed. FINDINGS Between March 20, 2017, and March 21, 2019, 113 general practice clinics were enrolled, but four clinics dropped out shortly after randomisation. Using an automated search of the electronic prescribing records of the remaining 109 clinics, we identified 13 657 eligible patients who were sent an introductory letter with 14 days to opt out. 13 514 of these patients were randomly assigned (per practice or at the individual patient level) to the usual care group (n=6531) or the intervention group (n=6983). Following randomisation, 149 (2%) of 6983 participants in the intervention group and 143 (2%) of 6531 participants in the usual care group, on further scrutiny, did not meet all eligibility criteria or withdrew from the study. Of the remaining 6834 participants in the intervention group, 2679 (39%) expressed an interest in undergoing the Cytosponge-TFF3 procedure. Of these, 1750 (65%) met all of the eligibility criteria on telephone screening and underwent the procedure. Most of these participants (1654 [95%]; median age 69 years) swallowed the Cytosponge successfully and produced a sample. 231 (3%) of 6834 participants had a positive Cytosponge-TFF3 result and were referred for an endoscopy. Patients who declined the offer of the Cytosponge-TFF3 procedure and all participants in the usual care group only had an endoscopy if deemed necessary by their general practitioner. During an average of 12 months of follow-up, 140 (2%) of 6834 participants in the intervention group and 13 (<1%) of 6388 participants in the usual care group were diagnosed with Barrett's oesophagus (absolute difference 18·3 per 1000 person-years [95% CI 14·8-21·8]; rate ratio adjusted for cluster randomisation 10·6 [95% CI 6·0-18·8], p<0·0001). Nine (<1%) of 6834 participants were diagnosed with dysplastic Barrett's oesophagus (n=4) or stage I oesophago-gastric cancer (n=5) in the intervention group, whereas no participants were diagnosed with dysplastic Barrett's oesophagus or stage I gastro-oesophageal junction cancer in the usual care group. Among 1654 participants in the intervention group who swallowed the Cytosponge device successfully, 221 (13%) underwent endoscopy after testing positive for TFF3 and 131 (8%, corresponding to 59% of those having an endoscopy) were diagnosed with Barrett's oesophagus or cancer. One patient had a detachment of the Cytosponge from the thread requiring endoscopic removal, and the most common side-effect was a sore throat in 63 (4%) of 1654 participants. INTERPRETATION In patients with gastro-oesophageal reflux, the offer of Cytosponge-TFF3 testing results in improved detection of Barrett's oesophagus. Cytosponge-TFF3 testing could also lead to the diagnosis of treatable dysplasia and early cancer. This strategy will lead to additional endoscopies with some false positive results. FUNDING Cancer Research UK, National Institute for Health Research, the UK National Health Service, Medtronic, and the Medical Research Council.
Collapse
Affiliation(s)
- Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK.
| | - Massimiliano di Pietro
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Maria O'Donovan
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Roberta Maroni
- Cancer Research UK and King's College London Cancer Prevention Trials Unit, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Beth Muldrew
- Cancer Research UK and King's College London Cancer Prevention Trials Unit, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Irene Debiram-Beecham
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Marcel Gehrung
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Judith Offman
- Cancer Prevention Group, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Monika Tripathi
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK; Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Samuel G Smith
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Benoit Aigret
- Cancer Research UK and King's College London Cancer Prevention Trials Unit, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Fiona M Walter
- The Primary Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Greg Rubin
- Institute of Population Health Sciences, Newcastle University, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Peter Sasieni
- Cancer Research UK and King's College London Cancer Prevention Trials Unit, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| |
Collapse
|
58
|
Accurate Nonendoscopic Detection of Barrett's Esophagus by Methylated DNA Markers: A Multisite Case Control Study. Am J Gastroenterol 2020; 115:1201-1209. [PMID: 32558685 PMCID: PMC7415629 DOI: 10.14309/ajg.0000000000000656] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Nonendoscopic Barrett's esophagus (BE) screening may help improve esophageal adenocarcinoma outcomes. We previously demonstrated promising accuracy of methylated DNA markers (MDMs) for the nonendoscopic diagnosis of BE using samples obtained from a capsule sponge-on-string (SOS) device. We aimed to assess the accuracy of these MDMs in an independent cohort using a commercial grade assay. METHODS BE cases had ≥ 1 cm of circumferential BE with intestinal metaplasia; controls had no endoscopic evidence of BE. The SOS device was withdrawn 8 minutes after swallowing, followed by endoscopy (the criterion standard). Highest performing MDMs from a previous study were blindly assessed on extracted bisulfite-converted DNA by target enrichment long-probe quantitative amplified signal (TELQAS) assays. Optimal MDM combinations were selected and analyzed using random forest modeling with in silico cross-validation. RESULTS Of 295 patients consented, 268 (91%) swallowed the SOS device; 112 cases and 89 controls met the pre-established inclusion criteria. The median BE length was 6 cm (interquartile range 4-9), and 50% had no dysplasia. The cross-validated sensitivity and specificity of a 5 MDM random forest model were 92% (95% confidence interval 85%-96%) and 94% (95% confidence interval 87%-98%), respectively. Model performance was not affected by age, gender, or smoking history but was influenced by the BE segment length. SOS administration was well tolerated (median [interquartile range] tolerability 2 [0, 4] on 10 scale grading), and 95% preferred SOS over endoscopy. DISCUSSION Using a minimally invasive molecular approach, MDMs assayed from SOS samples show promise as a safe and accurate nonendoscopic test for BE prediction.
Collapse
|
59
|
Krishna Chandar A, Sharma A, Chak A. Novel Screening Alternatives for Barrett Esophagus. Gastroenterol Hepatol (N Y) 2020; 16:238-245. [PMID: 34035726 PMCID: PMC8132638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Barrett esophagus (BE) is the only known premalignant precursor to esophageal adenocarcinoma (EAC), a deadly malignancy that carries a dismal prognosis. Guidelines currently recommend screening for BE only in high-risk populations, such as patients with chronic gastroesophageal reflux disease (GERD) and 1 or more additional risk factors. A GERD-centered approach to BE screening likely leads to a large number of missed EAC cases, as the true population prevalence of BE is thought to be much higher than current estimates. Mass screening for BE has been proposed but is fraught with challenges. Esophagogastroduodenoscopy screening is the current gold standard for BE detection, but it is expensive and cumbersome and carries a small potential for unwanted harms. Transnasal endoscopy is simple, cost-effective, and well tolerated, but it has not found widespread acceptance among physicians and patients. Esophageal capsule endoscopy, despite being well tolerated and accepted, has not been shown to be cost-effective. Newer minimally invasive, nonendoscopic techniques for BE screening have shown promise in prospective clinical trials. Pragmatic head-to-head trials comparing these techniques will help determine the path forward and could herald a new future for population-based BE screening.
Collapse
Affiliation(s)
- Apoorva Krishna Chandar
- Dr Chandar is an internal medicine resident in the Department of Medicine at University Hospitals Cleveland Medical Center in Cleveland, Ohio
- Dr Sharma is a clinical gastroenterology fellow at University Hospitals Cleveland Medical Center
- Dr Chak is the Brenda and Marshall B. Brown Master Clinician in Innovation and Discovery at University Hospitals Cleveland Medical Center and professor of medicine and oncology at Case Western Reserve University School of Medicine in Cleveland, Ohio
| | - Anamay Sharma
- Dr Chandar is an internal medicine resident in the Department of Medicine at University Hospitals Cleveland Medical Center in Cleveland, Ohio
- Dr Sharma is a clinical gastroenterology fellow at University Hospitals Cleveland Medical Center
- Dr Chak is the Brenda and Marshall B. Brown Master Clinician in Innovation and Discovery at University Hospitals Cleveland Medical Center and professor of medicine and oncology at Case Western Reserve University School of Medicine in Cleveland, Ohio
| | - Amitabh Chak
- Dr Chandar is an internal medicine resident in the Department of Medicine at University Hospitals Cleveland Medical Center in Cleveland, Ohio
- Dr Sharma is a clinical gastroenterology fellow at University Hospitals Cleveland Medical Center
- Dr Chak is the Brenda and Marshall B. Brown Master Clinician in Innovation and Discovery at University Hospitals Cleveland Medical Center and professor of medicine and oncology at Case Western Reserve University School of Medicine in Cleveland, Ohio
| |
Collapse
|
60
|
SHAHEEN NICHOLASJ. ENDOSCOPIC TREATMENT OF ESOPHAGEAL NEOPLASIA: A DECADE OF EVOLUTION. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2020; 131:297-314. [PMID: 32675869 PMCID: PMC7358467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Traditional therapy for early esophageal neoplasia has been esophagectomy. In the past decade, the approach to these conditions has rapidly evolved, such that endoscopic therapy has become the primary modality to treat patients with esophageal dysplasia and superficial carcinoma. A variety of modalities are available, including thermal methods, such as radiofrequency ablation and argon plasma coagulation; cryotherapy, including spray liquid nitrogen cryotherapy and balloon-based nitrous oxide cryotherapy; and tissue resection methods, such as endoscopic mucosal resection and endoscopic submucosal dissection. Level 1 evidence substantiates that patients treated with these therapies have a low risk of developing invasive cancer. These treatments demonstrate an excellent safety profile. Future work in this area will define the best modalities of treatment, assess the utility of endoscopic therapy in combination with radiation therapy and chemotherapy, and improve current screening regimens to allow earlier detection of neoplasia.
Collapse
Affiliation(s)
- NICHOLAS J. SHAHEEN
- Correspondence and reprint requests: Nicholas Shaheen, MD, MPH, University of North Carolina School of Medicine, 130 Mason Farm Road, Suite 4150, Chapel Hill, NC 27599-7080919-966-7047
| |
Collapse
|
61
|
Paterson AL, Gehrung M, Fitzgerald RC, O'Donovan M. Role of TFF3 as an adjunct in the diagnosis of Barrett's esophagus using a minimally invasive esophageal sampling device-The Cytosponge TM. Diagn Cytopathol 2019; 48:253-264. [PMID: 31814330 DOI: 10.1002/dc.24354] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/06/2019] [Accepted: 11/06/2019] [Indexed: 12/21/2022]
Abstract
The incidence of esophageal carcinoma continues to increase whilst its prognosis remains poor. The most dramatic reduction in mortality is likely to follow early diagnosis of the preinvasive precursor lesion, Barrett's esophagus (BE), coupled with treatment of dysplastic lesions. The major risk factor for BE is gastroesophageal reflux disease, however this is highly prevalent and only a small proportion of individuals have BE, therefore an endoscopy-based screening strategy to detect BE is unfeasible. Minimally invasive esophageal sampling devices offer an alternative, cost-effective strategy which can be deployed within an at-risk population in a primary care setting to identify individuals with probable BE who can then be referred for endoscopic confirmation. The device that has currently progressed furthest in clinical trials is the CytospongeTM which collects cells from the gastric cardia, gastroesophageal junction and along the whole esophageal length. The cell sample is processed into a formalin-fixed paraffin-embedded block and sections assessed for the presence of intestinal metaplasia. TFF3 immunohistochemistry has consistently been shown to be a valuable adjunct that increases the accuracy of the CytospongeTM test by highlighting early goblet cells which may be missed on morphological assessment and by allowing pseudogoblet cells to be differentiated from true goblet cells.
