1
|
Jiang W, Zhang B, Xu J, Xue L, Wang L. Current status and perspectives of esophageal cancer: a comprehensive review. Cancer Commun (Lond) 2024. [PMID: 39723635 DOI: 10.1002/cac2.12645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Esophageal cancer (EC) continues to be a significant global health concern, with two main subtypes: esophageal squamous cell carcinoma and esophageal adenocarcinoma. Prevention and changes in etiology, improvements in early detection, and refinements in the treatment have led to remarkable progress in the outcomes of EC patients in the past two decades. This seminar provides an in-depth analysis of advances in the epidemiology, disease biology, screening, diagnosis, and treatment landscape of esophageal cancer, focusing on the ongoing debate surrounding multimodality therapy. Despite significant advancements, EC remains a deadly disease, underscoring the need for continued research into early detection methods, understanding the molecular mechanisms, and developing effective treatments.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| | - Bo Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Jiaqi Xu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, P. R. China
| |
Collapse
|
2
|
Wang C, Shi ZZ. Exosomes in esophageal cancer: function and therapeutic prospects. Med Oncol 2024; 42:18. [PMID: 39601925 DOI: 10.1007/s12032-024-02543-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024]
Abstract
Esophageal cancer (EC) is one of the most common malignant tumors worldwide. Exosomes are a type of extracellular vesicles produced by eukaryotic cells and present in all body fluids. Recent studies have revealed that exosomes can be used as a tool for cell signaling and have great potential in cancer diagnosis and treatment strategies. This article reviews the research progress of exosomes in EC in recent years, mainly including the mechanism of action, diagnostic markers, therapeutic targets, and drug carriers. The challenges faced are discussed to provide guidelines for further research in future.
Collapse
Affiliation(s)
- Chong Wang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Zhi-Zhou Shi
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
3
|
Chien S, Glen P, Bryce G, Cruickshank N, Penman I, Robertson K, Phull P, Crumley A, Gunjaca I, Apollos J, Miller M, Fletcher J, Fullarton G. Clinical application of capsule sponge testing in symptomatic reflux disease: a national prospective cohort study. BMC Gastroenterol 2024; 24:431. [PMID: 39592951 PMCID: PMC11590222 DOI: 10.1186/s12876-024-03503-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Capsule sponge testing, using an oesophageal cell collection device with biomarkers, was implemented nationally across Scotland in 2020 for symptomatic reflux patients referred to secondary care for non-urgent endoscopy. The aim was to use capsule sponge testing as a triage tool to reduce pressures on the endoscopy service during COVID-19, focus endoscopy resources on those most likely to have pathology and streamline the patient pathway. This prospective cohort study presents the first real-world results and evaluates the clinical application of capsule sponge testing in symptomatic reflux disease based on endoscopic biopsy results. METHODS Over a 32-month period, all patients undergoing capsule sponge testing for investigation of reflux symptoms across 11 Scottish health boards were identified from prospectively maintained databases. Individual patient records were interrogated to collect baseline demographics, capsule sponge test result (TFF3/atypia/p53) and ongoing clinical management. Further analysis was performed on patients who subsequently underwent upper gastrointestinal (UGI) endoscopy. RESULTS 1385 tests were performed for reflux symptoms in 1305 patients. The median follow-up time was 20 months (IQR 12-27). 1103/1385 tests (79.6%) were negative for biomarkers. 913/1305 (70.0%) patients were discharged with no additional investigation required. 355/1305 patients (27.2%) underwent UGI endoscopy due to a positive or insufficient result or ongoing symptoms. With insufficient tests excluded, 52/314 patients (16.6%) had intestinal metaplasia (IM) on endoscopic biopsies, which strongly correlated with positive biomarkers (88.5% vs. 11.5%; p < 0.001), including 1 case of dysplasia. 10/1103 sponge negative patients (0.9%) had biopsies demonstrating IM (n = 6) or malignancy (n = 4): 1 patient was diagnosed with oesophageal adenocarcinoma 27 months later and 3 patients had gastric malignancy, which relies on symptom assessment to direct to endoscopy since the capsule sponge is an oesophageal test. CONCLUSIONS Capsule sponge testing is effectively at identifying the 30% of symptomatic reflux patients requiring further investigation with UGI endoscopy and aiding the diagnosis of Barrett's oesophagus in clinical practice. Judicious follow-up of the discharged group will be critical to validate the ongoing use of capsule sponge testing long-term.
Collapse
Affiliation(s)
- Siobhan Chien
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1QH, UK.
- Centre for Sustainable Delivery, Golden Jubilee National Hospital, Clydebank, Glasgow, G81 4DN, UK.
| | - Paul Glen
- Department of General Surgery, Queen Elizabeth University Hospital, Glasgow, G51 4TF, UK
| | - Gavin Bryce
- Department of General Surgery, University Hospital Wishaw, Wishaw, ML2 0DP, UK
| | - Neil Cruickshank
- Department of General Surgery, Victoria Hospital, Kirkcaldy, KY2 5AH, UK
| | - Ian Penman
- Centre for Liver & Digestive Disorders, Royal Infirmary of Edinburgh, Edinburgh, EH16 4SA, UK
| | - Kevin Robertson
- Department of General Surgery, University Hospital Crosshouse, Kilmarnock, KA2 0BE, UK
| | - Perminder Phull
- Department of Gastroenterology, Aberdeen Royal Infirmary, Aberdeen, AB25 2ZN, UK
| | - Andrew Crumley
- Department of General Surgery, Forth Valley Royal Hospital, Larbert, FK5 4WR, UK
| | - Ivan Gunjaca
- Department of Gastroenterology, Raigmore Hospital, Inverness, IV2 3UJ, UK
| | - Jeyakumar Apollos
- Department of General Surgery, Dumfries & Galloway Royal Infirmary, Dumfries, DG2 8RX, UK
| | - Michael Miller
- Department of Gastroenterology, Ninewells Hospital, Dundee, DD2 1SG, UK
| | - Jonathan Fletcher
- Department of Gastroenterology, Borders General Hospital, Melrose, TD6 9BS, UK
| | - Grant Fullarton
- Centre for Sustainable Delivery, Golden Jubilee National Hospital, Clydebank, Glasgow, G81 4DN, UK
| |
Collapse
|
4
|
Tinland J, Gauld C, Sujobert P, Giroux É. Diagnostic staging and stratification in psychiatry and oncology: clarifying their conceptual, epistemological and ethical implications. MEDICINE, HEALTH CARE, AND PHILOSOPHY 2024; 27:333-347. [PMID: 38760623 DOI: 10.1007/s11019-024-10207-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 05/19/2024]
Abstract
Staging and stratification are two diagnostic approaches that have introduced a more dynamic outlook on the development of diseases, thus participating in blurring the line between the normal and the pathological. First, diagnostic staging, aiming to capture how diseases evolve in time and/or space through identifiable and gradually more severe stages, may be said to lean on an underlying assumption of "temporal determinism". Stratification, on the other hand, allows for the identification of various prognostic or predictive subgroups based on specific markers, relying on a more "mechanistic" or "statistical" form of determinism. There are two medical fields in which these developments have played a significant role and have given rise to sometimes profound nosological transformations: oncology and psychiatry. Drawing on examples from these two fields, this paper aims to provide much needed conceptual clarifications on both staging and stratification in order to outline how several epistemological and ethical issues may, in turn, arise. We argue that diagnostic staging ought to be detached from the assumption of temporal determinism, though it should still play an essential role in adapting interventions to stage. In doing so, it would help counterbalance stratification's own epistemological and ethical shortcomings. In this sense, the reflections and propositions developed in psychiatry can offer invaluable insights regarding how adopting a more transdiagnostic and cross-cutting perspective on temporality and disease dynamics may help combine both staging and stratification in clinical practice.
Collapse
Affiliation(s)
- Julia Tinland
- Aix Marseille Univ, Inserm, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, ISSPAM ; Chaire Démocratie en santé et engagement des personnes concernées par le cancer, Marseille, France.
| | - Christophe Gauld
- Service de Psychopathologie de l'Enfant et de l'Adolescent, Hospices Civils de Lyon, Lyon, F-69000, France
- Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS & Université Claude Bernard Lyon 1, Lyon, F-69000, France
| | - Pierre Sujobert
- Équipe Lymphoma Immunobiology, Centre international de recherche en infectiologie, université Lyon 1, Faculté de médecine et de maïeutique Lyon Sud, Lyon, France
- Service d'hématologie Biologique, Hospices civils de Lyon, hôpital Lyon Sud, Lyon, France
| | - Élodie Giroux
- Professeure des Universités en philosophie des sciences à l'université Jean Moulin Lyon 3, Institut de recherches philosophiques de Lyon (IRPHIL), Lyon, France
| |
Collapse
|
5
|
Sharif K, de Santiago ER, David P, Afek A, Gralnek IM, Ben-Horin S, Lahat A. Ecogastroenterology: cultivating sustainable clinical excellence in an environmentally conscious landscape. Lancet Gastroenterol Hepatol 2024; 9:550-563. [PMID: 38554732 DOI: 10.1016/s2468-1253(23)00414-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 04/02/2024]
Abstract
Gastrointestinal practices, especially endoscopy, have a substantial environmental impact, marked by notable greenhouse gas emissions and waste generation. As the world struggles with climate change, there emerges a pressing need to re-evaluate and reform the environmental footprint within gastrointestinal medicine. The challenge lies in finding a harmonious balance between ensuring clinical effectiveness and upholding environmental responsibility. This task involves recognising that the most significant reduction in the carbon footprint of endoscopy is achieved by avoiding unnecessary procedures; addressing the use of single-use endoscopes and accessories; and extending beyond the procedural suites to include clinics, virtual care, and conferences, among other aspects of gastrointestinal practice. The emerging digital realm in health care is crucial, given the potential environmental advantages of virtual gastroenterological care. Through an in-depth analysis, this review presents a path towards sustainable gastrointestinal practices, emphasising integrated strategies that prioritise both patient care and environmental stewardship.