Collapse
Affiliation(s)
- Anna L Paterson
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Marcel Gehrung
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,MRC Cancer Unit, Hutchison/MRC Research Centre, Cambridge, UK
| | | | - Maria O'Donovan
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.,MRC Cancer Unit, Hutchison/MRC Research Centre, Cambridge, UK
| |
Collapse
|
62
|
Markar SR, Wiggins T, Antonowicz S, Chin ST, Romano A, Nikolic K, Evans B, Cunningham D, Mughal M, Lagergren J, Hanna GB. Assessment of a Noninvasive Exhaled Breath Test for the Diagnosis of Oesophagogastric Cancer. JAMA Oncol 2019; 4:970-976. [PMID: 29799976 PMCID: PMC6145735 DOI: 10.1001/jamaoncol.2018.0991] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Question What is the diagnostic accuracy of a breath test for esophagogastric cancer? Findings In a multicenter diagnostic study of 335 patients, including 172 patients with esophagogastric cancer, the breath test demonstrated good diagnostic accuracy. Meaning This study suggests the potential of breath analysis as a noninvasive tool in the diagnosis of esophagogastric cancer. Importance Early esophagogastric cancer (OGC) stage presents with nonspecific symptoms. Objective The aim of this study was to determine the accuracy of a breath test for the diagnosis of OGC in a multicenter validation study. Design, Setting, and Participants Patient recruitment for this diagnostic validation study was conducted at 3 London hospital sites, with breath samples returned to a central laboratory for selected ion flow tube mass spectrometry (SIFT-MS) analysis. Based on a 1:1 cancer:control ratio, and maintaining a sensitivity and specificity of 80%, the sample size required was 325 patients. All patients with cancer were on a curative treatment pathway, and patients were recruited consecutively. Among the 335 patients included; 172 were in the control group and 163 had OGC. Interventions Breath samples were collected using secure 500-mL steel breath bags and analyzed by SIFT-MS. Quality assurance measures included sampling room air, training all researchers in breath sampling, regular instrument calibration, and unambiguous volatile organic compounds (VOCs) identification by gas chromatography mass spectrometry. Main Outcomes and Measures The risk of cancer was identified based on a previously generated 5-VOCs model and compared with histopathology-proven diagnosis. Results Patients in the OGC group were older (median [IQR] age 68 [60-75] vs 55 [41-69] years) and had a greater proportion of men (134 [82.2%]) vs women (81 [47.4%]) compared with the control group. Of the 163 patients with OGC, 123 (69%) had tumor stage T3/4, and 106 (65%) had nodal metastasis on clinical staging. The predictive probabilities generated by this 5-VOCs diagnostic model were used to generate a receiver operator characteristic curve, with good diagnostic accuracy, area under the curve of 0.85. This translated to a sensitivity of 80% and specificity of 81% for the diagnosis of OGC. Conclusions and Relevance This study shows the potential of breath analysis in noninvasive diagnosis of OGC in the clinical setting. The next step is to establish the diagnostic accuracy of the test among the intended population in primary care where the test will be applied.
Collapse
Affiliation(s)
- Sheraz R Markar
- Department Surgery & Cancer, Imperial College London, United Kingdom
| | - Tom Wiggins
- Department Surgery & Cancer, Imperial College London, United Kingdom
| | - Stefan Antonowicz
- Department Surgery & Cancer, Imperial College London, United Kingdom
| | - Sung-Tong Chin
- Department Surgery & Cancer, Imperial College London, United Kingdom
| | - Andrea Romano
- Department Surgery & Cancer, Imperial College London, United Kingdom
| | - Konstantin Nikolic
- Institute of Biomedical Engineering, Imperial College London, United Kingdom
| | - Benjamin Evans
- Institute of Biomedical Engineering, Imperial College London, United Kingdom
| | - David Cunningham
- Department of Oncology, Royal Marsden Hospital, London, United Kingdom
| | - Muntzer Mughal
- Department of Surgery, University College London Hospital, United Kingdom
| | - Jesper Lagergren
- Department of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| | - George B Hanna
- Department Surgery & Cancer, Imperial College London, United Kingdom
| |
Collapse
|
63
|
Trindade AJ, Navaneethan U, Aslanian HR, Bhutani MS, Krishnan K, Lichtenstein DR, Melson J, Pannala R, Parsi MA, Schulman AR, Sethi A, Trikudanathan G, Watson RR, Maple JT. Advances in the diagnosis and surveillance of Barrett's esophagus (with videos). Gastrointest Endosc 2019; 90:325-334. [PMID: 31113535 DOI: 10.1016/j.gie.2019.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Most patients diagnosed with esophageal adenocarcinoma do not carry a known diagnosis of Barrett's esophagus (BE), suggesting that an improved approach to screening may potentially be of benefit. The use of dysplasia as a biomarker and random biopsy protocols for its detection has limitations. In addition, detecting and appropriately classifying dysplasia in patients with known BE can be difficult. METHODS This document reviews several technologies with a recently established or potential role in the diagnosis and/or surveillance of BE as well as risk stratification for progression to esophageal adenocarcinoma. RESULTS Two technologies were reviewed for imaging or tissue sampling: (1) wide-area transepithelial sampling and (2) volumetric laser endomicroscopy. Four technologies were reviewed for molecular and biomarker technologies for diagnosis and risk stratification: (1) Cytosponge, (2) mutational load, (3) fluorescence in situ hybridization, and (4) immunohistochemistry. CONCLUSION Several technologies discussed in this document may improve dysplasia detection in BE in a wide-field manner. Moreover, the addition of different biomarkers may aid in enhanced risk stratification to optimize approaches to surveillance or treatment for patients with BE.
Collapse
Affiliation(s)
- Arvind J Trindade
- Zucker School of Medicine at Hofstra/Northwell, Northwell Health System, New Hyde Park, New York
| | | | - Harry R Aslanian
- Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Manoop S Bhutani
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Kumar Krishnan
- Division of Gastroenterology, Department of Internal Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - David R Lichtenstein
- Division of Gastroenterology, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Joshua Melson
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Rahul Pannala
- Division of Gastroenterology and Hepatology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Mansour A Parsi
- Department of Gastroenterology and Hepatology, Tulane University, New Orleans, Louisiana
| | - Allison R Schulman
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan
| | - Amrita Sethi
- Division of Digestive and Liver Diseases, Columbia University Medical Center-New York-Presbyterian, New York, New York
| | - Guru Trikudanathan
- Division of Gastroenterology, University of Minnesota, Minneapolis, Minnesota
| | - Rabindra R Watson
- Interventional Endoscopy Services, California Pacific Medical Center, San Francisco, California
| | - John T Maple
- Division of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
64
|
Li S, Chung DC, Mullen JT. Screening high-risk populations for esophageal and gastric cancer. J Surg Oncol 2019; 120:831-846. [PMID: 31373005 DOI: 10.1002/jso.25656] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022]
Abstract
Cancers of the esophagus and stomach remain important causes of mortality worldwide, in large part because they are most often diagnosed at advanced stages. Thus, it is imperative that we identify and treat these cancers in earlier stages. Due to significant heterogeneity in incidence and risk factors for these cancers, it has been challenging to develop standardized screening recommendations. This review summarizes the current recommendations for screening populations at high risk of developing esophagogastric cancers.
Collapse
Affiliation(s)
- Selena Li
- Departments of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Daniel C Chung
- Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - John T Mullen
- Departments of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
65
|
Abstract
PURPOSE OF REVIEW There has been an exponential increase in the incidence of esophageal adenocarcinoma (EAC) over the last half century. Barrett's esophagus (BE) is the only known precursor lesion of EAC. Screening for BE in high-risk populations has been advocated with the aim of identifying BE, followed by endoscopic surveillance to detect dysplasia and early stage cancer, with the intent that treatment can improve outcomes. We aimed to review BE screening methodologies currently recommended and in development. RECENT FINDINGS Unsedated transnasal endoscopy allows for visualization of the distal esophagus, with potential for biopsy acquisition, and can be done in the office setting. Non-endoscopic screening methods being developed couple the use of swallowable esophageal cell sampling devices with BE specific biomarkers, as well as trefoil factor 3, methylated DNA markers, and microRNAs. This approach has promising accuracy. Circulating and exhaled volatile organic compounds and the foregut microbiome are also being explored as means of detecting EAC and BE in a non-invasive manner. Non-invasive diagnostic techniques have shown promise in the detection of BE and may be effective methods of screening high-risk patients.
Collapse
Affiliation(s)
- Don C Codipilly
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Prasad G Iyer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
66
|
Gastrointestinal Endoscopy Editorial Board top 10 topics: advances in GI endoscopy in 2018. Gastrointest Endosc 2019; 90:35-43. [PMID: 30928425 DOI: 10.1016/j.gie.2019.03.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/18/2019] [Indexed: 12/11/2022]
Abstract
The American Society for Gastrointestinal Endoscopy's Gastrointestinal Endoscopy Editorial Board reviewed original endoscopy-related articles published during 2018 in Gastrointestinal Endoscopy and 10 other leading medical and gastroenterology journals. Votes from each individual member were tallied to identify a consensus list of 10 topic areas of major advances in GI endoscopy. Individual board members summarized important findings published in these 10 areas of adenoma detection, bariatric endoscopy, EMR/submucosal dissection/full-thickness resection, artificial intelligence, expandable metal stents for palliation of biliary obstruction, pancreatic therapy with lumen-apposing metal stents, endoscope reprocessing, Barrett's esophagus, interventional EUS, and GI bleeding. This document summarizes these "Top 10" endoscopic advances of 2018.