Collapse
Affiliation(s)
- Kassem Sharif
- Department of Gastroenterology, Sheba Medical Centre, Ramat Gan, Israel; Department of Internal Medicine B, Sheba Medical Centre, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Enrique Rodriguez de Santiago
- Gastroenterology and Hepatology, Hospital Universitario Ramón y Cajal, Universidad de Alcalá, IRYCIS, CIBERehd, ISCIII, Madrid, Spain
| | - Paula David
- Department of Internal Medicine B, Sheba Medical Centre, Ramat Gan, Israel
| | - Arnon Afek
- Department of Gastroenterology, Sheba Medical Centre, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ian M Gralnek
- Ellen and Pinchas Mamber Institute of Gastroenterology and Hepatology, Emek Medical Centre, Afula, Israel; Rappaport Faculty of Medicine Technion Israel Institute of Technology, Haifa, Israel
| | - Shomron Ben-Horin
- Department of Gastroenterology, Sheba Medical Centre, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Lahat
- Department of Gastroenterology, Sheba Medical Centre, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
6
|
Chien S, Glen P, Penman I, Cruickshank N, Bryce G, Crumley A, Phull P, Miller M, Fletcher J, Gunjaca I, Apollos J, Robertson K, Fullarton G. Oesophageal cell collection device and biomarker testing to identify high-risk Barrett's patients requiring endoscopic investigation. Br J Surg 2024; 111:znae117. [PMID: 38736137 PMCID: PMC11089076 DOI: 10.1093/bjs/znae117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/25/2024] [Accepted: 04/11/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Barrett's oesophagus surveillance places significant burden on endoscopy services yet is vital to detect early cancerous change. Oesophageal cell collection device (OCCD) testing was introduced across Scotland for Barrett's surveillance in response to the COVID-19 pandemic. This national pragmatic retrospective study presents the CytoSCOT programme results and evaluates whether OCCD testing is successfully identifying high-risk Barrett's patients requiring urgent endoscopy. METHODS All patients undergoing OCCD testing for Barrett's surveillance across 11 Scottish health boards over a 32-month period were identified. Patients who underwent endoscopy within 12 months of OCCD test were included. Individual patient records were interrogated to record clinical information and OCCD test result to categorize patients into risk groups. Endoscopic histopathology results were analysed according to risk group and segment length. Patients were deemed high risk if the OCCD test demonstrated atypia and/or p53 positivity. RESULTS 4204 OCCD tests were performed in 3745 patients: 608 patients underwent endoscopy within 12 months and were included in this analysis. Patients with longer Barrett's segments were significantly more likely to have an abnormal OCCD test. 50/608 patients (8.2%) had high-grade dysplasia or cancer on endoscopic biopsies: this equates to 1.3% of the total group (50/3745). 46/50 patients (92.0%) were deemed high risk, triggering urgent endoscopy: this rose to 100% with insufficient tests removed. There were no cancers diagnosed within 12 months post-OCCD in the low-risk group. CONCLUSION OCCD testing is an effective triage tool to identify high-risk patients with Barrett's oesophagus requiring further investigation with endoscopy within the real-world setting.
Collapse
Affiliation(s)
- Siobhan Chien
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Centre for Sustainable Delivery, Golden Jubilee National Hospital, Glasgow, UK
| | - Paul Glen
- Department of General Surgery, Queen Elizabeth University Hospital, Glasgow, UK
| | - Ian Penman
- Centre for Liver & Digestive Disorders, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | | - Gavin Bryce
- Department of General Surgery, University Hospital Wishaw, Wishaw, UK
| | - Andrew Crumley
- Department of General Surgery, Forth Valley Royal Hospital, Larbert, UK
| | - Perminder Phull
- Department of Gastroenterology, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Michael Miller
- Department of Gastroenterology, Ninewells Hospital, Dundee, UK
| | - Jonathan Fletcher
- Department of Gastroenterology, Borders General Hospital, Melrose, UK
| | - Ivan Gunjaca
- Department of Gastroenterology, Raigmore Hospital, Inverness, UK
| | - Jeyakumar Apollos
- Department of General Surgery, Dumfries & Galloway Royal Infirmary, Dumfries, UK
| | - Kevin Robertson
- Department of General Surgery, University Hospital Crosshouse, Kilmarnock, UK
| | - Grant Fullarton
- Centre for Sustainable Delivery, Golden Jubilee National Hospital, Glasgow, UK
| |
Collapse
|
7
|
Chien S, Glen P, Penman I, Bryce G, Cruickshank N, Miller M, Crumley A, Fletcher J, Phull P, Gunjaca I, Robertson K, Apollos J, Fullarton G. National adoption of an esophageal cell collection device for Barrett's esophagus surveillance: impact on delay to investigation and pathological findings. Dis Esophagus 2024; 37:doae002. [PMID: 38267082 DOI: 10.1093/dote/doae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/30/2023] [Accepted: 01/04/2024] [Indexed: 01/26/2024]
Abstract
High quality Barrett's esophagus surveillance is crucial to detect early neoplastic changes. An esophageal cell collection device (OCCD) was introduced as a triage tool for Barrett's surveillance. This study aims to evaluate whether the Scottish OCCD program (CytoSCOT) has reduced delays to Barrett's surveillance, and whether delayed surveillance negatively impacts endoscopic pathology. All patients undergoing OCCD testing for Barrett's surveillance across 11 Scottish health boards between 14/9/2020 and 13/9/2022 were identified. Patients were dichotomised into two groups (Year 1 vs. Year 2), with individual records interrogated to record demographics, recommended surveillance interval, time from last endoscopy to OCCD test, and OCCD result. Patients were deemed high-risk if the OCCD demonstrated atypia and/or p53 positivity. Further analysis was performed on patients who underwent endoscopy within 12 months of OCCD testing. A total of 3223 OCCD tests were included in the analysis (1478 in Year 1; 1745 in Year 2). In Year 1 versus Year 2, there was a longer median delay to surveillance (9 vs. 5 months; P < 0.001), increased proportion of patients with delayed surveillance (72.6% vs. 57.0%; P < 0.001), and more high-risk patients (12.0% vs. 5.3%; P < 0.001). 425/3223 patients (13.2%) were further investigated with upper gastrointestinal endoscopy, 57.9% of which were high-risk. As surveillance delay increased beyond 24 months, high-risk patients were significantly more likely to develop dysplasia or malignancy (P = 0.004). Delayed Barrett's esophagus surveillance beyond 24 months is associated with increased risk of pre-cancerous pathology. The CytoSCOT program has reduced delays in surveillance, promoting earlier detection of dysplasia and reducing burden on endoscopy services.
Collapse
Affiliation(s)
- Siobhan Chien
- Centre for Sustainable Delivery, Golden Jubilee National Hospital, Clydebank, Glasgow G81 4DN, UK
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1QH, UK
| | - Paul Glen
- Department of General Surgery, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Ian Penman
- Centre for Liver & Digestive Disorders, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK
| | - Gavin Bryce
- Department of General Surgery, University Hospital Wishaw, Wishaw ML2 0DP, UK
| | - Neil Cruickshank
- Department of General Surgery, Victoria Hospital, Kirkcaldy KY2 5AH, UK
| | - Michael Miller
- Department of Gastroenterology, Ninewells Hospital, Dundee DD2 1SG, UK
| | - Andrew Crumley
- Department of General Surgery, Forth Valley Royal Hospital, Larbert FK5 4WR, UK
| | - Jonathan Fletcher
- Department of Gastroenterology, Borders General Hospital, Melrose TD6 9BS, UK
| | - Perminder Phull
- Department of Gastroenterology, Aberdeen Royal Infirmary, Aberdeen AB25 2ZN, UK
| | - Ivan Gunjaca
- Department of Gastroenterology, Raigmore Hospital, Inverness IV2 3UJ, UK
| | - Kevin Robertson
- Department of General Surgery, University Hospital Crosshouse, Kilmarnock KA2 0BE, UK
| | - Jeyakumar Apollos
- Department of General Surgery, Dumfries & Galloway Royal Infirmary, Dumfries DG2 8RX, UK
| | - Grant Fullarton
- Centre for Sustainable Delivery, Golden Jubilee National Hospital, Clydebank, Glasgow G81 4DN, UK
| |
Collapse
|
8
|
Bouzid K, Sharma H, Killcoyne S, Castro DC, Schwaighofer A, Ilse M, Salvatelli V, Oktay O, Murthy S, Bordeaux L, Moore L, O'Donovan M, Thieme A, Nori A, Gehrung M, Alvarez-Valle J. Enabling large-scale screening of Barrett's esophagus using weakly supervised deep learning in histopathology. Nat Commun 2024; 15:2026. [PMID: 38467600 PMCID: PMC10928093 DOI: 10.1038/s41467-024-46174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Abstract
Timely detection of Barrett's esophagus, the pre-malignant condition of esophageal adenocarcinoma, can improve patient survival rates. The Cytosponge-TFF3 test, a non-endoscopic minimally invasive procedure, has been used for diagnosing intestinal metaplasia in Barrett's. However, it depends on pathologist's assessment of two slides stained with H&E and the immunohistochemical biomarker TFF3. This resource-intensive clinical workflow limits large-scale screening in the at-risk population. To improve screening capacity, we propose a deep learning approach for detecting Barrett's from routinely stained H&E slides. The approach solely relies on diagnostic labels, eliminating the need for expensive localized expert annotations. We train and independently validate our approach on two clinical trial datasets, totaling 1866 patients. We achieve 91.4% and 87.3% AUROCs on discovery and external test datasets for the H&E model, comparable to the TFF3 model. Our proposed semi-automated clinical workflow can reduce pathologists' workload to 48% without sacrificing diagnostic performance, enabling pathologists to prioritize high risk cases.
Collapse
Affiliation(s)
| | | | | | | | | | - Max Ilse
- Microsoft Health Futures, Cambridge, UK
| | | | | | | | | | - Luiza Moore
- Department of Histopathology, Addenbrookes Hospital, Cambridge University NHS Foundation Trust, Cambridge, UK
| | - Maria O'Donovan
- Cyted Ltd, Cambridge, UK
- Department of Histopathology, Addenbrookes Hospital, Cambridge University NHS Foundation Trust, Cambridge, UK
| | | | | | | | | |
Collapse
|
9
|
Abstract
The cumulative pool of cell-free DNA (cfDNA) molecules within bodily fluids represents a highly dense and multidimensional information repository. This "biological mirror" provides real-time insights into the composition, function, and dynamics of the diverse genomes within the body, enabling significant advancements in personalized molecular medicine. However, effective use of this information necessitates meticulous classification of distinct cfDNA subtypes with exceptional precision. While cfDNA molecules originating from different sources exhibit numerous genetic, epigenetic, and physico-chemical variations, they also share common features that complicate analyses. Considerable progress has been achieved in mapping the landscape of cfDNA features, their clinical correlations, and optimizing extraction procedures, analytical approaches, bioinformatics pipelines, and machine learning algorithms. Nevertheless, preanalytical workflows, despite their profound impact on cfDNA measurements, have not progressed at a corresponding pace. In this perspective article, we emphasize the pivotal role of robust preanalytical procedures in the development and clinical integration of cfDNA assays, highlighting persistent obstacles and emerging challenges.