Collapse
|
67
|
Peters Y, Al-Kaabi A, Shaheen NJ, Chak A, Blum A, Souza RF, Di Pietro M, Iyer PG, Pech O, Fitzgerald RC, Siersema PD. Barrett oesophagus. Nat Rev Dis Primers 2019; 5:35. [PMID: 31123267 DOI: 10.1038/s41572-019-0086-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Barrett oesophagus (BE), the only known histological precursor of oesophageal adenocarcinoma (EAC), is a condition in which the squamous epithelium of the oesophagus is replaced by columnar epithelium as an adaptive response to gastro-oesophageal reflux. EAC has one of the fastest rising incidences of cancers in Western countries and has a dismal prognosis. BE is usually detected during endoscopic examination, and diagnosis is confirmed by the histological presence of intestinal metaplasia. Advances in genomics and transcriptomics have improved our understanding of the pathogenesis and malignant progression of intestinal metaplasia. As the majority of EAC cases are diagnosed in individuals without a known history of BE, screening for BE could potentially decrease disease-related mortality. Owing to the pre-malignant nature of BE, endoscopic surveillance of patients with BE is imperative for early detection and treatment of dysplasia to prevent further progression to invasive EAC. Developments in endoscopic therapy have resulted in a major shift in the treatment of patients with BE who have dysplasia or early EAC, from surgical resection to endoscopic resection and ablation. In addition to symptom control by optimization of lifestyle and pharmacological therapy with proton pump inhibitors, chemopreventive strategies based on NSAIDs and statins are currently being investigated for BE management.
Collapse
Affiliation(s)
- Yonne Peters
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ali Al-Kaabi
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nicholas J Shaheen
- Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amitabh Chak
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew Blum
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Rhonda F Souza
- Department of Medicine and the Center for Esophageal Diseases, Baylor University Medical Center at Dallas and the Center for Esophageal Research, Baylor Scott and White Research Institute, Dallas, TX, USA
| | | | - Prasad G Iyer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Oliver Pech
- Department of Gastroenterology, St John of God Hospital, Regensburg, Germany
| | | | - Peter D Siersema
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, Netherlands.
| |
Collapse
|
68
|
Zhou Z, Kalatskaya I, Russell D, Marcon N, Cirocco M, Krzyzanowski PM, Streutker C, Liang H, Litle VR, Godfrey TE, Stein L. Combined EsophaCap cytology and MUC2 immunohistochemistry for screening of intestinal metaplasia, dysplasia and carcinoma. Clin Exp Gastroenterol 2019; 12:219-229. [PMID: 31190949 PMCID: PMC6527096 DOI: 10.2147/ceg.s186958] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 03/30/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose: The incidence of esophageal adenocarcinoma (EAC) has increased by 700% in Western countries over the last 30 years. Although clinical guidelines call for endoscopic surveillance for EAC among high-risk populations, fewer than 5% of new EAC patients are under surveillance at the time of diagnosis. We studied the accuracy of combined cytopathology and MUC2 immunohistochemistry (IHC) for screening of Intestinal Metaplasia (IM), dysplasia and EAC, using specimens collected from the EsophaCap swallowable encapsulated cytology sponge from Canada and United States. Patients and methods: By comparing the EsophaCap cytological diagnosis with concurrent endoscopic biopsies performed on the same patients in 28 cases, we first built up the cytology diagnostic categories and criteria. Based on these criteria, 136 cases were evaluated by both cytology and MUC2 IHC with blinded to patient biopsy diagnosis. Results: We first set up categories and criteria for cytological diagnosis of EscophaCap samples. Based on these, we divided our evaluated cytological samples into two groups: non-IM group and IM or dysplasia or adenocarcinoma group. Using the biopsy as our gold standard to screen IM, dysplasia and EAC by combined cytology and MUC2 IHC, the sensitivity and specificity were 68% and 91%, respectively, which is in the range of clinically useful cytological screening tests such as the cervical Pap smear. Conclusions: Combined EsophaCap cytology and MUC2 IHC could be a good screening test for IM and Beyond.
Collapse
Affiliation(s)
- Zhongren Zhou
- Department of Pathology & Immunology, Washington University, Saint Louis, MO, USA
| | - Irina Kalatskaya
- Department of Adaptive Oncology, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Donna Russell
- Department of Pathology & Immunology, Washington University, Saint Louis, MO, USA
| | - Norman Marcon
- Division of Gastroenterology, Department of Internal Medicine, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Maria Cirocco
- Division of Gastroenterology, Department of Internal Medicine, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Paul M Krzyzanowski
- Department of Adaptive Oncology, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Cathy Streutker
- Division of Gastroenterology, Department of Internal Medicine, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Hua Liang
- Department of Statistics, George Washington University, Washington, DC, USA
| | - Virginia R Litle
- Department of Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Tony E Godfrey
- Department of Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Lincoln Stein
- Department of Adaptive Oncology, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
69
|
Moinova HR, LaFramboise T, Lutterbaugh JD, Chandar AK, Dumot J, Faulx A, Brock W, De la Cruz Cabrera O, Guda K, Barnholtz-Sloan JS, Iyer PG, Canto MI, Wang JS, Shaheen NJ, Thota PN, Willis JE, Chak A, Markowitz SD. Identifying DNA methylation biomarkers for non-endoscopic detection of Barrett's esophagus. Sci Transl Med 2019; 10:10/424/eaao5848. [PMID: 29343623 DOI: 10.1126/scitranslmed.aao5848] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/08/2017] [Indexed: 12/17/2022]
Abstract
We report a biomarker-based non-endoscopic method for detecting Barrett's esophagus (BE) based on detecting methylated DNAs retrieved via a swallowable balloon-based esophageal sampling device. BE is the precursor of, and a major recognized risk factor for, developing esophageal adenocarcinoma. Endoscopy, the current standard for BE detection, is not cost-effective for population screening. We performed genome-wide screening to ascertain regions targeted for recurrent aberrant cytosine methylation in BE, identifying high-frequency methylation within the CCNA1 locus. We tested CCNA1 DNA methylation as a BE biomarker in cytology brushings of the distal esophagus from 173 individuals with or without BE. CCNA1 DNA methylation demonstrated an area under the curve of 0.95 for discriminating BE-related metaplasia and neoplasia cases versus normal individuals, performing identically to methylation of VIM DNA, an established BE biomarker. When combined, the resulting two biomarker panel was 95% sensitive and 91% specific. These results were replicated in an independent validation cohort of 149 individuals who were assayed using the same cutoff values for test positivity established in the training population. To progress toward non-endoscopic esophageal screening, we engineered a well-tolerated, swallowable, encapsulated balloon device able to selectively sample the distal esophagus within 5 min. In balloon samples from 86 individuals, tests of CCNA1 plus VIM DNA methylation detected BE metaplasia with 90.3% sensitivity and 91.7% specificity. Combining the balloon sampling device with molecular assays of CCNA1 plus VIM DNA methylation enables an efficient, well-tolerated, sensitive, and specific method of screening at-risk populations for BE.
Collapse
Affiliation(s)
- Helen R Moinova
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Thomas LaFramboise
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - James D Lutterbaugh
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Apoorva Krishna Chandar
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - John Dumot
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Ashley Faulx
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Wendy Brock
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | | | - Kishore Guda
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Prasad G Iyer
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Marcia I Canto
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | - Jean S Wang
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nicholas J Shaheen
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Prashanti N Thota
- Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Joseph E Willis
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA. .,Department of Pathology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.,University Hospitals Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Amitabh Chak
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.,University Hospitals Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Sanford D Markowitz
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.,University Hospitals Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
70
|
Patel A, Gyawali CP. Screening for Barrett's Esophagus: Balancing Clinical Value and Cost-effectiveness. J Neurogastroenterol Motil 2019; 25:181-188. [PMID: 30827080 PMCID: PMC6474698 DOI: 10.5056/jnm18156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/25/2018] [Accepted: 12/08/2018] [Indexed: 12/12/2022] Open
Abstract
In predisposed individuals with long standing gastroesophageal reflux disease (GERD), esophageal squamous mucosa can transform into columnar mucosa with intestinal metaplasia, commonly called Barrett’s esophagus (BE). Barrett’s mucosa can develop dysplasia, which can be a precursor for esophageal adenocarcinoma (EAC). However, most EAC cases are identified when esophageal symptoms develop, without prior BE or GERD diagnoses. While several gastrointestinal societies have published BE screening guidelines, these vary, and many recommendations are not based on high quality evidence. These guidelines are concordant in recommending targeted screening of predisposed individuals (eg, long standing GERD symptoms with age > 50 years, male sex, Caucasian race, obesity, and family history of BE or EAC), and against population based screening, or screening of GERD patients without risk factors. Targeted endoscopic screening programs provide earlier diagnosis of high grade dysplasia and EAC, and offer potential for endoscopic therapy, which can improve prognosis and outcome. On the other hand, endoscopic screening of the general population, unselected GERD patients, patients with significant comorbidities or patients with limited life expectancy is not cost-effective. New screening modalities, some of which do not require endoscopy, have the potential to reduce costs and expand access to screening for BE.
Collapse
Affiliation(s)
- Amit Patel
- Division of Gastroenterology, Duke University School of Medicine, and the Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - C Prakash Gyawali
- Division of Gastroenterology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
71
|
Kaz AM, Grady WM. Novel Barrett's esophagus screening assays based on swallowable devices: will they change the game? Transl Gastroenterol Hepatol 2019; 4:25. [PMID: 31143846 PMCID: PMC6509432 DOI: 10.21037/tgh.2019.04.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 11/06/2022] Open
Affiliation(s)
- Andrew M. Kaz
- Gastroenterology Section, VA Puget Sound Health Care System, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - William M. Grady
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
72
|
Abstract
OBJECTIVE To identify patient factors that are associated with emergency presentation of esophageal and gastric cancer, and further to evaluate long-term prognosis in this cohort. BACKGROUND The incidence of emergency presentation is variable, with the prognosis of patients stabilized and discharged to return for elective surgery unknown. METHODS The primary admission of patients with esophageal or gastric cancer within the Hospital Episode Statistics database (1997-2012) was used to classify as emergency or elective diagnosis. Multivariate regression analyses were used to identify patient factors associated with emergency diagnosis and prognosis. RESULTS A total of 35,807 (29.4%) and 45,866 (39.6%) patients with esophageal and gastric cancer presented as an emergency over the study period. Age ≥70, female sex, non-white ethnicity, Charlson comorbidity index score ≥3 and more deprived Townsend index were independent predictors of emergency cancer diagnosis. Emergency diagnosis was an independent predictor of increased 5-year mortality for all patients with esophageal cancer [hazard ratio (HR) = 1.63, 95% confidence interval (CI) 1.61-1.65] and gastric cancer (HR = 1.20, 95% CI 1.16-1.23). Specifically patients receiving surgery on an elective follow-up admission with an initial emergency diagnosis had a poorer prognosis (esophageal cancer: HR = 1.35, 95% CI 1.27-1.44, gastric cancer: HR = 1.13. 95% CI 1.04-1.22), with a significant increase in liver recurrence (esophageal cancer: 7.1% vs 4.9%; P < 0.001, gastric cancer: 7.0% vs 4.8%; P < 0.001) compared to patients referred electively. CONCLUSIONS Emergency presentation of esophageal and gastric cancer is associated with a poor prognosis, due to the increased incidence of metastatic disease at diagnosis and a higher recurrence rate after surgery.