Collapse
Affiliation(s)
- Abel J Bronkhorst
- Munich Biomarker Research Center, Institute of Laboratory Medicine, German Heart Center, Technical University Munich, Munich, Germany
| | - Stefan Holdenrieder
- Munich Biomarker Research Center, Institute of Laboratory Medicine, German Heart Center, Technical University Munich, Munich, Germany
| |
Collapse
|
10
|
Landy R, Killcoyne S, Tang C, Juniat S, O’Donovan M, Goel N, Gehrung M, Fitzgerald RC. Real-world implementation of non-endoscopic triage testing for Barrett's oesophagus during COVID-19. QJM 2023; 116:659-666. [PMID: 37220898 PMCID: PMC10497181 DOI: 10.1093/qjmed/hcad093] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/27/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND The Coronavirus pandemic (COVID-19) curtailed endoscopy services, adding to diagnostic backlogs. Building on trial evidence for a non-endoscopic oesophageal cell collection device coupled with biomarkers (Cytosponge), an implementation pilot was launched for patients on waiting lists for reflux and Barrett's oesophagus surveillance. AIMS (i) To review reflux referral patterns and Barrett's surveillance practices. (ii) To evaluate the range of Cytosponge findings and impact on endoscopy services. DESIGN AND METHODS Cytosponge data from centralized laboratory processing (trefoil factor 3 (TFF3) for intestinal metaplasia (IM), haematoxylin & eosin for cellular atypia and p53 for dysplasia) over a 2-year period were included. RESULTS A total of 10 577 procedures were performed in 61 hospitals in England and Scotland, of which 92.5% (N = 9784/10 577) were sufficient for analysis. In the reflux cohort (N = 4074 with gastro-oesophageal junction sampling), 14.7% had one or more positive biomarkers (TFF3: 13.6% (N = 550/4056), p53: 0.5% (21/3974), atypia: 1.5% (N = 63/4071)), requiring endoscopy. Among samples from individuals undergoing Barrett's surveillance (N = 5710 with sufficient gland groups), TFF3-positivity increased with segment length (odds ratio = 1.37 per cm (95% confidence interval: 1.33-1.41, P < 0.001)). Some surveillance referrals (21.5%, N = 1175/5471) had ≤1 cm segment length, of which 65.9% (707/1073) were TFF3 negative. Of all surveillance procedures, 8.3% had dysplastic biomarkers (4.0% (N = 225/5630) for p53 and 7.6% (N = 430/5694) for atypia), increasing to 11.8% (N = 420/3552) in TFF3+ cases with confirmed IM and 19.7% (N = 58/294) in ultra-long segments. CONCLUSIONS Cytosponge-biomarker tests enabled targeting of endoscopy services to higher-risk individuals, whereas those with TFF3 negative ultra-short segments could be reconsidered regarding their Barrett's oesophagus status and surveillance requirements. Long-term follow-up will be important in these cohorts.
Collapse
Affiliation(s)
- R Landy
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - S Killcoyne
- Cyted Ltd, 22 Station Road, Cambridge CB1 2JD, UK
| | - C Tang
- Cyted Ltd, 22 Station Road, Cambridge CB1 2JD, UK
| | - S Juniat
- Cyted Ltd, 22 Station Road, Cambridge CB1 2JD, UK
| | - M O’Donovan
- Cyted Ltd, 22 Station Road, Cambridge CB1 2JD, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - N Goel
- Cyted Ltd, 22 Station Road, Cambridge CB1 2JD, UK
| | - M Gehrung
- Cyted Ltd, 22 Station Road, Cambridge CB1 2JD, UK
| | - R C Fitzgerald
- Department of Oncology, Early Cancer Institute, University of Cambridge, Cambridge CB2 0XZ, UK
| |
Collapse
|
11
|
Berman AG, Orchard WR, Gehrung M, Markowetz F. SliDL: A toolbox for processing whole-slide images in deep learning. PLoS One 2023; 18:e0289499. [PMID: 37549131 PMCID: PMC10406329 DOI: 10.1371/journal.pone.0289499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/20/2023] [Indexed: 08/09/2023] Open
Abstract
The inspection of stained tissue slides by pathologists is essential for the early detection, diagnosis and monitoring of disease. Recently, deep learning methods for the analysis of whole-slide images (WSIs) have shown excellent performance on these tasks, and have the potential to substantially reduce the workload of pathologists. However, WSIs present a number of unique challenges for analysis, requiring special consideration of image annotations, slide and image artefacts, and evaluation of WSI-trained model performance. Here we introduce SliDL, a Python library for performing pre- and post-processing of WSIs. SliDL makes WSI data handling easy, allowing users to perform essential processing tasks in a few simple lines of code, bridging the gap between standard image analysis and WSI analysis. We introduce each of the main functionalities within SliDL: from annotation and tile extraction to tissue detection and model evaluation. We also provide 'code snippets' to guide the user in running SliDL. SliDL has been designed to interact with PyTorch, one of the most widely used deep learning libraries, allowing seamless integration into deep learning workflows. By providing a framework in which deep learning methods for WSI analysis can be developed and applied, SliDL aims to increase the accessibility of an important application of deep learning.
Collapse
Affiliation(s)
- Adam G. Berman
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - William R. Orchard
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Marcel Gehrung
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
12
|
Honing J, Fitzgerald RC. Categorizing Risks within Barrett's Esophagus To Guide Surveillance and Interception; Suggesting a New Framework. Cancer Prev Res (Phila) 2023; 16:313-320. [PMID: 37259801 PMCID: PMC10234311 DOI: 10.1158/1940-6207.capr-22-0447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/08/2023] [Accepted: 04/05/2023] [Indexed: 06/02/2023]
Abstract
Barrett's esophagus is a precancerous condition that can progress in a stepwise manner to dysplasia and eventually esophageal adenocarcinoma (EAC). Once diagnosed, patients with Barrett's esophagus are kept on surveillance to detect progression so that timely intervention can occur with endoscopic therapy. Several demographic and clinical risk factors are known to increase progression toward EAC, such as longer Barrett's segments, and these patients are kept on tighter surveillance. While p53 IHC has been advocated as an adjunct to histopathologic diagnosis, use of this biomarker is variable, and no other molecular factors are currently applied. Given the new evidence available, it is time to consider whether other risk factors or tools could be applied in clinical practice to decide on closer or attenuated surveillance. In this commentary, we summarize the most relevant risk factors for Barrett's esophagus progression, highlight the most promising novel risk stratification tools-including nonendoscopic triage and commercial biomarker panels, and propose a new framework suggesting how to incorporate risk stratification into clinical practice.
Collapse
Affiliation(s)
- Judith Honing
- Early Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
13
|
Berman AG, Tan WK, O'Donovan M, Markowetz F, Fitzgerald RC. Quantification of TFF3 expression from a non-endoscopic device predicts clinically relevant Barrett's oesophagus by machine learning. EBioMedicine 2022; 82:104160. [PMID: 35843173 PMCID: PMC9297109 DOI: 10.1016/j.ebiom.2022.104160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Intestinal metaplasia (IM) is pre-neoplastic with variable cancer risk. Cytosponge-TFF3 test can detect IM. We aimed to 1) assess whether quantitative TFF3 scores can distinguish clinically relevant Barrett's oesophagus (BO) (C≥1 or M≥3) from focal IM pathologies (C<1, M<3 or IM of gastro-oesophageal junction); 2) whether TFF3 counts can be automated to inform clinical practice. METHODS Patients from the Barett's oEsophagus Screening Trial 2 (BEST2) case-control and BEST3 randomised trials were used. For aim 1, TFF3-positive glands were scored manually and correlated with clinical diagnosis. For aim 2, machine learning approach was used to obtain TFF3 count and logistic regression with cross-validation was trained on the BEST2 dataset (n = 529) and tested in the BEST3 dataset (n = 158). FINDINGS Patients with clinically relevant BO had higher mean TFF3 gland count compared to focal IM pathologies (mean difference 4.14; 95% confidence interval, CI 2.76-5.52, p < 0.001). The mean class-balanced validation accuracy was 0.84 (95% CI 0.77-0.90), and precision of 0.95 (95% CI 0.87-1.00) for detecting clinically relevant BO. Applying this model on BEST3 showed precision of 0.91 (95% CI 0.85-0.97) for focal IM pathologies with a class-balanced accuracy of 0.77 (95% CI 0.69-0.84). Using this model, 55% of patients (87/158) in BEST3 would fall below the threshold for clinically relevant BO and could avoid gastroscopy, while only missing 5.1% of patients (8/158). INTERPRETATION Automated Cytosponge-TFF3 gland quantification may enable thresholds to be set to trigger confirmatory gastroscopy to minimize overdiagnosis of focal IM pathologies with very low cancer-associated risk. FUNDING Cancer Research UK (12088/16893 and C14478/A21047).
Collapse
Affiliation(s)
- Adam G Berman
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - W Keith Tan
- Early Cancer Institute, Department of Oncology, Hutchison Building, University of Cambridge, Cambridge, UK; Department of Gastroenterology, Addenbrooke's Hospital, Cambridge University NHS Foundation Trust, Cambridge UK
| | - Maria O'Donovan
- Department of Histopathology, Addenbrookes Hospital, Cambridge University NHS Foundation Trust, Cambridge, UK; Cyted Ltd, Cambridge, UK
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| | - Rebecca C Fitzgerald
- Early Cancer Institute, Department of Oncology, Hutchison Building, University of Cambridge, Cambridge, UK; Department of Gastroenterology, Addenbrooke's Hospital, Cambridge University NHS Foundation Trust, Cambridge UK.
| |
Collapse
|
14
|
Paulson TG, Galipeau PC, Oman KM, Sanchez CA, Kuhner MK, Smith LP, Hadi K, Shah M, Arora K, Shelton J, Johnson M, Corvelo A, Maley CC, Yao X, Sanghvi R, Venturini E, Emde AK, Hubert B, Imielinski M, Robine N, Reid BJ, Li X. Somatic whole genome dynamics of precancer in Barrett's esophagus reveals features associated with disease progression. Nat Commun 2022; 13:2300. [PMID: 35484108 PMCID: PMC9050715 DOI: 10.1038/s41467-022-29767-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/25/2022] [Indexed: 01/08/2023] Open
Abstract
While the genomes of normal tissues undergo dynamic changes over time, little is understood about the temporal-spatial dynamics of genomes in premalignant tissues that progress to cancer compared to those that remain cancer-free. Here we use whole genome sequencing to contrast genomic alterations in 427 longitudinal samples from 40 patients with stable Barrett’s esophagus compared to 40 Barrett’s patients who progressed to esophageal adenocarcinoma (ESAD). We show the same somatic mutational processes are active in Barrett’s tissue regardless of outcome, with high levels of mutation, ESAD gene and focal chromosomal alterations, and similar mutational signatures. The critical distinction between stable Barrett’s versus those who progress to cancer is acquisition and expansion of TP53−/− cell populations having complex structural variants and high-level amplifications, which are detectable up to six years prior to a cancer diagnosis. These findings reveal the timing of common somatic genome dynamics in stable Barrett’s esophagus and define key genomic features specific to progression to esophageal adenocarcinoma, both of which are critical for cancer prevention and early detection strategies. Barrett’s esophagus is a pre-malignant condition that can progress to esophageal cancer. Here, the authors carry out whole genome sequencing of samples from patients who did or did not progress to cancer and find that mutations in many genes occur regardless of progression status, but also find features associated with progressive disease.
Collapse
Affiliation(s)
- Thomas G Paulson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA.
| | - Patricia C Galipeau
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA
| | - Kenji M Oman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA
| | - Carissa A Sanchez
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA
| | - Mary K Kuhner
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195-5065, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, 98195-5065, USA
| | - Lucian P Smith
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195-5065, USA
| | - Kevin Hadi
- New York Genome Center (NYGC), New York, NY, 10013, USA
| | - Minita Shah
- New York Genome Center (NYGC), New York, NY, 10013, USA
| | - Kanika Arora
- New York Genome Center (NYGC), New York, NY, 10013, USA
| | | | - Molly Johnson
- New York Genome Center (NYGC), New York, NY, 10013, USA
| | - Andre Corvelo
- New York Genome Center (NYGC), New York, NY, 10013, USA
| | - Carlo C Maley
- Arizona Cancer Evolution Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - Xiaotong Yao
- New York Genome Center (NYGC), New York, NY, 10013, USA
| | | | | | | | | | - Marcin Imielinski
- New York Genome Center (NYGC), New York, NY, 10013, USA.,Department of Pathology and Laboratory Medicine, Englander Institute for Precision Medicine, Institute for Computational Biomedicine and Meyer Cancer Center, Weill Cornell Medical College, New York, NY, 10065, USA
| | | | - Brian J Reid
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA.,Department of Genome Sciences, University of Washington, Seattle, WA, 98195-5065, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, 98195-5065, USA.,Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Xiaohong Li
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109-1024, USA.
| |
Collapse
|
15
|
Ratcliffe E, Britton J, Hamdy S, McLaughlin J, Ang Y. Developing patient-orientated Barrett's oesophagus services: the role of dedicated services. BMJ Open Gastroenterol 2022; 9:bmjgast-2021-000829. [PMID: 35193888 PMCID: PMC8867250 DOI: 10.1136/bmjgast-2021-000829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/09/2022] [Indexed: 11/03/2022] Open
Abstract
Introduction Barrett’s oesophagus (BO) is common and is a precursor to oesophageal adenocarcinoma with a 0.33% per annum risk of progression. Surveillance and follow-up services for BO have been shown to be lacking, with studies showing inadequate adherence to guidelines and patients reporting a need for greater disease-specific knowledge. This review explores the emerging role of dedicated services for patients with BO. Methods A literature search of PubMed, MEDLINE, Embase, Emcare, HMIC, BNI, CiNAHL, AMED and PsycINFO in regard to dedicated BO care pathways was undertaken. Results Prospective multicentre and randomised trials were lacking. Published cohort data are encouraging with improvements in guideline adherence with dedicated services, with one published study showing significant improvements in dysplasia detection rates. Accuracy of allocation to surveillance endoscopy has been shown to hold cost savings, and a study of a dedicated clinic showed increased discharges from unnecessary surveillance. Training modalities for BO surveillance and dysplasia detection exist, which could be used to educate a BO workforce. Qualitative and quantitative studies have shown patients report high levels of cancer worry and poor disease-specific knowledge, but few studies have explored follow-up care models despite being a patient and clinician priority for research. Conclusions Cost–benefit analysis for dedicated services, considering both financial and environmental impacts, and more robust clinical data must be obtained to support this model of care in the wider health service. Greater understanding is needed of the root causes for poor guideline adherence, and disease-specific models of care should be designed around clinical and patient-reported outcomes to address the unmet needs of patients with BO.
Collapse
Affiliation(s)
- Elizabeth Ratcliffe
- Gastroenterology, Wrightington Wigan and Leigh NHS Foundation Trust, Leigh, UK .,School of Medical Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
| | - James Britton
- Department of Gastroenterology, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - Shaheen Hamdy
- School of Medical Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK.,Department of Gastroenterology, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - John McLaughlin
- School of Medical Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK.,Department of Gastroenterology, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - Yeng Ang
- School of Medical Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK.,Department of Gastroenterology, Northern Care Alliance NHS Foundation Trust, Salford, UK
| |
Collapse
|
16
|
Maity AK, Stone TC, Ward V, Webster AP, Yang Z, Hogan A, McBain H, Duku M, Ho KMA, Wolfson P, Graham DG, Beck S, Teschendorff AE, Lovat LB. Novel epigenetic network biomarkers for early detection of esophageal cancer. Clin Epigenetics 2022; 14:23. [PMID: 35164838 PMCID: PMC8845366 DOI: 10.1186/s13148-022-01243-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/04/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Early detection of esophageal cancer is critical to improve survival. Whilst studies have identified biomarkers, their interpretation and validity is often confounded by cell-type heterogeneity. RESULTS Here we applied systems-epigenomic and cell-type deconvolution algorithms to a discovery set encompassing RNA-Seq and DNA methylation data from esophageal adenocarcinoma (EAC) patients and matched normal-adjacent tissue, in order to identify robust biomarkers, free from the confounding effect posed by cell-type heterogeneity. We identify 12 gene-modules that are epigenetically deregulated in EAC, and are able to validate all 12 modules in 4 independent EAC cohorts. We demonstrate that the epigenetic deregulation is present in the epithelial compartment of EAC-tissue. Using single-cell RNA-Seq data we show that one of these modules, a proto-cadherin module centered around CTNND2, is inactivated in Barrett's Esophagus, a precursor lesion to EAC. By measuring DNA methylation in saliva from EAC cases and controls, we identify a chemokine module centered around CCL20, whose methylation patterns in saliva correlate with EAC status. CONCLUSIONS Given our observations that a CCL20 chemokine network is overactivated in EAC tissue and saliva from EAC patients, and that in independent studies CCL20 has been found to be overactivated in EAC tissue infected with the bacterium F. nucleatum, a bacterium that normally inhabits the oral cavity, our results highlight the possibility of using DNAm measurements in saliva as a proxy for changes occurring in the esophageal epithelium. Both the CTNND2/CCL20 modules represent novel promising network biomarkers for EAC that merit further investigation.
Collapse
Affiliation(s)
- Alok K Maity
- CAS Key Lab of Computational Biology, Shanghai Institute for Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Timothy C Stone
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Vanessa Ward
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Amy P Webster
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Zhen Yang
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Aine Hogan
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Hazel McBain
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Margaraet Duku
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Kai Man Alexander Ho
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - Paul Wolfson
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK
| | - David G Graham
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK.,Division of GI Services, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK
| | | | - Stephan Beck
- UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Andrew E Teschendorff
- CAS Key Lab of Computational Biology, Shanghai Institute for Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
| | - Laurence B Lovat
- Division of Surgery and Interventional Science, University College London, Gower Street, London, WC1E 6BT, UK. .,Division of GI Services, University College London Hospitals NHS Foundation Trust, 235 Euston Road, London, NW1 2BU, UK.
| |
Collapse
|
17
|
Masqué-Soler N, Gehrung M, Kosmidou C, Li X, Diwan I, Rafferty C, Atabakhsh E, Markowetz F, Fitzgerald RC. Computational pathology aids derivation of microRNA biomarker signals from Cytosponge samples. EBioMedicine 2022; 76:103814. [PMID: 35051729 PMCID: PMC8883000 DOI: 10.1016/j.ebiom.2022.103814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/22/2021] [Accepted: 01/04/2022] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND Non-endoscopic cell collection devices combined with biomarkers can detect Barrett's intestinal metaplasia and early oesophageal cancer. However, assays performed on multi-cellular samples lose information about the cell source of the biomarker signal. This cross-sectional study examines whether a bespoke artificial intelligence-based computational pathology tool could ascertain the cellular origin of microRNA biomarkers, to inform interpretation of the disease pathology, and confirm biomarker validity. METHODS The microRNA expression profiles of 110 targets were assessed with a custom multiplexed panel in a cohort of 117 individuals with reflux that took a Cytosponge test. A computational pathology tool quantified the amount of columnar epithelium present in pathology slides, and results were correlated with microRNA signals. An independent cohort of 139 Cytosponges, each from an individual patient, was used to validate the findings via qPCR. FINDINGS Seventeen microRNAs are upregulated in BE compared to healthy squamous epithelia, of which 13 remain upregulated in dysplasia. A pathway enrichment analysis confirmed association to neoplastic and cell cycle regulation processes. Ten microRNAs positively correlated with columnar epithelium content, with miRNA-192-5p and -194-5p accurately detecting the presence of gastric cells (AUC 0.97 and 0.95). In contrast, miR-196a-5p is confirmed as a specific BE marker. INTERPRETATION Computational pathology tools aid accurate cellular attribution of molecular signals. This innovative design with multiplex microRNA coupled with artificial intelligence has led to discovery of a quality control metric suitable for large scale application of the Cytosponge. Similar approaches could aid optimal interpretation of biomarkers for clinical use. FUNDING Funded by the NIHR Cambridge Biomedical Research Centre, the Medical Research Council, the Rosetrees and Stoneygate Trusts, and CRUK core grants.
Collapse
Affiliation(s)
- Neus Masqué-Soler
- MRC Cancer Unit, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK.
| | - Marcel Gehrung
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Cassandra Kosmidou
- MRC Cancer Unit, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Xiaodun Li
- MRC Cancer Unit, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Izzuddin Diwan
- Abcam Inc., 1 Kendall Sq B2304, Cambridge, MA, 02139, United States
| | - Conor Rafferty
- Abcam Inc., 1 Kendall Sq B2304, Cambridge, MA, 02139, United States
| | - Elnaz Atabakhsh
- Abcam Inc., 1 Kendall Sq B2304, Cambridge, MA, 02139, United States
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | | |
Collapse
|
18
|
Eluri S, Paterson A, Lauren BN, O’Donovan M, Bhandari P, di Pietro M, Lee M, Haidry R, Lovat L, Ragunath K, Hur C, Fitzgerald RC, Shaheen NJ. Utility and Cost-Effectiveness of a Nonendoscopic Approach to Barrett's Esophagus Surveillance After Endoscopic Therapy. Clin Gastroenterol Hepatol 2022; 20:e51-e63. [PMID: 33581357 PMCID: PMC8352994 DOI: 10.1016/j.cgh.2021.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/28/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS A non-endoscopic approach to Barrett's esophagus (BE) surveillance after radiofrequency ablation (RFA) would offer a less invasive method for monitoring. We assessed the test characteristics and cost-effectiveness of the Cytosponge (Medtronic, Minneapolis, MN) in post-RFA patients. METHODS We performed a multicenter study of dysplastic BE patients after at least one round of RFA. A positive Cytosponge before endoscopy was defined as intestinal metaplasia (IM) on cytological assessment and/or TFF3 immunohistochemistry. Sensitivity, specificity, and receiver operator characteristic (ROC) curves were calculated. Multivariable regression was used to estimate the odds of a positive Cytosponge in BE. A microsimulation cost-effectiveness model was performed to assess outcomes of various surveillance strategies: endoscopy-only, Cytosponge-only, and alternating endoscopy/Cytosponge. RESULTS Of 234 patients, Cytosponge adequately sampled the distal esophagus in 175 (75%). Of the 142 with both endoscopic and histologic data, 19 (13%) had residual/recurrent BE. For detecting any residual Barrett's, Cytosponge had a sensitivity of 74%, specificity of 85%, accuracy of 84%, and ROC curve showed an area under the curve of 0.74. The adjusted odds of a positive Cytosponge in BE were 17.1 (95% CI, 5.2-55.9). Cytosponge-only surveillance dominated all the surveillance strategies, being both less costly and more effective. Cytosponge-only surveillance required <1/4th the endoscopies, resulting in only 0.69 additional EAC cases/1000 patients, and no increase in EAC deaths when compared to currently-practiced endoscopy-only surveillance. CONCLUSIONS A positive Cytosponge test was strongly associated with residual BE after ablation. While the assay needs further refinement in this context, it could serve as a cost-effective surveillance examination.