Collapse
|
73
|
Wang Z, Kambhampati Thiruvengadam S, Cheng Y, Ma K, Simsek C, Tieu AH, Abraham JM, Liu X, Prasath V, Duncan M, Stark A, Trick A, Tsai HL, Wang H, He Y, Khashab MA, Ngamruengphong S, Shin EJ, Wang TH, Meltzer SJ. Methylation Biomarker Panel Performance in EsophaCap Cytology Samples for Diagnosing Barrett's Esophagus: A Prospective Validation Study. Clin Cancer Res 2019; 25:2127-2135. [PMID: 30670490 PMCID: PMC6594757 DOI: 10.1158/1078-0432.ccr-18-3696] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/28/2018] [Accepted: 01/17/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE Barrett's esophagus is the only known precursor of esophageal adenocarcinoma (EAC). Although endoscopy and biopsy are standard methods for Barrett's esophagus diagnosis, their high cost and risk limit their use as a screening modality. Here, we sought to develop a Barrett's esophagus detection method based on methylation status in cytology samples captured by EsophaCap using a streamlined sensitive technique, methylation on beads (MOB). EXPERIMENTAL DESIGN We conducted a prospective cohort study on 80 patients (52 in the training set; 28 in the test set). We used MOB to extract and bisulfite-convert DNA, followed by quantitative methylation-specific PCR to assess methylation levels of 8 previously selected candidate markers. Lasso regression was applied to establish a prediction model in the training set, which was then tested on the independent test set. RESULTS In the training set, five of eight candidate methylation biomarkers (p16, HPP1, NELL1, TAC1, and AKAP12) were significantly higher in Barrett's esophagus patients than in controls. We built a four-biomarker-plus-age lasso regression model for Barrett's esophagus diagnosis. The AUC was 0.894, with sensitivity 94.4% [95% confidence interval (CI), 71%-99%] and specificity 62.2% (95% CI, 44.6%-77.3%) in the training set. This model also performed with high accuracy for Barrett's esophagus diagnosis in an independent test set: AUC = 0.929 (P < 0.001; 95% CI, 0.810%-1%), with sensitivity=78.6% (95% CI, 48.8%-94.3%) and specificity = 92.8% (95% CI, 64.1%-99.6%). CONCLUSIONS EsophaCap, in combination with an epigenetic biomarker panel and the MOB method, is a promising, well-tolerated, low-cost esophageal sampling strategy for Barrett's esophagus diagnosis. This approach merits further prospective studies in larger populations.
Collapse
Affiliation(s)
- Zhixiong Wang
- Gastrointestinal Surgical Center, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Center of Gastric Cancer, Sun Yat-Sen University, Guangzhou, China
| | - Swetha Kambhampati Thiruvengadam
- Division of Hematology/Oncology, Department of Medicine, Helen Diller Family Comprehensive Cancer Center, UCSF Medical Center, San Francisco, California
- Department of Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - Yulan Cheng
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ke Ma
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cem Simsek
- Division of Gastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan H Tieu
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John M Abraham
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xi Liu
- Department of Pathology, the First Affiliated Hospital of Xi' an Jiaotong University, Xi' an, Shaanxi, China
| | - Vishnu Prasath
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Mark Duncan
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Alejandro Stark
- Departments of Mechanical Engineering and Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Alexander Trick
- Departments of Mechanical Engineering and Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Hua-Ling Tsai
- Division of Biostatistics, Department of Oncology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hao Wang
- Division of Biostatistics, Department of Oncology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yulong He
- Gastrointestinal Surgical Center, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Center of Gastric Cancer, Sun Yat-Sen University, Guangzhou, China
- Gastrointestinal Surgical Center, Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Mouen A Khashab
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Saowanee Ngamruengphong
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eun J Shin
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tza-Huei Wang
- Department of Medicine, Johns Hopkins Hospital, Baltimore, Maryland
| | - Stephen J Meltzer
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
74
|
Sanghi V, Thota PN. Barrett's esophagus: novel strategies for screening and surveillance. Ther Adv Chronic Dis 2019; 10:2040622319837851. [PMID: 30937155 PMCID: PMC6435879 DOI: 10.1177/2040622319837851] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
Barrett’s esophagus is the precursor lesion for esophageal adenocarcinoma. Screening and surveillance of Barrett’s esophagus are undertaken with the goal of earlier detection and lowering the mortality from esophageal adenocarcinoma. The widely used technique is standard esophagogastroduodenoscopy with biopsies per the Seattle protocol for screening and surveillance of Barrett’s esophagus. Surveillance intervals vary depending on the degree of dysplasia with endoscopic eradication therapy confined to patients with Barrett’s esophagus and confirmed dysplasia. In this review, we present various novel techniques for screening of Barrett’s esophagus such as unsedated transnasal endoscopy, cytosponge with trefoil factor-3, balloon cytology, esophageal capsule endoscopy, liquid biopsy, electronic nose, and oral microbiome. In addition, advanced imaging techniques such as narrow band imaging, dye-based chromoendoscopy, confocal laser endomicroscopy, volumetric laser endomicroscopy, and wide-area transepithelial sampling with computer-assisted three-dimensional analysis developed for better detection of dysplasia are also reviewed.
Collapse
Affiliation(s)
- Vedha Sanghi
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Prashanthi N Thota
- Esophageal Center, Department of Gastroenterology and Hepatology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
75
|
Konda VJA, Souza RF. Barrett's Esophagus and Esophageal Carcinoma: Can Biomarkers Guide Clinical Practice? Curr Gastroenterol Rep 2019; 21:14. [PMID: 30868278 DOI: 10.1007/s11894-019-0685-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
PURPOSE OF REVIEW Despite gastrointestinal societal recommendations for endoscopic screening and surveillance of Barrett's esophagus, the rates of esophageal adenocarcinoma continue to rise. Furthermore, this current practice is costly to patients and the medical system without clear evidence of reduction in cancer mortality. The use of biomarkers to guide screening, surveillance, and treatment strategies might alleviate some of these issues. RECENT FINDINGS Incredible advances in biomarker identification, biomarker assays, and minimally-invasive modalities to acquire biomarkers have shown promising results. We will highlight recently published, key studies demonstrating where we are with using biomarkers for screening and surveillance in clinical practice, and what is on the horizon regarding novel non-invasive and minimally invasive methods to acquire biomarkers. Proof-of principle studies using in silico models demonstrate that biomarker-guided screening, surveillance, and therapeutic intervention strategies can be cost-effective and can reduce cancer deaths in patients with Barrett's esophagus.
Collapse
Affiliation(s)
- Vani J A Konda
- Department of Medicine and the Center for Esophageal Diseases, Baylor University Medical Center, Dallas, TX, 75246, USA
- The Center for Esophageal Research, Baylor Scott and White Research Institute, Baylor University Medical Center, 2 Hoblitzelle, Suite 250, 3500 Gaston Avenue, Dallas, TX, 75246, USA
| | - Rhonda F Souza
- Department of Medicine and the Center for Esophageal Diseases, Baylor University Medical Center, Dallas, TX, 75246, USA.
- The Center for Esophageal Research, Baylor Scott and White Research Institute, Baylor University Medical Center, 2 Hoblitzelle, Suite 250, 3500 Gaston Avenue, Dallas, TX, 75246, USA.
| |
Collapse
|
76
|
Sami SS, Iyer PG, Pophali P, Halland M, di Pietro M, Ortiz-Fernandez-Sordo J, White JR, Johnson M, Guha IN, Fitzgerald RC, Ragunath K. Acceptability, Accuracy, and Safety of Disposable Transnasal Capsule Endoscopy for Barrett's Esophagus Screening. Clin Gastroenterol Hepatol 2019; 17:638-646.e1. [PMID: 30081223 PMCID: PMC6330075 DOI: 10.1016/j.cgh.2018.07.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/03/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Screening for Barrett's esophagus (BE) with conventional esophagogastroduodenoscopy (C-EGD) is expensive. We assessed the performance of a clinic-based, single use transnasal capsule endoscope (EG Scan II) for the detection of BE, compared to C-EGD as the reference standard. METHODS We performed a prospective multicenter cohort study of patients with and without BE recruited from 3 referral centers (1 in the United States and 2 in the United Kingdom). Of 200 consenting participants, 178 (89%) completed both procedures (11% failed EG Scan due to the inability to intubate the nasopharynx). The mean age of participants was 57.9 years and 67% were male. The prevalence of BE was 53%. All subjects underwent the 2 procedures on the same day, performed by blinded endoscopists. Patients completed preference and validated tolerability (10-point visual analogue scale [VAS]) questionnaires within 14 days of the procedures. RESULTS A higher proportion of patients preferred the EG Scan (54.2%) vs the C-EGD (16.7%) (P < .001) and the EG Scan had a higher VAS score (7.2) vs the C-EGD (6.4) (P = .0004). No serious adverse events occurred. The EG Scan identified any length BE with a sensitivity value of 0.90 (95% CI, 0.83-0.96) and a specificity value of 0.91 (95% CI, 0.82-0.96). The EG Scan identified long segment BE with a sensitivity value of 0.95 and short segment BE with a sensitivity values of 0.87. CONCLUSIONS In a prospective study, we found the EG Scan to be safe and to detect BE with higher than 90% sensitivity and specificity. A higher proportion of patients preferred the EG Scan to C-EGD. This device might be used as a clinic-based tool to screen populations at risk for BE. ISRCTN registry identifier: 70595405; ClinicalTrials.gov no: NCT02066233.
Collapse
Affiliation(s)
- Sarmed S Sami
- Barrett's Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota; Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Prasad G Iyer
- Barrett's Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Prachi Pophali
- Barrett's Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Magnus Halland
- Barrett's Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Massimiliano di Pietro
- Cambridge University Hospitals NHS Trust and MRC Cancer Unit, Hutchinson/MRC Research Center, University of Cambridge, Cambridge, United Kingdom
| | - Jacobo Ortiz-Fernandez-Sordo
- National Institute for Health Research Biomedical Research Center in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust and the University of Nottingham, Queens Medical Center Campus, Nottingham, United Kingdom
| | - Jonathan R White
- National Institute for Health Research Biomedical Research Center in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust and the University of Nottingham, Queens Medical Center Campus, Nottingham, United Kingdom
| | - Michele Johnson
- Barrett's Esophagus Unit, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Indra Neil Guha
- National Institute for Health Research Biomedical Research Center in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust and the University of Nottingham, Queens Medical Center Campus, Nottingham, United Kingdom
| | - Rebecca C Fitzgerald
- Cambridge University Hospitals NHS Trust and MRC Cancer Unit, Hutchinson/MRC Research Center, University of Cambridge, Cambridge, United Kingdom
| | - Krish Ragunath
- National Institute for Health Research Biomedical Research Center in Gastrointestinal and Liver Diseases at Nottingham University Hospitals NHS Trust and the University of Nottingham, Queens Medical Center Campus, Nottingham, United Kingdom.
| |
Collapse
|
77
|
Januszewicz W, Tan WK, Lehovsky K, Debiram-Beecham I, Nuckcheddy T, Moist S, Kadri S, di Pietro M, Boussioutas A, Shaheen NJ, Katzka DA, Dellon ES, Fitzgerald RC. Safety and Acceptability of Esophageal Cytosponge Cell Collection Device in a Pooled Analysis of Data From Individual Patients. Clin Gastroenterol Hepatol 2019; 17:647-656.e1. [PMID: 30099104 PMCID: PMC6370042 DOI: 10.1016/j.cgh.2018.07.043] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/10/2018] [Accepted: 07/28/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Diagnosis and surveillance of Barrett's esophagus (BE) and eosinophilic esophagitis (EoE) have become emerging public health issues. Cytosponge is a novel, minimally invasive esophageal cell collection device. We aimed to assess the data on safety and acceptability of this device. METHODS We performed a patient-level review of 5 prospective trials assessing Cytosponge performance in patients with reflux disease, BE and EoE in primary and secondary care. Acceptability of Cytosponge and subsequent endoscopy were recorded with visual analogue scale (VAS), wherein 0 and 10 denoted lowest and highest acceptability. Median VAS scores were compared using a Mann-Whitney test. The number of attempts, failures in swallowing the device and occurrence of adverse events were analyzed. Risk factors for failure in swallowing were analyzed using a multivariate regression model. RESULTS In total, 2672 Cytosponge procedures were performed, in 2418 individuals from 2008 through 2017. There were 2 adverse events related to the device: a minor pharyngeal bleed and a case of detachment (<1:2000). The median acceptability score for the Cytosponge was 6.0 (interquartile range [IQR], 5.0-8.0), which was higher than the score for endoscopy without sedation (median 5.0; IQR, 3.0-7.0; P < .001) and lower than the score for endoscopy with sedation (median 8.0; IQR, 5.0-9.0; P < .001). Nearly all patients (91.1%) successfully swallowed the Cytosponge, most on the first attempt (90.1%). Failure to swallow the device was more likely to occur in secondary care (odds ratio, 5.13; 95% CI, 1.48-17.79; P < .01). CONCLUSIONS The Cytosponge test is a safe procedure with good acceptability ratings in a variety of health care settings.
Collapse
Affiliation(s)
- Wladyslaw Januszewicz
- MRC Cancer Unit, University of Cambridge, Cambridge UK,Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Centre for Postgraduate Education, Warsaw, Poland
| | - Wei Keith Tan
- MRC Cancer Unit, University of Cambridge, Cambridge UK,Department of Gastroenterology, Addenbrookes Hospital, Cambridge University NHS Foundation Trust, Cambridge, UK
| | - Katie Lehovsky
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Susan Moist
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Sudarshan Kadri
- Department of Gastroenterology, University Hospital Leicester, Leicester, UK
| | | | - Alex Boussioutas
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas J. Shaheen
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, USA
| | - David A. Katzka
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Evan S. Dellon
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, USA
| | | | | |
Collapse
|
78
|
Thota PN, Chak A. Is Mass Screening for Barrett's Esophagus a Myth or Reality? Clin Gastroenterol Hepatol 2019; 17:610-612. [PMID: 30267863 PMCID: PMC6445641 DOI: 10.1016/j.cgh.2018.09.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Prashanthi N. Thota
- Department of Gastroenterology and Hepatology, Cleveland
Clinic, Cleveland, OH 44195
| | - Amitabh Chak
- Department of Gastroenterology and Hepatology University
Hospitals Cleveland Medical Center, Case Western Reserve University School of
Medicine, Cleveland, OH, 44118
| |
Collapse
|
79
|
Falk GW. 2017 David Sun Lecture: Screening and Surveillance of Barrett's Esophagus: Where Are We Now and What Does the Future Hold? Am J Gastroenterol 2019; 114:64-70. [PMID: 30361622 DOI: 10.1038/s41395-018-0374-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Barrett's esophagus and esophageal adenocarcinoma continue to present considerable management challenges in the Western world. Despite our best efforts to date, the prognosis of advanced esophageal adenocarcinoma remains poor and far too many individuals with esophageal adenocarcinoma have not had a prior endoscopy to detect Barrett's esophagus. As such, current strategies of screening for Barrett's esophagus and subsequent surveillance need to be further optimized. Screening today is limited to high definition white light endoscopy in high-risk patient populations and as such has multiple limitations. However, a variety of exciting new techniques including risk prediction tools, tethered capsule endomicroscopy, a cytology sponge, breath testing for exhaled volatile organic compounds, and assessment of the oral microbiome are now under study in an effort to develop less expensive population-based screening methods. Similarly, endoscopic surveillance, as currently practiced has a variety of limitations. Inexpensive readily available adjuncts are already available to optimize surveillance including increased inspection time in an effort to detect mucosal or vascular abnormalities, special attention to the right hemisphere of the esophagus, and utilization of narrow band imaging or other electronic chromoendoscopy techniques. To improve endoscopic surveillance, a variety of new paradigms are under study including wide area trans-epithelial sampling, advanced endoscopic imaging, molecular imaging, clinical risk stratification and utilization of biomarkers of increased risk. However, progress will be challenging due to the complexity of esophageal cancer biology and the rarity of progression to cancer among patients with nondysplastic Barrett's epithelium.
Collapse
Affiliation(s)
- Gary W Falk
- Division of Gastroenterology, Department of Medicine Hospital of the University of Pennsylvania, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
80
|
Tan WK, Muldrew B, Khan Z, Fitzgerald RC. A crosssectional analysis of Facebook comments to study public perception of a new diagnostic test called the Cytosponge. Dis Esophagus 2019; 32:5098582. [PMID: 30239646 PMCID: PMC7051844 DOI: 10.1093/dote/doy085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Indexed: 12/11/2022]
Abstract
Social media provides a useful platform for informal discussions about healthcare. Acceptability is key to the uptake of diagnostic devices and this can be difficult to gauge from questionnaires and qualitative studies. The aim of this study is to investigate whether Facebook could be used to gauge public perception toward uptake of a new diagnostic test for Barrett's esophagus called the Cytosponge. We retrospectively reviewed Facebook comments relating to a video on the Cytosponge. We categorized comments into: (1) Positive, (2) Negative, (3) Unknown and (4) Questions. Recurring themes that arose were compared to a qualitative study on the Cytosponge. The video received 22.5 million views and 2837 comments within four months. Of these, 525 comments were positive, 215 were unknown, 179 were negative, 71 were questions, and 1847 were 'Tagged' comments. Among positive comments, recurrent themes were that it was innovative, could lead to early cancer-detection, and more favorable than endoscopy. Among negative comments, a recurring theme was concern about the risk of gagging and vomiting. Among 'questions', a recurring theme was related to the risk of Cytosponge detachment. We compared our analysis to a published qualitative study and found similar themes arose across both studies. Facebook provides a rich source of qualitative data, which could be used to augment studies to gauge public perception toward a new diagnostic test.
Collapse
Affiliation(s)
- Wei Keith Tan
- MRC Cancer Unit, University of Cambridge, Cambridge, UK,Department of Gastroenterology, Addenbrooke’s Hospital, Cambridge, UK
| | - Beth Muldrew
- Barts Clinical Trials Unit, Queen Mary University of London, Wolfson Institute of Preventive Medicine, London, UK
| | - Zohrah Khan
- Barts Clinical Trials Unit, Queen Mary University of London, Wolfson Institute of Preventive Medicine, London, UK
| | | |
Collapse
|
81
|
Abstract
In Western countries, the incidence of esophageal adenocarcinoma has increased rapidly in parallel with its premalignant condition, Barrett esophagus (BE). Unlike colonoscopy, endoscopic screening for BE is not currently recommended for all patients; however, surveillance endoscopy is advocated for patients with established BE. Novel imaging and sampling techniques have been developed and investigated for the purpose of improving the detection of Barrett esophagus, dysplasia, and neoplasia. This article discusses several screening and surveillance techniques, including Seattle protocol, chromoendoscopy, electronic chromoendoscopy, wide area transepithelial sampling with 3-dimensional analysis, nonendoscopic sampling devices, and transnasal endoscopy.