Collapse
Affiliation(s)
- Swathi Eluri
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, North Carolina.
| | - Anna Paterson
- MRC Cancer Unit, University of Cambridge, Cambridge, United Kingdom
| | - Brianna N. Lauren
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center
| | - Maria O’Donovan
- MRC Cancer Unit, University of Cambridge, Cambridge, United Kingdom
| | - Pradeep Bhandari
- Department of Gastroenterology, Queen Alexandra Hospital, Portsmouth, United Kingdom
| | | | - Minyi Lee
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center
| | - Rehan Haidry
- Division of Surgery and Interventional Science, University College London Hospital, London, UK
| | - Laurence Lovat
- Division of Surgery and Interventional Science, University College London Hospital, London, UK
| | - Krish Ragunath
- Nottingham Digestive Diseases Center, NIHR Biomedical Research Centre, University of Nottingham, Queens Medical Centre Campus, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Chin Hur
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center
| | | | - Nicholas J. Shaheen
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, NC,Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, NC
| |
Collapse
|
19
|
Pilonis ND, Killcoyne S, Tan WK, O'Donovan M, Malhotra S, Tripathi M, Miremadi A, Debiram-Beecham I, Evans T, Phillips R, Morris DL, Vickery C, Harrison J, di Pietro M, Ortiz-Fernandez-Sordo J, Haidry R, Kerridge A, Sasieni PD, Fitzgerald RC. Use of a Cytosponge biomarker panel to prioritise endoscopic Barrett's oesophagus surveillance: a cross-sectional study followed by a real-world prospective pilot. Lancet Oncol 2022; 23:270-278. [PMID: 35030332 PMCID: PMC8803607 DOI: 10.1016/s1470-2045(21)00667-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Endoscopic surveillance is recommended for patients with Barrett's oesophagus because, although the progression risk is low, endoscopic intervention is highly effective for high-grade dysplasia and cancer. However, repeated endoscopy has associated harms and access has been limited during the COVID-19 pandemic. We aimed to evaluate the role of a non-endoscopic device (Cytosponge) coupled with laboratory biomarkers and clinical factors to prioritise endoscopy for Barrett's oesophagus. METHODS We first conducted a retrospective, multicentre, cross-sectional study in patients older than 18 years who were having endoscopic surveillance for Barrett's oesophagus (with intestinal metaplasia confirmed by TFF3 and a minimum Barrett's segment length of 1 cm [circumferential or tongues by the Prague C and M criteria]). All patients had received the Cytosponge and confirmatory endoscopy during the BEST2 (ISRCTN12730505) and BEST3 (ISRCTN68382401) clinical trials, from July 7, 2011, to April 1, 2019 (UK Clinical Research Network Study Portfolio 9461). Participants were divided into training (n=557) and validation (n=334) cohorts to identify optimal risk groups. The biomarkers evaluated were overexpression of p53, cellular atypia, and 17 clinical demographic variables. Endoscopic biopsy diagnosis of high-grade dysplasia or cancer was the primary endpoint. Clinical feasibility of a decision tree for Cytosponge triage was evaluated in a real-world prospective cohort from Aug 27, 2020 (DELTA; ISRCTN91655550; n=223), in response to COVID-19 and the need to provide an alternative to endoscopic surveillance. FINDINGS The prevalence of high-grade dysplasia or cancer determined by the current gold standard of endoscopic biopsy was 17% (92 of 557 patients) in the training cohort and 10% (35 of 344) in the validation cohort. From the new biomarker analysis, three risk groups were identified: high risk, defined as atypia or p53 overexpression or both on Cytosponge; moderate risk, defined by the presence of a clinical risk factor (age, sex, and segment length); and low risk, defined as Cytosponge-negative and no clinical risk factors. The risk of high-grade dysplasia or intramucosal cancer in the high-risk group was 52% (68 of 132 patients) in the training cohort and 41% (31 of 75) in the validation cohort, compared with 2% (five of 210) and 1% (two of 185) in the low-risk group, respectively. In the real-world setting, Cytosponge results prospectively identified 39 (17%) of 223 patients as high risk (atypia or p53 overexpression, or both) requiring endoscopy, among whom the positive predictive value was 31% (12 of 39 patients) for high-grade dysplasia or intramucosal cancer and 44% (17 of 39) for any grade of dysplasia. INTERPRETATION Cytosponge atypia, p53 overexpression, and clinical risk factors (age, sex, and segment length) could be used to prioritise patients for endoscopy. Further investigation could validate their use in clinical practice and lead to a substantial reduction in endoscopy procedures compared with current surveillance pathways. FUNDING Medical Research Council, Cancer Research UK, Innovate UK.
Collapse
Affiliation(s)
| | - Sarah Killcoyne
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - W Keith Tan
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Maria O'Donovan
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK; Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Shalini Malhotra
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK; Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Monika Tripathi
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK; Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Ahmad Miremadi
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK; Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Irene Debiram-Beecham
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Tara Evans
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Rosemary Phillips
- Department of Gastroenterology, Princess Alexandra Hospital NHS Trust, Harlow, UK
| | - Danielle L Morris
- Department of Gastroenterology, East and North Herts NHS Trust, Stevenage, UK
| | - Craig Vickery
- Department of Surgery, West Suffolk Hospital, Bury St Edmunds, UK
| | - Jon Harrison
- Department of Gastroenterology, Harrogate District Hospital, Harrogate, UK
| | | | - Jacobo Ortiz-Fernandez-Sordo
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| | - Rehan Haidry
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| | - Abigail Kerridge
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Peter D Sasieni
- Cancer Prevention Group in Clinical Trials Unit, King's Clinical Trials Unit, King's College London, London, UK
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Schmidt M, Hackett RJ, Baker AM, McDonald SAC, Quante M, Graham TA. Evolutionary dynamics in Barrett oesophagus: implications for surveillance, risk stratification and therapy. Nat Rev Gastroenterol Hepatol 2022; 19:95-111. [PMID: 34728819 DOI: 10.1038/s41575-021-00531-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Cancer development is a dynamic evolutionary process characterized by marked intratumoural heterogeneity at the genetic, epigenetic and phenotypic levels. Barrett oesophagus, the pre-malignant condition to oesophageal adenocarcinoma (EAC), is an exemplary system to longitudinally study the evolution of malignancy. Evidence has emerged of Barrett oesophagus lesions pre-programmed for progression to EAC many years before clinical detection, indicating a considerable window for therapeutic intervention. In this Review, we explore the mechanisms underlying clonal expansion and contraction that establish the Barrett oesophagus clonal mosaicism over time and space and discuss intrinsic genotypic and extrinsic environmental drivers that direct the evolutionary trajectory of Barrett oesophagus towards a malignant phenotype. We propose that understanding and exploiting the evolutionary dynamics of Barrett oesophagus will identify novel therapeutic targets, improve prognostic tools and offer the opportunity for personalized surveillance programmes geared to prevent progression to EAC.
Collapse
Affiliation(s)
- Melissa Schmidt
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), München, Germany
| | - Richard J Hackett
- Clonal Dynamics in Epithelia Group; Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ann-Marie Baker
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Stuart A C McDonald
- Clonal Dynamics in Epithelia Group; Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Quante
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), München, Germany
- Department of Medicine II, Universitaetsklinikum Freiburg, Freiburg, Germany
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
21
|
Acceptability and Adequacy of a Non-endoscopic Cell Collection Device for Diagnosis of Barrett's Esophagus: Lessons Learned. Dig Dis Sci 2022; 67:177-186. [PMID: 33532971 DOI: 10.1007/s10620-021-06833-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/07/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Endoscopic screening for Barrett's esophagus (BE) is common, costly, and underperformed in at-risk people. A non-endoscopic cell collection device can be used to collect esophageal cells, enabling BE screening. AIMS This study assessed the acceptability and adequacy of a commercial non-endoscopic cell collection device in a US population. METHODS Six sites enrolled patients with confirmed BE or heartburn/regurgitation for ≥ 6 months. Patients underwent administration of the device, consisting of a sponge encapsulated in a capsule. The capsule dwelled in the stomach for 7.5 min and was retracted via an attached suture. An adequate sample was ≥ 1 columnar cell by H&E staining. Sample quality was rated using a 0-5 scale, with 0 = no columnar cells and 5 = plentiful groups. Trefoil Factor 3 (TFF3) staining was performed. Accuracy was assessed using esophagogastroduodenoscopy (EGD)/biopsy as the gold standard. RESULTS Of 191 patients, 99.5% successfully swallowed the device. Overall sample adequacy was 91% (171/188), with 84% (158/188) high quality. The detachment rate was 2/190 (1%). Overall sensitivity, specificity, and accuracy of the assay with TFF3 staining were 76%, 77%, and 76%. Sensitivity, specificity, and accuracy for ≥ 3 cm BE were 86%, 77%, and 82%. Asked if willing to repeat the procedure, 93% would, and 65% indicated a preference for the device over EGD. CONCLUSIONS This study demonstrated a high rate of sample adequacy and promising acceptability of this non-endoscopic sampling device in a US population. Diagnostic characteristics suggest that non-endoscopic assessment of BE deserves further development as an alternative to endoscopy.
Collapse
|
22
|
McGowan EC, Aceves SS. Noninvasive tests for eosinophilic esophagitis: Ready for use? Ann Allergy Asthma Immunol 2021; 129:27-34. [PMID: 34706258 DOI: 10.1016/j.anai.2021.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To summarize the existing literature for several promising minimally invasive tests to measure disease activity in eosinophilic esophagitis (EoE). DATA SOURCES Literature searches were performed using PubMed. Keyword combinations included eosinophilic esophagitis and minimally invasive techniques, including the esophageal string test, Cytosponge, transnasal endoscopy, technetium-labeled heparin, and noninvasive biomarkers. STUDY SELECTIONS Retrospective and prospective observational studies, peer-reviewed reviews, and systematic reviews were selected. Data were reviewed and summarized. RESULTS Various techniques have been developed in recent years to measure disease activity in EoE without the need for conventional endoscopy. Our review summarizes the data on these techniques, the benefits and limitations, and future directions for implementation in both research and clinical care. CONCLUSION Tremendous progress has been made toward developing minimally invasive techniques to measure disease activity in EoE. Each of the techniques mentioned in this review has advantages and disadvantages, and some are closer to widespread use than others.