Collapse
Affiliation(s)
- Yoshihiro Komatsu
- Esophageal and Lung Institute, Allegheny Health Network, Western Pennsylvania Hospital, Suite 158, Mellon Pavilion, 4815 Liberty Avenue, Pittsburgh, PA 15224, USA
| | - Kirsten M Newhams
- Esophageal and Lung Institute, Allegheny Health Network, Western Pennsylvania Hospital, Suite 158, Mellon Pavilion, 4815 Liberty Avenue, Pittsburgh, PA 15224, USA
| | - Blair A Jobe
- Esophageal and Lung Institute, Allegheny Health Network, Western Pennsylvania Hospital, Suite 158, Mellon Pavilion, 4815 Liberty Avenue, Pittsburgh, PA 15224, USA.
| |
Collapse
|
82
|
Safety and efficacy of a minimally invasive cell sampling device ('Cytosponge') in the diagnosis of esophageal pathology: a systematic review. Eur J Gastroenterol Hepatol 2018; 30:1261-1269. [PMID: 30044236 DOI: 10.1097/meg.0000000000001210] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Esophageal adenocarcinoma is an increasingly common cause of morbidity and mortality in developed countries. Most cases are considered the consequence of chronic gastroesophageal reflux disease, with subsequent Barrett's metaplasia and dysplasia. Because progression from Barrett's metaplasia to cancer occurs over many years, endoscopic screening and surveillance programs have been established, albeit with little or no consideration for cost-effectiveness. As an alternative to the expensive and resource-demanding endoscopic surveillance, the Cytosponge has been developed to sample the esophageal mucosa efficiently. The device is a compressed mesh sponge encapsulated in an ingestible gelatin pill attached to a string. After swallowing, the capsule dissolves allowing the sponge to expand in the stomach. As it is pulled out, cells are collected from the esophagogastric junction and throughout the esophagus. The cellular samples can be analyzed by cytology, immunohistochemistry, and molecular markers. We conducted a systematic review of all recent relevant studies to help define the role of this novel technology, including studies of screening and surveillance of Barrett's esophagus, esophageal squamous dysplasia detection, detection of eosinophilic esophagitis, and evaluation of benign esophageal diseases. With the major limitation that most studies were performed by a single investigative group that developed the technology, the device yielded overall impressive results against the endoscopy/biopsy gold standard. Patient acceptability was high. If these promising early results are validated by other investigators in other populations, the Cytosponge represents an important new advance in the detection of esophageal pathology that could potentially decrease the burden of endoscopic esophageal sampling.
Collapse
|
83
|
Chettouh H, Mowforth O, Galeano-Dalmau N, Bezawada N, Ross-Innes C, MacRae S, Debiram-Beecham I, O’Donovan M, Fitzgerald RC. Methylation panel is a diagnostic biomarker for Barrett's oesophagus in endoscopic biopsies and non-endoscopic cytology specimens. Gut 2018; 67:1942-1949. [PMID: 29084829 PMCID: PMC6176521 DOI: 10.1136/gutjnl-2017-314026] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Barrett's oesophagus is a premalignant condition that occurs in the context of gastro-oesophageal reflux. However, most Barrett's cases are undiagnosed because of reliance on endoscopy. We have developed a non-endoscopic tool: the Cytosponge, which when combined with trefoil factor 3 immunohistochemistry, can diagnose Barrett's oesophagus. We investigated whether a quantitative methylation test that is not reliant on histopathological analysis could be used to diagnose Barrett's oesophagus. DESIGN Differentially methylated genes between Barrett's and normal squamous oesophageal biopsies were identified from whole methylome data and confirmed using MethyLight PCR in biopsy samples of squamous oesophagus, gastric cardia and Barrett's oesophagus. Selected genes were then tested on Cytosponge BEST2 trial samples comprising a pilot cohort (n=20 cases, n=10 controls) and a validation cohort (n=149 cases, n=129 controls). RESULTS Eighteen genes were differentially methylated in patients with Barrett'soesophagus compared with squamous controls. Hypermethylation of TFPI2, TWIST1, ZNF345 and ZNF569 was confirmed in Barrett's biopsies compared with biopsies from squamous oesophagus and gastric cardia (p<0.05). When tested in Cytosponge samples, these four genes were hypermethylated in patients with Barrett's oesophagus compared with patients with reflux symptoms (p<0.001). The optimum biomarker to diagnose Barrett's oesophagus was TFPI2 with a sensitivity and specificity of 82.2% and 95.7%, respectively. CONCLUSION TFPI2, TWIST1, ZNF345 and ZNF569 methylation have promise as diagnostic biomarkers for Barrett's oesophagus when used in combination with a simple and cost effective non-endoscopic cell collection device.
Collapse
Affiliation(s)
- Hamza Chettouh
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Oliver Mowforth
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Núria Galeano-Dalmau
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Navya Bezawada
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Caryn Ross-Innes
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Shona MacRae
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Irene Debiram-Beecham
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Maria O’Donovan
- Department of Histopathology, Addenbrooke’s Hospital, Cambridge, UK
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
84
|
Li X, Kleeman S, Coburn SB, Fumagalli C, Perner J, Jammula S, Pfeiffer RM, Orzolek L, Hao H, Taylor PR, Miremadi A, Galeano-Dalmau N, Lao-Sirieix P, Tennyson M, MacRae S, Cook MB, Fitzgerald RC. Selection and Application of Tissue microRNAs for Nonendoscopic Diagnosis of Barrett's Esophagus. Gastroenterology 2018; 155:771-783.e3. [PMID: 29906417 PMCID: PMC6120784 DOI: 10.1053/j.gastro.2018.05.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/27/2018] [Accepted: 05/31/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS MicroRNA (miRNA) is highly stable in biospecimens and provides tissue-specific profiles, making it a useful biomarker of carcinogenesis. We aimed to discover a set of miRNAs that could accurately discriminate Barrett's esophagus (BE) from normal esophageal tissue and to test its diagnostic accuracy when applied to samples collected by a noninvasive esophageal cell sampling device. METHODS We analyzed miRNA expression profiles of 2 independent sets of esophageal biopsy tissues collected during endoscopy from 38 patients with BE and 26 patients with normal esophagus (controls) using Agilent microarray and Nanostring nCounter assays. Consistently up-regulated miRNAs were quantified by real-time polymerase chain reaction in esophageal tissues collected by Cytosponge from patients with BE vs without BE. miRNAs were expressed from plasmids and antisense oligonucleotides were expressed in normal esophageal squamous cells; effects on proliferation and gene expression patterns were analyzed. RESULTS We identified 15 miRNAs that were significantly up-regulated in BE vs control tissues. Of these, 11 (MIR215, MIR194, MIR 192, MIR196a, MIR199b, MIR10a, MIR145, MIR181a, MIR30a, MIR7, and MIR199a) were validated in Cytosponge samples. The miRNAs with the greatest increases in BE tissues (7.9-fold increase in expression or more, P < .0001: MIR196a, MIR192, MIR194, and MIR215) each identified BE vs control tissues with area under the curve (AUC) values of 0.82 or more. We developed an optimized multivariable logistic regression model, based on expression levels of 6 miRNAs (MIR7, MIR30a, MIR181a, MIR192, MIR196a, and MIR199a), that identified patients with BE with an AUC value of 0.89, 86.2% sensitivity, and 91.6% specificity. Expression level of MIR192, MIR196a, MIR199a, combined that of trefoil factor 3, identified patients with BE with an AUC of 0.93, 93.1% sensitivity, and 93.7% specificity. Hypomethylation was observed in the promoter region of the highly up-regulated cluster MIR192-MIR194. Overexpression of these miRNAs in normal esophageal squamous cells increased their proliferation, via GRHL3 and PTEN signaling. CONCLUSIONS In analyses of miRNA expression patterns of BE vs non-BE tissues, we identified a profile that can identify Cytosponge samples from patients with BE with an AUC of 0.93. Expression of MIR194 is increased in BE samples via epigenetic mechanisms that might be involved in BE pathogenesis.
Collapse
Affiliation(s)
- Xiaodun Li
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Sam Kleeman
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Sally B. Coburn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, Maryland
| | - Carlo Fumagalli
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Juliane Perner
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Sriganesh Jammula
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Ruth M. Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, Maryland
| | - Linda Orzolek
- Johns Hopkins Medical Institutions Deep Sequencing and Microarray Core, Baltimore, Maryland
| | - Haiping Hao
- Johns Hopkins Medical Institutions Deep Sequencing and Microarray Core, Baltimore, Maryland
| | - Philip R. Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, Maryland
| | | | - Núria Galeano-Dalmau
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Pierre Lao-Sirieix
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Maria Tennyson
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Shona MacRae
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Michael B. Cook
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, Maryland,Reprint requests Address requests for reprints to: Michael B. Cook, PhD, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD
| | - Rebecca C. Fitzgerald
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK,Rebecca C. Fitzgerald, MD, MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Box 197, Cambridge Biomedical Campus, Cambridge, UK CB2 0XZ.
| |
Collapse
|
85
|
Offman J, Muldrew B, O’Donovan M, Debiram-Beecham I, Pesola F, Kaimi I, Smith SG, Wilson A, Khan Z, Lao-Sirieix P, Aigret B, Walter FM, Rubin G, Morris S, Jackson C, Sasieni P, Fitzgerald RC, on behalf of the BEST3 Trial team. Barrett's oESophagus trial 3 (BEST3): study protocol for a randomised controlled trial comparing the Cytosponge-TFF3 test with usual care to facilitate the diagnosis of oesophageal pre-cancer in primary care patients with chronic acid reflux. BMC Cancer 2018; 18:784. [PMID: 30075763 PMCID: PMC6091067 DOI: 10.1186/s12885-018-4664-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/10/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Early detection of oesophageal cancer improves outcomes; however, the optimal strategy for identifying patients at increased risk from the pre-cancerous lesion Barrett's oesophagus (BE) is not clear. The Cytosponge, a novel non-endoscopic sponge device, combined with the biomarker Trefoil Factor 3 (TFF3) has been tested in four clinical studies. It was found to be safe, accurate and acceptable to patients. The aim of the BEST3 trial is to evaluate if the offer of a Cytosponge-TFF3 test in primary care for patients on long term acid suppressants leads to an increase in the number of patients diagnosed with BE. METHODS The BEST3 trial is a pragmatic multi-site cluster-randomised controlled trial set in primary care in England. Approximately 120 practices will be randomised 1:1 to either the intervention arm, invitation to a Cytosponge-TFF3 test, or the control arm usual care. Inclusion criteria are men and women aged 50 or over with records of at least 6 months of prescriptions for acid-suppressants in the last year. Patients in the intervention arm will receive an invitation to have a Cytosponge-TFF3 test in their general practice. Patients with a positive TFF3 test will receive an invitation for an upper gastro-intestinal endoscopy at their local hospital-based endoscopy clinic to test for BE. The primary objective is to compare histologically confirmed BE diagnosis between the intervention and control arms to determine whether the offer of the Cytosponge-TFF3 test in primary care results in an increase in BE diagnosis within 12 months of study entry. DISCUSSION The BEST3 trial is a well-powered pragmatic trial testing the use of the Cytosponge-TFF3 test in the same population that we envisage it being used in clinical practice. The data generated from this trial will enable NICE and other clinical bodies to decide whether this test is suitable for routine clinical use. TRIAL REGISTRATION This trial was prospectively registered with the ISRCTN Registry on 19/01/2017, trial number ISRCTN68382401 .