Collapse
Affiliation(s)
- Emily C McGowan
- Division of Allergy and Immunology, University of Virginia School of Medicine, Charlottesville, Virginia; Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Seema S Aceves
- Division of Allergy and Immunology, Departments of Pediatrics and Medicine, University of California San Diego, San Diego, California; Rady Children's Hospital-San Diego, San Diego, California
| |
Collapse
|
23
|
Liang K, Ahsen OO, Murphy A, Zhang J, Nguyen TH, Potsaid B, Figueiredo M, Huang Q, Mashimo H, Fujimoto JG. Tethered capsule en face optical coherence tomography for imaging Barrett's oesophagus in unsedated patients. BMJ Open Gastroenterol 2021; 7:bmjgast-2020-000444. [PMID: 32883714 PMCID: PMC7473663 DOI: 10.1136/bmjgast-2020-000444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/14/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022] Open
Abstract
Objective Barrett’s oesophagus (BE) screening outside the endoscopy suite can identify patients for surveillance and reduce mortality. Tethered capsule optical coherence tomography (OCT) can volumetrically image oesophageal mucosa in unsedated patients and detect features of BE. We investigated ultrahigh-speed tethered capsule swept-source OCT (SS-OCT), improved device design, developed procedural techniques and measured capsule contact, longitudinal pullback non-uniformity and patient toleration. Design OCT was performed in 16 patients prior to endoscopic surveillance/treatment. Unsedated patients swallowed the capsule with sips of water and the tether was pulled back to image the oesophagus. SS-OCT at 1 000 000 A-scans/s enabled imaging 10 cm oesophageal lengths in 10 s with 30 µm transverse and 8 µm axial resolution. Capsule contact, longitudinal image coverage and patient toleration were assessed. Results Nine patients had non-dysplastic BE, three had ablative treatment-naïve neoplasia and four had prior ablation for dysplasia. Dry swallows facilitated capsule transit through the lower oesophageal sphincter (LES), and waiting 10 s before pullback reduced swallow induced LES relaxation. Slow nasal inhalation facilitated capsule retrieval and minimised gag reflex. The procedure was well tolerated. Ultrahigh-speed SS-OCT generated cross-sectional and subsurface en face images showing BE features, while subsurface en face images were required to assess the gastro-oesophageal junction. Candidate features of dysplasia were also identified which could inform follow-up endoscopy/biopsy. BE features were seen in all patients with histologically confirmed BE. Mean capsule contact over BE was 75%±27% for all patients and better in short segment BE. Mean longitudinal image coverage over BE was 59%±34% and better for long segment BE. Conclusions Ultrahigh-speed tethered capsule SS-OCT can image en face and cross-sectional mucosal features over wide areas. Device and procedure optimisation improved performance. BE features could be identified in all patients, but limited capsule contact and longitudinal coverage could cause sampling errors for focal pathologies.
Collapse
Affiliation(s)
- Kaicheng Liang
- Department of Electrical Engineering and Computer Science, and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Osman O Ahsen
- Department of Electrical Engineering and Computer Science, and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Annalee Murphy
- Gastroenterology, VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Jason Zhang
- Department of Electrical Engineering and Computer Science, and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Tan H Nguyen
- Department of Electrical Engineering and Computer Science, and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Benjamin Potsaid
- Department of Electrical Engineering and Computer Science, and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Marisa Figueiredo
- Gastroenterology, VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Qin Huang
- Gastroenterology, VA Boston Healthcare System, Boston, Massachusetts, USA.,Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Hiroshi Mashimo
- Gastroenterology, VA Boston Healthcare System, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - James G Fujimoto
- Department of Electrical Engineering and Computer Science, and Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
24
|
Madabhushi A, Toro P, Willis JE. Artificial Intelligence in Surveillance of Barrett's Esophagus. Cancer Res 2021; 81:3446-3448. [PMID: 34252041 PMCID: PMC9494280 DOI: 10.1158/0008-5472.can-21-1511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 11/16/2022]
Abstract
A study by Waterhouse and colleagues in a previous issue of Cancer Research describes the development and prospective validation of an artificial intelligence approach in conjunction with spectral imaging to enhance endoscopic detection of Barrett's esophagus-related neoplasia. The authors developed a novel spectral endoscope with external optics suitable for routine Barrett's esophagus surveillance with diffuse tissue reflectance to define multispectral data correlated with histopathology. A convolutional neural network was trained on the absis of the spectral signatures acquired as part of a small, prospective clinical trial to distinguish Barrett's esophagus from Barrett's esophagus neoplasia. The results from the study suggest the utility of artificial intelligence for diagnosis of Barrett's esophagus.See related article by Waterhouse et al., Cancer Res 2021;81:3415-25.
Collapse
Affiliation(s)
- Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio.
- Louis Stokes VA Medical Center, Cleveland, Ohio
| | - Paula Toro
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Joseph E Willis
- University Hospitals Cleveland Medical Center, Cleveland, Ohio
- Department of Pathology, Case Western Reserve University, Cleveland Ohio
| |
Collapse
|
25
|
Global burden and epidemiology of Barrett oesophagus and oesophageal cancer. Nat Rev Gastroenterol Hepatol 2021; 18:432-443. [PMID: 33603224 DOI: 10.1038/s41575-021-00419-3] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
Abstract
Oesophageal cancer is a global health problem; in 2018 there were more than 572,000 people newly diagnosed with oesophageal cancer worldwide. There are two main histological subtypes of oesophageal cancer, oesophageal adenocarcinoma (EAC) and oesophageal squamous cell carcinoma (ESCC), and there has been a dramatic shift in its epidemiology. While the incidence of EAC and its precursor lesion, Barrett oesophagus, has increased in Western populations over the past four decades, the incidence of ESCC has declined in most parts of the world over the same period. ESCC still accounts for the vast majority of all oesophageal cancer cases diagnosed worldwide each year. Prognosis for patients with oesophageal cancer is strongly related to stage at diagnosis. As most patients are diagnosed with late-stage disease, overall 5-year survival for oesophageal cancer remains <20%. Knowledge of epidemiology and risk factors for oesophageal cancer is essential for public health and clinical decisions about risk stratification, screening and prevention. The goal of this Review is to establish the current epidemiology of oesophageal cancer, with a particular focus on the Western world and the increasing incidence of EAC and Barrett oesophagus.
Collapse
|
26
|
Yusuf A, Fitzgerald RC. Screening for Barrett's Oesophagus: Are We Ready for it? ACTA ACUST UNITED AC 2021; 19:321-336. [PMID: 33746508 PMCID: PMC7962426 DOI: 10.1007/s11938-021-00342-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 01/10/2023]
Abstract
Purpose of review The targeted approach adopted for Barrett’s oesophagus (BO) screening is sub-optimal considering the large proportion of BO cases that are currently missed. We reviewed the literature highlighting recent technological advancements in efforts to counteract this challenge. We also provided insights into strategies that can improve the outcomes from current BO screening practises. Recent findings The standard method for BO detection, endoscopy, is invasive and expensive and therefore inappropriate for mass screening. On the other hand, endoscopy is more cost-effective for screening a high-risk population. A consensus has however not been reached on who should be screened. Risk prediction algorithms have been tested as an enrichment pre-screening tool reporting modest AUC’s but require more prospective evaluation studies. Less invasive endoscopy methods like trans-nasal endoscopy, oesophageal capsule endsocopy and non-endoscopic cell collection devices like the Cytosponge coupled with biomarker analysis have shown promise in BO detection with randomised clinical trial evidence. Summary A three-tier precision cancer programme whereby risk prediction algorithms and non-endoscopic minimally invasive cell collection devices are used to triage test a wider pool of individuals may improve the detection rate of current screening practises with minimal cost implications.
Collapse
Affiliation(s)
- Aisha Yusuf
- Medical Research Council Cancer Unit, Hutchison/Medical Research Council Research Centre, University of Cambridge, Cambridge, CB2 0XZ United Kingdom
| | - Rebecca C Fitzgerald
- Medical Research Council Cancer Unit, Hutchison/Medical Research Council Research Centre, University of Cambridge, Cambridge, CB2 0XZ United Kingdom
| |
Collapse
|
27
|
Chang JW, Haller E, Dellon ES. Dietary Management of Eosinophilic Esophagitis: Man Versus Food or Food Versus Man? Gastroenterol Clin North Am 2021; 50:59-75. [PMID: 33518169 DOI: 10.1016/j.gtc.2020.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
An alternative to pharmacologic management of eosinophilic esophagitis, elimination of food antigens for diet therapy is an effective first-line treatment strategy to induce and maintain symptomatic, histologic, and endoscopic disease remission. The 3 dietary strategies for eosinophilic esophagitis include elemental diet, empiric elimination diet, and targeted elimination diet. We review the studies supporting various diet therapy strategies, practical considerations and challenges for applying an elimination diet, and novel testing to identify triggers and optimize food reintroduction.
Collapse
Affiliation(s)
- Joy W Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, 3912 Taubman Center, 1500 E. Medical Center Drive, SPC 5362, Ann Arbor, MI 48109, USA.
| | - Emily Haller
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, 3912 Taubman Center, 1500 E. Medical Center Drive, SPC 5362, Ann Arbor, MI 48109, USA
| | - Evan S Dellon
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, CB #7080, Bioinformatics Building, 130 Mason Farm Road, Chapel Hill, NC 27599, USA
| |
Collapse
|
28
|
Middleton DRS, Mmbaga BT, O'Donovan M, Abedi‐Ardekani B, Debiram‐Beecham I, Nyakunga‐Maro G, Maro V, Bromwich M, Daudi A, Ngowi T, Minde R, Claver J, Mremi A, Mwasamwaja A, Schüz J, Fitzgerald RC, McCormack V. Minimally invasive esophageal sponge cytology sampling is feasible in a Tanzanian community setting. Int J Cancer 2021; 148:1208-1218. [PMID: 33128785 PMCID: PMC7839767 DOI: 10.1002/ijc.33366] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/19/2020] [Accepted: 10/02/2020] [Indexed: 12/24/2022]
Abstract
Esophageal sponge cytology is an endoscopy alternative well accepted by patients with extensive data for accuracy in the context of adenocarcinoma. Few studies have assessed its feasibility in asymptomatic community members, and fewer still in East Africa, where esophageal squamous cell carcinoma (ESCC) rates are high. We aimed to assess the feasibility of a capsule-based diagnosis of esophageal squamous dysplasia (ESD), an ESCC precursor, which may benefit epidemiological and early detection research. We collected Cytosponge collections in 102 asymptomatic adults from Kilimanjaro, Tanzania. Uptake, acceptability and safety were assessed. Participants scored acceptability immediately following the procedure and 7 days later on a scale of 0 (least) to 10 (most acceptable). Slides from paraffin-embedded cell clots were read by two pathologists for ESD and other pathologies. All participants (52 men, 50 women, aged 30-77) swallowed the device at first attempt, 100 (98%) of which gave slides of adequate cellularity. Acceptability scores were 10 (53%), 9 (24%), 8 (21%), 7 (2%) and 6 (1%), with no differences by age, sex or time of asking. Cytological findings were esophageal inflammation (4%), atypical squamous cells of uncertain significance (1%), low-grade dysplasia (1%), gastritis (22%) and suspected intestinal metaplasia (6%). Setting-specific logistical and ethical considerations of study implementation are discussed. We demonstrate the safety, acceptability and feasibility of Cytosponge sampling in this setting, paving the way for innovative etiology and early-detection research. Targeted sampling strategies and biomarker development will underpin the success of such initiatives. The study protocol is registered on ClinicalTrials.gov (NCT04090554).