Collapse
Affiliation(s)
- Judith Offman
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
| | - Beth Muldrew
- Cancer Prevention Trials Unit, Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Maria O’Donovan
- Department of Histopathology, Addenbrooke’s Hospital, Cambridge, UK
| | - Irene Debiram-Beecham
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Francesca Pesola
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
| | - Irene Kaimi
- Cancer Prevention Trials Unit, Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Samuel G. Smith
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Ashley Wilson
- Cancer Prevention Trials Unit, Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Zohrah Khan
- Cancer Prevention Trials Unit, Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | | | - Benoit Aigret
- Cancer Prevention Trials Unit, Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Fiona M. Walter
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Greg Rubin
- Institute of Health and Society, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle University, Newcastle upon Tyne, UK
| | - Steve Morris
- Department of Applied Health Research, University College London, London, UK
| | | | - Peter Sasieni
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
- Cancer Prevention Trials Unit, Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Rebecca C. Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - on behalf of the BEST3 Trial team
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
- Cancer Prevention Trials Unit, Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
- Department of Histopathology, Addenbrooke’s Hospital, Cambridge, UK
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
- Astra Zeneca, Cambridge, UK
- The Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Institute of Health and Society, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle University, Newcastle upon Tyne, UK
- Department of Applied Health Research, University College London, London, UK
- MRC Biostatistic Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
86
|
Iyer PG, Taylor WR, Johnson ML, Lansing RL, Maixner KA, Yab TC, Simonson JA, Devens ME, Slettedahl SW, Mahoney DW, Berger CK, Foote PH, Smyrk TC, Wang KK, Wolfsen HC, Ahlquist DA. Highly Discriminant Methylated DNA Markers for the Non-endoscopic Detection of Barrett's Esophagus. Am J Gastroenterol 2018; 113:1156-1166. [PMID: 29891853 DOI: 10.1038/s41395-018-0107-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/04/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Minimally invasive methods have been described to detect Barrett's esophagus (BE), but are limited by subjectivity and suboptimal accuracy. We identified methylated DNA markers (MDMs) for BE in tissue and assessed their accuracy on whole esophagus brushings and capsule sponge samples. METHODS Step 1: Unbiased whole methylome sequencing was performed on DNA from BE and normal squamous esophagus (SE) tissue. Discriminant MDM candidates were validated on an independent patient cohort (62 BE cases, 30 controls) by quantitative methylation specific PCR (qMSP). Step 2: Selected MDMs were further evaluated on whole esophageal brushings (49 BE cases, 36 controls). 35 previously sequenced esophageal adenocarcinoma (EAC) MDMs were also evaluated. Step 3: 20 BE cases and 20 controls were randomized to swallow capsules sponges (25 mm, 10 pores or 20 pores per inch (ppi)) followed endoscopy. DNA yield, tolerability, and mucosal injury were compared. Best MDM assays were performed on this cohort. RESULTS Step 1: 19 MDMs with areas under the ROC curve (AUCs) >0.85 were carried forward. Step 2: On whole esophageal brushings, 80% of individual MDM candidates showed high accuracy for BE (AUCs 0.84-0.94). Step 3: The capsule sponge was swallowed and withdrawn in 98% of subjects. Tolerability was superior with the 10 ppi sponge with minimal mucosal injury and abundant DNA yield. A 2-marker panel (VAV3 + ZNF682) yielded excellent BE discrimination (AUC = 1). CONCLUSIONS Identified MDMs discriminate BE with high accuracy. BE detection appears safe and feasible with a capsule sponge. Corroboration in larger studies is warranted. ClinicalTrials.gov number NCT02560623.
Collapse
Affiliation(s)
- Prasad G Iyer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - William R Taylor
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Michele L Johnson
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Ramona L Lansing
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Kristyn A Maixner
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Tracy C Yab
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Julie A Simonson
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Mary E Devens
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Seth W Slettedahl
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Douglas W Mahoney
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Calise K Berger
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Patrick H Foote
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Thomas C Smyrk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Kenneth K Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Herbert C Wolfsen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - David A Ahlquist
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA. Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA. Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA. Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
87
|
Abstract
New improved methods are required for the early detection of esophageal adenocarcinoma in order to reduce mortality from this aggressive cancer. In this review we discuss different screening methods which are currently under evaluation ranging from image-based methods to cell collection devices coupled with biomarkers. As Barrett's esophagus is a low prevalence disease, potential screening tests must be applied to an enriched population to reduce the false-positive rate and improve the cost-effectiveness of the program.
Collapse
Affiliation(s)
- Maria O'Donovan
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Hills Road, Cambridge, CB2 0XZ, UK
- Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Hills Road, Cambridge, CB2 0XZ, UK.
| |
Collapse
|
88
|
Abstract
The cost-effectiveness of screening and surveillance for Barrett's esophagus continues to evolve as the incidence of esophageal adenocarcinoma increases, biomarkers enhance the identification of individuals at highest risk for developing cancer, and endoscopic eradication of Barrett's esophagus improves. Screening to detect Barrett's esophagus may be cost-effective in selected high-risk groups based on age, race, sex and other factors such as symptoms of heartburn. Currently, endoscopic eradication therapy for Barrett's esophagus and high-grade dysplasia is a cost-effective intervention, while endoscopic therapy for non-dysplastic Barrett's esophagus is not a cost-effective strategy. As diagnosis of low-grade dysplasia improves, endoscopic eradication therapy may also prove to be a cost-effective intervention.
Collapse
Affiliation(s)
- John M Inadomi
- Division of Gastroenterology, Department of Medicine, University of Washington School of Medicine, 1959 NE Pacific Street, Box 356424, Seattle, WA, 98195, USA.
| | - Nina Saxena
- Division of Gastroenterology, Department of Medicine, University of Washington School of Medicine, 1959 NE Pacific Street, Box 356424, Seattle, WA, 98195, USA
| |
Collapse
|
89
|
Abstract
The currently recommended approach to managing cancer risk for patients with Barrett's esophagus is endoscopic surveillance including a biopsy protocol to sample the esophageal tissue randomly to detect dysplasia. However, there are numerous limitations in this practice that rely on the histopathological grading of dysplasia alone to make clinical decisions. The availability of in silico models demonstrating the potential cost-effectiveness of biomarker-based stratification has increased interest in finding a clinically relevant "Barrett's biomarker." The success of endoscopic eradication therapy in preventing neoplastic progression of dysplastic Barrett's esophagus has promoted the desire to stratify non-dysplastic Barrett's esophagus to those with "high risk" that may benefit from endotherapy. Furthermore, on the other end of the spectrum, there is interest in searching for a "low risk" marker that may identify those that would not likely benefit from endoscopy screening or surveillance. This review highlights recent data from the genomics (r)evolution revealing new genetic biomarkers of susceptibility to the development of Barrett's esophagus and novel pathways for its neoplastic progression, addresses the development of new modes of tissue sampling and imaging to detect early neoplasia in Barrett's esophagus, and discusses current progress in moving biomarkers from the laboratory into clinical practice in the era of precision medicine.
Collapse
|
90
|
|
91
|
Spechler SJ, Katzka DA, Fitzgerald RC. New Screening Techniques in Barrett's Esophagus: Great Ideas or Great Practice? Gastroenterology 2018; 154:1594-1601. [PMID: 29577931 DOI: 10.1053/j.gastro.2018.03.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Stuart J Spechler
- Divisions of Gastroenterology and Hepatology, Baylor University Medical Center at Dallas, Dallas, Texas
| | | | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchinson/MRC Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
92
|
Screening patients for Barrett esophagus: Why, who, and how. TECHNIQUES IN GASTROINTESTINAL ENDOSCOPY 2018. [DOI: 10.1016/j.tgie.2018.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
93
|
Ireland CJ, Gordon AL, Thompson SK, Watson DI, Whiteman DC, Reed RL, Esterman A. Validation of a risk prediction model for Barrett's esophagus in an Australian population. Clin Exp Gastroenterol 2018; 11:135-142. [PMID: 29628770 PMCID: PMC5878665 DOI: 10.2147/ceg.s158627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Esophageal adenocarcinoma is a disease that has a high mortality rate, the only known precursor being Barrett's esophagus (BE). While screening for BE is not cost-effective at the population level, targeted screening might be beneficial. We have developed a risk prediction model to identify people with BE, and here we present the external validation of this model. MATERIALS AND METHODS A cohort study was undertaken to validate a risk prediction model for BE. Individuals with endoscopy and histopathology proven BE completed a questionnaire containing variables previously identified as risk factors for this condition. Their responses were combined with data from a population sample for analysis. Risk scores were derived for each participant. Overall performance of the risk prediction model in terms of calibration and discrimination was assessed. RESULTS Scores from 95 individuals with BE and 636 individuals from the general population were analyzed. The Brier score was 0.118, suggesting reasonable overall performance. The area under the receiver operating characteristic was 0.83 (95% CI 0.78-0.87). The Hosmer-Lemeshow statistic was p=0.14. Minimizing false positives and false negatives, the model achieved a sensitivity of 74% and a specificity of 73%. CONCLUSION This study has validated a risk prediction model for BE that has a higher sensitivity than previous models.
Collapse
Affiliation(s)
- Colin J Ireland
- School of Nursing and Midwifery, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Andrea L Gordon
- School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sarah K Thompson
- Discipline of Surgery, University of Adelaide, Adelaide, SA, Australia
| | - David I Watson
- Department of Surgery, Flinders University, Bedford Park, SA, Australia
| | - David C Whiteman
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Richard L Reed
- Discipline of General Practice, Flinders University, Bedford Park, SA, Australia
| | - Adrian Esterman
- School of Nursing and Midwifery, Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
94
|
Diem M, Ergin A, Remiszewski S, Mu X, Akalin A, Raz D. Infrared micro-spectroscopy of human tissue: principles and future promises. Faraday Discuss 2018; 187:9-42. [PMID: 27075634 DOI: 10.1039/c6fd00023a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This article summarizes the methods employed, and the progress achieved over the past two decades in applying vibrational (Raman and IR) micro-spectroscopy to problems of medical diagnostics and cellular biology. During this time, several research groups have verified the enormous information contained in vibrational spectra; in fact, information on protein, lipid and metabolic composition of cells and tissues can be deduced by decoding the observed vibrational spectra. This decoding process is aided by the availability of computer workstations and advanced algorithms for data analysis. Furthermore, commercial instrumentation for the fast collection of both Raman and infrared micro-spectral data has enabled the collection of images of cells and tissues based solely on vibrational spectroscopic data. The progress in the field has been manifested by a steady increase in the number and quality of publications submitted by established and new research groups in vibrational spectroscopy in the biological and biomedical arenas.
Collapse
Affiliation(s)
- Max Diem
- Laboratory for Spectral Diagnosis (LSpD), Department of Chemistry and Chemical Biology, Northeastern University, 316 Hurtig Hall, 360 Huntington Ave, Boston, MA, USA. and Cireca Theranostics, LLC, 19 Blackstone St, Cambridge, MA, USA
| | - Ayşegül Ergin
- Cireca Theranostics, LLC, 19 Blackstone St, Cambridge, MA, USA
| | | | - Xinying Mu
- Cireca Theranostics, LLC, 19 Blackstone St, Cambridge, MA, USA and Department of Mathematics and Statistics and Program in Bioinformatics, Boston University, Boston, MA, USA
| | - Ali Akalin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dan Raz
- Division of Thoracic Surgery, City of Hope Medical Center, Duarte, CA, USA
| |
Collapse
|
95
|
Rubin G, Walter F, Emery J, de Wit N. Reimagining the diagnostic pathway for gastrointestinal cancer. Nat Rev Gastroenterol Hepatol 2018; 15:181-188. [PMID: 29410534 DOI: 10.1038/nrgastro.2018.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A crisis is looming for the diagnosis of gastrointestinal cancers, one grounded only partly in the steady increase in their overall incidence. Public demand for diagnostic tests to be undertaken early and at lower levels of risk is reflected in early diagnosis being a widely held policy objective for reasons of both clinical outcome and patient experience. In the UK, urgent referrals for suspected lower gastrointestinal cancer have increased by 78% in the past 6 years, with parallel increases in endoscopy and imaging activity. Such growth in demand is unsustainable with current models of care. If gastrointestinal cancer diagnosis is to be affordable, the roles of professionals and their interactions with each other will need to be reframed while retaining public confidence in the process. In this Perspective, we consider how the relationship between medical specialists and generalists could be redefined to make better use of the skills of each while delivering optimal clinical outcomes and a good patient experience.
Collapse
Affiliation(s)
- Greg Rubin
- Institute of Health and Society, Newcastle University, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle upon Tyne, NE1 4LP, UK
| | - Fiona Walter
- Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Jon Emery
- Centre for Cancer Research and Department of General Practice, University of Melbourne, Victoria Comprehensive Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3010, Australia
| | - Niek de Wit
- Julius Center for Health Sciences and Primary Care University Medical Center, Utrecht, Netherlands
| |
Collapse
|
96
|
Abstract
BACKGROUND Gastroesophageal reflux disease (GERD) affects an estimated 20% of the population in the USA, and its prevalence is increasing worldwide. About 10-15% of patients with GERD will develop Barrett's esophagus (BE). AIMS The aims of this study were to review the available evidence of the pathophysiology of BE and the role of anti-reflux surgery in the treatment of this disease. RESULTS The transformation of the squamous epithelium into columnar epithelium with goblet cells is due to the chronic injury produced by repeated reflux episodes. It involves genetic mutations that in some patients may lead to high-grade dysplasia and cancer. There is no strong evidence that anti-reflux surgery is associated with resolution or improvement in BE, and its indications should be the same as for other GERD patients without BE. CONCLUSIONS Patients with BE without dysplasia require endoscopic surveillance, while those with low- or high-grade dysplasia should have consideration of endoscopic eradication therapy followed by surveillance. New endoscopic treatment modalities are being developed, which hold the promise to improve the management of patients with BE.
Collapse
|
97
|
Old O, Lloyd G, Isabelle M, Almond LM, Kendall C, Baxter K, Shepherd N, Shore A, Stone N, Barr H. Automated cytological detection of Barrett's neoplasia with infrared spectroscopy. J Gastroenterol 2018; 53:227-235. [PMID: 28501919 DOI: 10.1007/s00535-017-1344-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 04/17/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND Development of a nonendoscopic test for Barrett's esophagus would revolutionize population screening and surveillance for patients with Barrett's esophagus. Swallowed cell collection devices have recently been developed to obtain cytology brushings from the esophagus: automated detection of neoplasia in such samples would enable large-scale screening and surveillance. METHODS Fourier transform infrared (FTIR) spectroscopy was used to develop an automated tool for detection of Barrett's esophagus and Barrett's neoplasia in esophageal cell samples. Cytology brushings were collected at endoscopy, cytospun onto slides and FTIR images were measured. An automated cell recognition program was developed to identify individual cells on the slide. RESULTS Cytology review and contemporaneous histology was used to inform a training dataset containing 141 cells from 17 patients. A classification model was constructed by principal component analysis fed linear discriminant analysis, then tested by leave-one-sample-out cross validation. With application of this training model to whole slide samples, a threshold voting system was used to classify samples according to their constituent cells. Across the entire dataset of 115 FTIR maps from 66 patients, whole samples were classified with sensitivity and specificity respectively as follows: normal squamous cells 79.0% and 81.1%, nondysplastic Barrett's esophagus cells 31.3% and 100%, and neoplastic Barrett's esophagus cells 83.3% and 62.7%. CONCLUSIONS Analysis of esophageal cell samples can be performed with FTIR spectroscopy with reasonable sensitivity for Barrett's neoplasia, but with poor specificity with the current technique.
Collapse
Affiliation(s)
- Oliver Old
- Biophotonics Research Unit, Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK. .,University of Exeter Medical School, Royal Devon and Exeter NHS Foundation Trust, Exeter, EX2 5DW, UK.
| | - Gavin Lloyd
- Biophotonics Research Unit, Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK.,School of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Martin Isabelle
- Biophotonics Research Unit, Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK
| | - L Max Almond
- Biophotonics Research Unit, Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK.,Heartlands Hospital, Bordesley Green East, Birmingham, B9 5SS, UK
| | - Catherine Kendall
- Biophotonics Research Unit, Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK.,School of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Karol Baxter
- Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK
| | - Neil Shepherd
- Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK
| | - Angela Shore
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Royal Devon and Exeter NHS Foundation Trust, Exeter, EX2 5DW, UK
| | - Nick Stone
- School of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Hugh Barr
- Biophotonics Research Unit, Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK.,Gloucestershire Hospitals NHS Foundation Trust, Great Western Road, Gloucester, GL1 3NN, UK
| |
Collapse
|
98
|
Abstract
OPINION STATEMENT PURPOSE OF REVIEW: There is a pressing need for effective strategies to halt the increase in both the incidence and mortality of esophageal adenocarcinoma (EAC). Screening for Barrett's esophagus, which is the only known precursor of EAC, remains a ripe area for research, particularly with regard to identifying the target population, screening tools, and management of screen-detected populations. This review aims to explore in depth the rationale for screening for Barrett's esophagus, recent biotechnological advances which may have the potential of making screening feasible, and also highlight the challenges which will have to be overcome in order make screening for BE a realistic prospect. RECENT FINDINGS Imaging techniques such as portable transnasal endoscopy have the advantage of providing an immediate diagnosis of Barrett's esophagus as well as other significant pathologies such as reflux esophagitis and cancer; however, larger studies in non-enriched community screening populations are required to evaluate their feasibility. The capsule sponge is a cell-sampling device coupled with a biomarker, which has been most extensively evaluated with very promising results as regards feasibility, acceptability, accuracy, and cost-effectiveness. Its effectiveness in increasing the detection of Barrett's esophagus in primary care is currently being evaluated. Several Barrett's esophagus risk prediction scores have been developed with variable degrees of accuracy. Several minimally and non-invasive screening techniques have been studied including imaging and cell-sampling devices. Barrett's risk assessment models need to be further validated in independent, relevant screening populations with clear cut-offs for recommending screening to be defined.
Collapse
Affiliation(s)
- Sarmed S Sami
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Prasad G Iyer
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
99
|
Straumann A, Katzka DA. Diagnosis and Treatment of Eosinophilic Esophagitis. Gastroenterology 2018; 154:346-359. [PMID: 28756235 DOI: 10.1053/j.gastro.2017.05.066] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/16/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
Eosinophilic esophagitis (EoE) is a new disease. It is caused by a T-helper type 2 cell response to food antigens in contact with the esophageal mucosa. Although no single feature defines EoE, a constellation of compatible demographic, clinical, endoscopic, and histologic findings establish the diagnosis. Children present with symptoms and endoscopic patterns characteristic of inflammation, whereas adolescents and adults have manifestations of fibrosis and gross esophageal strictures. Clinical and endoscopic scoring systems have helped to standardize diagnosis. There is controversy in EoE research over the optimal endpoint for treatment. Although the most common endpoint is a reduced number of eosinophils in biopsies, changes in symptoms and endoscopic features are becoming important targets of therapy. We should improve our understanding of EoE progression and the need for maintenance therapy, and continue development of diagnostic tools that avoid endoscopy and biopsy analyses to more easily monitor disease activity.
Collapse
Affiliation(s)
- Alex Straumann
- Swiss EoE Clinic, Praxis Römerhof, Olten, Switzerland; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - David A Katzka
- Swiss EoE Clinic, Praxis Römerhof, Olten, Switzerland; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
100
|
di Pietro M, Canto MI, Fitzgerald RC. Endoscopic Management of Early Adenocarcinoma and Squamous Cell Carcinoma of the Esophagus: Screening, Diagnosis, and Therapy. Gastroenterology 2018; 154:421-436. [PMID: 28778650 PMCID: PMC6104810 DOI: 10.1053/j.gastro.2017.07.041] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/16/2022]
Abstract
Because the esophagus is easily accessible with endoscopy, early diagnosis and curative treatment of esophageal cancer is possible. However, diagnosis is often delayed because symptoms are not specific during early stages of tumor development. The onset of dysphagia is associated with advanced disease, which has a survival at 5 years lower than 15%. Population screening by endoscopy is not cost-effective, but a number of alternative imaging and cell analysis technologies are under investigation. The ideal screening test should be inexpensive, well tolerated, and applicable to primary care. Over the past 10 years, significant progress has been made in endoscopic diagnosis and treatment of dysplasia (squamous and Barrett's), and early esophageal cancer using resection and ablation technologies supported by evidence from randomized controlled trials. We review the state-of-the-art technologies for early diagnosis and minimally invasive treatment, which together could reduce the burden of disease.
Collapse
Affiliation(s)
| | - Marcia I Canto
- Division of Gastroenterology and Hepatology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | | |
Collapse
|