Collapse
Affiliation(s)
- Daniel R. S. Middleton
- Section of Environment and RadiationInternational Agency for Research on Cancer (IARC/WHO)LyonFrance
| | - Blandina T. Mmbaga
- Kilimanjaro Clinical Research InstituteMoshiTanzania
- Kilimanjaro Christian Medical CentreMoshiTanzania
- Kilimanjaro Christian Medical University CollegeMoshiTanzania
| | - Maria O'Donovan
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Behnoush Abedi‐Ardekani
- Genetic Cancer Susceptibility Group, Section of GeneticsInternational Agency for Research on Cancer (IARC/WHO)LyonFrance
| | | | - Gissela Nyakunga‐Maro
- Kilimanjaro Clinical Research InstituteMoshiTanzania
- Kilimanjaro Christian Medical CentreMoshiTanzania
- Kilimanjaro Christian Medical University CollegeMoshiTanzania
| | - Venance Maro
- Kilimanjaro Christian Medical CentreMoshiTanzania
- Kilimanjaro Christian Medical University CollegeMoshiTanzania
| | - Martin Bromwich
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Amini Daudi
- Kilimanjaro Clinical Research InstituteMoshiTanzania
- Kilimanjaro Christian Medical CentreMoshiTanzania
| | - Timothy Ngowi
- Kilimanjaro Clinical Research InstituteMoshiTanzania
| | - Rehema Minde
- Kilimanjaro Christian Medical CentreMoshiTanzania
| | | | - Alex Mremi
- Kilimanjaro Christian Medical CentreMoshiTanzania
- Kilimanjaro Christian Medical University CollegeMoshiTanzania
| | - Amos Mwasamwaja
- Kilimanjaro Clinical Research InstituteMoshiTanzania
- Kilimanjaro Christian Medical CentreMoshiTanzania
- Kilimanjaro Christian Medical University CollegeMoshiTanzania
| | - Joachim Schüz
- Section of Environment and RadiationInternational Agency for Research on Cancer (IARC/WHO)LyonFrance
| | | | - Valerie McCormack
- Section of Environment and RadiationInternational Agency for Research on Cancer (IARC/WHO)LyonFrance
| |
Collapse
|
29
|
Weke K, Singh A, Uwugiaren N, Alfaro JA, Wang T, Hupp TR, O'Neill JR, Vojtesek B, Goodlett DR, Williams SM, Zhou M, Kelly RT, Zhu Y, Dapic I. MicroPOTS Analysis of Barrett's Esophageal Cell Line Models Identifies Proteomic Changes after Physiologic and Radiation Stress. J Proteome Res 2021; 20:2195-2205. [PMID: 33491460 PMCID: PMC8155554 DOI: 10.1021/acs.jproteome.0c00629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Moving from macroscale
preparative systems in proteomics to micro-
and nanotechnologies offers researchers the ability to deeply profile
smaller numbers of cells that are more likely to be encountered in
clinical settings. Herein a recently developed microscale proteomic
method, microdroplet processing in one pot for trace samples (microPOTS),
was employed to identify proteomic changes in ∼200 Barrett’s
esophageal cells following physiologic and radiation stress exposure.
From this small population of cells, microPOTS confidently identified
>1500 protein groups, and achieved a high reproducibility with
a Pearson’s
correlation coefficient value of R > 0.9 and over
50% protein overlap from replicates. A Barrett’s cell line
model treated with either lithocholic acid (LCA) or X-ray had 21 (e.g.,
ASNS, RALY, FAM120A, UBE2M, IDH1, ESD) and 32 (e.g., GLUL, CALU, SH3BGRL3,
S100A9, FKBP3, AGR2) overexpressed proteins, respectively, compared
to the untreated set. These results demonstrate the ability of microPOTS
to routinely identify and quantify differentially expressed proteins
from limited numbers of cells.
Collapse
Affiliation(s)
- Kenneth Weke
- University of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, Poland
| | - Ashita Singh
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K.,Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Naomi Uwugiaren
- University of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, Poland
| | - Javier A Alfaro
- University of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, Poland.,Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K
| | - Tongjie Wang
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K
| | - Ted R Hupp
- University of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, Poland.,Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K
| | - J Robert O'Neill
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K.,Cambridge Oesophagogastric Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, U.K
| | - Borek Vojtesek
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - David R Goodlett
- University of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, Poland.,University of Victoria - Genome British Columbia Proteomics Centre, Victoria, BC V8Z 7X8, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Sarah M Williams
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Mowei Zhou
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Ryan T Kelly
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Ying Zhu
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Irena Dapic
- University of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, Poland
| |
Collapse
|
30
|
Struyvenberg MR, de Groof AJ, Bergman JJ, van der Sommen F, de With PHN, Konda VJA, Curvers WL. Advanced Imaging and Sampling in Barrett's Esophagus: Artificial Intelligence to the Rescue? Gastrointest Endosc Clin N Am 2021; 31:91-103. [PMID: 33213802 DOI: 10.1016/j.giec.2020.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Because the current Barrett's esophagus (BE) surveillance protocol suffers from sampling error of random biopsies and a high miss-rate of early neoplastic lesions, many new endoscopic imaging and sampling techniques have been developed. None of these techniques, however, have significantly increased the diagnostic yield of BE neoplasia. In fact, these techniques have led to an increase in the amount of visible information, yet endoscopists and pathologists inevitably suffer from variations in intra- and interobserver agreement. Artificial intelligence systems have the potential to overcome these endoscopist-dependent limitations.
Collapse
Affiliation(s)
- Maarten R Struyvenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Albert J de Groof
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Jacques J Bergman
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Fons van der Sommen
- Department of Electrical Engineering, VCA group, Eindhoven University of Technology, Groene Loper 19, 5612 AP Eindhoven, the Netherlands
| | - Peter H N de With
- Department of Electrical Engineering, VCA group, Eindhoven University of Technology, Groene Loper 19, 5612 AP Eindhoven, the Netherlands
| | - Vani J A Konda
- Department of Gastroenterology and Hepatology, Baylor University Medical Center, 3500 Gaston Ave, Dallas, TX 75246, USA
| | - Wouter L Curvers
- Department of Gastroenterology and Hepatology, Catharina Hospital Eindhoven, Michelangelolaan 2, 5623 EJ Eindhoven, the Netherlands.
| |
Collapse
|
31
|
Progress in Screening for Barrett's Esophagus: Beyond Standard Upper Endoscopy. Gastrointest Endosc Clin N Am 2021; 31:43-58. [PMID: 33213799 DOI: 10.1016/j.giec.2020.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The rapid increase in the incidence of esophageal adenocarcinoma in Western populations over the past 4 decades and its associated poor prognosis, unless detected early has generated great interest in screening for the precursor lesion Barrett's esophagus (BE). Recently, there have been significant developments in imaging-based modalities and esophageal cell-sampling devices coupled with biomarker assays. In this review, the authors discuss the rationale for screening for BE and the factors to consider for targeting the at-risk population. They also explore future avenues for research in this area.
Collapse
|
32
|
Salta S, Macedo-Silva C, Miranda-Gonçalves V, Lopes N, Gigliano D, Guimarães R, Farinha M, Sousa O, Henrique R, Jerónimo C. A DNA methylation-based test for esophageal cancer detection. Biomark Res 2020; 8:68. [PMID: 33292587 PMCID: PMC7691099 DOI: 10.1186/s40364-020-00248-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022] Open
Abstract
Background Esophageal cancer (ECa) is the 7th most incident cancer and the 6th leading cause of cancer-related death. Most patients are diagnosed with locally advanced or metastatic disease, enduring poor survival. Biomarkers enabling early cancer detection may improve patient management, treatment effectiveness, and survival, are urgently needed. In this context, epigenetic-based biomarkers such as DNA methylation are potential candidates. Methods Herein, we sought to identify and validate DNA methylation-based biomarkers for early detection and prediction of response to therapy in ECa patients. Promoter methylation levels were assessed in a series of treatment-naïve ECa, post-neoadjuvant treatment ECa, and normal esophagus tissues, using quantitative methylation-specific PCR for COL14A1, GPX3, and ZNF569. Results ZNF569 methylation (ZNF569me) levels significantly differed between ECa and normal samples (p < 0.001). Moreover, COL14A1 methylation (COL14A1me) and GPX3 methylation (GPX3me) levels discriminated adenocarcinomas and squamous cell carcinomas, respectively, from normal samples (p = 0.002 and p = 0.009, respectively). COL14A1me & ZNF569me accurately identified adenocarcinomas (82.29%) whereas GPX3me & ZNF569me identified squamous cell carcinomas with 81.73% accuracy. Furthermore, ZNF569me and GPX3me levels significantly differed between normal and pre-treated ECa. Conclusion The biomarker potential of a specific panel of methylated genes for ECa was confirmed. These might prove useful for early detection and might allow for the identification of minimal residual disease after adjuvant therapy.
Collapse
Affiliation(s)
- Sofia Salta
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Catarina Macedo-Silva
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Vera Miranda-Gonçalves
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Nair Lopes
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Davide Gigliano
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal
| | - Rita Guimarães
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal
| | - Mónica Farinha
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal
| | - Olga Sousa
- Department of Radiation Oncology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal
| | - Rui Henrique
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar- University of Porto , Rua de Jorge Viterbo Ferreira, 228, Porto, 4050-313, Portugal
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal. .,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar- University of Porto , Rua de Jorge Viterbo Ferreira, 228, Porto, 4050-313, Portugal.
| |
Collapse
|
33
|
Practical early cancer detection: distinguishing stable from unstable genomes in pre-cancerous tissues. Br J Cancer 2020; 124:683-685. [PMID: 33154569 PMCID: PMC7884797 DOI: 10.1038/s41416-020-01142-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 09/24/2020] [Accepted: 10/16/2020] [Indexed: 01/23/2023] Open
Abstract
Barrett’s oesophagus has been known for many years to display early changes to the genome consistent with the risk for oesophageal adenocarcinoma. Recently we have shown that this information can be used without knowledge of individual gene mutations to accurately predict a patient’s future risk of malignant progression.
Collapse
|
34
|
Amano Y, Ishimura N, Ishihara S. Is Malignant Potential of Barrett's Esophagus Predictable by Endoscopy Findings? Life (Basel) 2020; 10:E244. [PMID: 33081277 PMCID: PMC7602941 DOI: 10.3390/life10100244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/01/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Given that endoscopic findings can be used to predict the potential of neoplastic progression in Barrett's esophagus (BE) cases, the detection rate of dysplastic Barrett's lesions may become higher even in laborious endoscopic surveillance because a special attention is consequently paid. However, endoscopic findings for effective detection of the risk of neoplastic progression to esophageal adenocarcinoma (EAC) have not been confirmed, though some typical appearances are suggestive. In the present review, endoscopic findings that can be used predict malignant potential to EAC in BE cases are discussed. Conventional results obtained with white light endoscopy, such as length of BE, presence of esophagitis, ulceration, hiatal hernia, and nodularity, are used as indicators of a higher risk of neoplastic progression. However, there are controversies in some of those findings. Absence of palisade vessels may be also a new candidate predictor, as that reveals degree of intense inflammation and of cyclooxygenase-2 protein expression with accelerated cellular proliferation. Furthermore, an open type of mucosal pattern and enriched stromal blood vessels, which can be observed by image-enhanced endoscopy, including narrow band imaging, have been confirmed as factors useful for prediction of neoplastic progression of BE because they indicate more frequent cyclooxygenase-2 protein expression along with accelerated cellular proliferation. Should the malignant potential of BE be shown predictable by these endoscopic findings, that would simplify methods used for an effective surveillance, because patients requiring careful monitoring would be more easily identified. Development in the near future of a comprehensive scoring system for BE based on clinical factors, biomarkers and endoscopic predictors is required.
Collapse
Affiliation(s)
- Yuji Amano
- Department of Endoscopy, New Tokyo Hospital, 1271 Wanagaya, Matsudo, Chiba 270-2232, Japan
| | - Norihisa Ishimura
- Department of Internal Medicine II, Faculty of Medicine, Shimane University, Shimane 693-8501, Japan; (N.I.); (S.I.)
| | - Shunji Ishihara
- Department of Internal Medicine II, Faculty of Medicine, Shimane University, Shimane 693-8501, Japan; (N.I.); (S.I.)
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
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: 139] [Impact Index Per Article: 27.8] [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
|
38
|
Abstract
Because of the rising incidence and lethality of esophageal adenocarcinoma, Barrett's esophagus (BE) is an increasingly important premalignant target for cancer prevention. BE-associated neoplasia can be safely and effectively treated with endoscopic eradication therapy (EET), incorporating tissue resection and ablation. Because EET has proliferated, managing patients after complete eradication of intestinal metaplasia has taken on increasing importance. Recurrence after complete eradication of intestinal metaplasia occurs in 8%-10% of the patients yearly, and the incidence may remain constant over time. Most recurrences occur at the gastroesophageal junction, whereas those in the tubular esophagus are endoscopically visible and distally located. A simplified biopsy protocol limited to the distal aspect of the BE segment, in addition to gastroesophageal junction sampling, may enhance efficiency and cost without significantly reducing recurrence detection. Similarly, research suggests that current surveillance intervals may be excessively frequent, failing to reflect the cancer risk reduction of EET. If validated, longer surveillance intervals could reduce the burden of resource-intensive endoscopic surveillance. Several important questions in post-EET management remain unanswered, including surveillance duration, the significance of gastric cardia intestinal metaplasia, and the role of advanced imaging and nonendoscopic sampling techniques in detecting recurrence. These merit further research to enhance quality of care and promote a more evidence-based approach.
Collapse
|
39
|
Ungerer V, Bronkhorst AJ, Holdenrieder S. Preanalytical variables that affect the outcome of cell-free DNA measurements. Crit Rev Clin Lab Sci 2020; 57:484-507. [DOI: 10.1080/10408363.2020.1750558] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Vida Ungerer
- Institute for Laboratory Medicine, German Heart Centre, Technical University Munich, Munich, Germany
| | - Abel J. Bronkhorst
- Institute for Laboratory Medicine, German Heart Centre, Technical University Munich, Munich, Germany
| | - Stefan Holdenrieder
- Institute for Laboratory Medicine, German Heart Centre, Technical University Munich, Munich, Germany
| |
Collapse
|
40
|
Rosenfeld A, Graham DG, Jevons S, Ariza J, Hagan D, Wilson A, Lovat SJ, Sami SS, Ahmad OF, Novelli M, Rodriguez Justo M, Winstanley A, Heifetz EM, Ben-Zecharia M, Noiman U, Fitzgerald RC, Sasieni P, Lovat LB. Development and validation of a risk prediction model to diagnose Barrett's oesophagus (MARK-BE): a case-control machine learning approach. Lancet Digit Health 2020; 2:E37-E48. [PMID: 32133440 PMCID: PMC7056359 DOI: 10.1016/s2589-7500(19)30216-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background Screening for Barrett's Oesophagus (BE) relies on endoscopy which is invasive and has a low yield. This study aimed to develop and externally validate a simple symptom and risk-factor questionnaire to screen for patients with BE. Methods Questionnaires from 1299 patients in the BEST2 case-controlled study were analysed: 880 had BE including 40 with invasive oesophageal adenocarcinoma (OAC) and 419 were controls. This was randomly split into a training cohort of 776 patients and an internal validation cohort of 523 patients. External validation included 398 patients from the BOOST case-controlled study: 198 with BE (23 with OAC) and 200 controls. Identification of independently important diagnostic features was undertaken using machine learning techniques information gain (IG) and correlation based feature selection (CFS). Multiple classification tools were assessed to create a multi-variable risk prediction model. Internal validation was followed by external validation in the independent dataset. Findings The BEST2 study included 40 features. Of these, 24 added IG but following CFS, only 8 demonstrated independent diagnostic value including age, gender, smoking, waist circumference, frequency of stomach pain, duration of heartburn and acid taste and taking of acid suppression medicines. Logistic regression offered the highest prediction quality with AUC (area under the receiver operator curve) of 0.87. In the internal validation set, AUC was 0.86. In the BOOST external validation set, AUC was 0.81. Interpretation The diagnostic model offers valid predictions of diagnosis of BE in patients with symptomatic gastroesophageal reflux, assisting in identifying who should go forward to invasive testing. Overweight men who have been taking stomach medicines for a long time may merit particular consideration for further testing. The risk prediction tool is quick and simple to administer but will need further calibration and validation in a prospective study in primary care. Funding Charles Wolfson Trust and Guts UK.
Collapse
Affiliation(s)
- Avi Rosenfeld
- Department of Industrial Engineering Jerusalem College of Technology (JCT), Jerusalem, Israel
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
| | - David G Graham
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
- Gastrointestinal Services, University College London Hospital (UCLH), London, United Kingdom
| | - Sarah Jevons
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
| | - Jose Ariza
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
- Gastrointestinal Services, University College London Hospital (UCLH), London, United Kingdom
| | - Daryl Hagan
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
| | - Ash Wilson
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
| | - Samuel J Lovat
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
| | - Sarmed S Sami
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
- Gastrointestinal Services, University College London Hospital (UCLH), London, United Kingdom
| | - Omer F Ahmad
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
- Gastrointestinal Services, University College London Hospital (UCLH), London, United Kingdom
| | - Marco Novelli
- Dept of Pathology, University College London Hospital (UCLH), London, United Kingdom
| | | | - Alison Winstanley
- Dept of Pathology, University College London Hospital (UCLH), London, United Kingdom
| | - Eliyahu M Heifetz
- Department of Health Informatics, Jerusalem College of Technology (JCT), Jerusalem, Israel
| | - Mordehy Ben-Zecharia
- Department of Health Informatics, Jerusalem College of Technology (JCT), Jerusalem, Israel
| | - Uria Noiman
- Department of Health Informatics, Jerusalem College of Technology (JCT), Jerusalem, Israel
| | | | - Peter Sasieni
- Cancer Prevention Trials Unit, Queen Mary University of London, London, United Kingdom
- School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Laurence B Lovat
- GENIE GastroENterological IntervEntion Group, Department for Targeted Intervention, University College London (UCL), London, United Kingdom
- Gastrointestinal Services, University College London Hospital (UCLH), London, United Kingdom
| |
Collapse
|
41
|
Petrillo A, Smyth EC. 27 years of stomach cancer: painting a global picture. Lancet Gastroenterol Hepatol 2020; 5:5-6. [DOI: 10.1016/s2468-1253(19)30357-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/26/2022]
|
42
|
Porter RJ, Murray GI, Brice DP, Petty RD, McLean MH. Novel biomarkers for risk stratification of Barrett's oesophagus associated neoplastic progression-epithelial HMGB1 expression and stromal lymphocytic phenotype. Br J Cancer 2019; 122:545-554. [PMID: 31831860 PMCID: PMC7028982 DOI: 10.1038/s41416-019-0685-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 10/30/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The incidence of oesophageal adenocarcinoma is increasing globally. Barrett's oesophagus (BO) is a pre-malignant condition with no biomarker to risk stratify those at highest risk of dysplasia and malignant transformation. METHODS Subcellular epithelial protein (HMGB1, p53, RUNX3) expression, alongside expression of CD20, CD4, CD8 and Foxp3 to characterise stromal B lymphocyte, and helper, cytotoxic and regulatory T-lymphocyte cell infiltrate, respectively, was assessed by immunohistochemistry in 218 human tissue samples including normal oesophageal/gastric biopsies (n = 39), BO (non-dysplasia, dysplasia, non-dysplastic background from progressors to dysplasia or cancer, n = 121) and oesophageal adenocarcinoma (n = 58). RESULTS There is a dynamic subcellular epithelial expression of HMGB1 (loss of nuclear, emergence of cytoplasmic), associated with epithelial p53 expression and differential immune cell phenotype in oesophageal neoplastic progression. We identify a protein signature and lymphocyte infiltrate in non-dysplastic BO when progressive disease (dysplasia or adenocarcinoma) is present but not histologically represented in the biopsied field. There is a dynamic stromal lymphocytic infiltrate in oesophageal neoplastic progression. CONCLUSIONS This data reveals novel insights into the microenvironment of BO and progression towards cancer and identifies a novel high-risk biomarker of disease progression to aid surveillance strategies to identify early progression and impact future incidence of oesophageal cancer.
Collapse
Affiliation(s)
- Ross J Porter
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Graeme I Murray
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Daniel P Brice
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Russell D Petty
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, DD1 1GZ, UK
| | - Mairi H McLean
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
43
|
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
|
44
|
Singh T, Sanghi V, Thota PN. Current management of Barrett esophagus and esophageal adenocarcinoma. Cleve Clin J Med 2019; 86:724-732. [PMID: 31710585 DOI: 10.3949/ccjm.86a.18106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Barrett esophagus is found in 5% to 15% of patients with gastroesophageal reflux disease and is a precursor of esophageal adenocarcinoma, yet the condition often goes undiagnosed. Patients with reflux disease who are male, over age 50, or white, and who smoke or have central obesity or a family history of Barrett esophagus or esophageal adenocarcinoma, should undergo initial screening endoscopy and, if no dysplasia is noted, surveillance endoscopy every 3 to 5 years. Dysplasia is treated with endoscopic eradication by ablation, resection, or both. Chemoprotective agents are being studied to prevent progression to dysplasia in Barrett esophagus. The authors discuss current recommendations for screening and management.
Collapse
Affiliation(s)
- Tavankit Singh
- Department of Gastroenterology and Hepatology, Cleveland Clinic
| | - Vedha Sanghi
- Department of Internal Medicine, Cleveland Clinic; Clinical Instructor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Prashanthi N Thota
- Medical Director, Esophageal Center, Digestive Disease and Surgery Institute, Cleveland Clinic; Clinical Assistant Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| |
Collapse
|
45
|
Pech O. Screening and Prevention of Barrett's Esophagus. Visc Med 2019; 35:210-214. [PMID: 31602381 PMCID: PMC6738192 DOI: 10.1159/000501918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/03/2019] [Indexed: 12/20/2022] Open
Abstract
Barrett's esophagus (BO) is a risk factor for esophageal adenocarcinoma. However, screening for BO is difficult since it is not yet clear who should be screened and which screening method is cost-effective. Screening methods could be upper endoscopy at the time of the first screening colonoscopy, transnasal endoscopy, esophageal capsule endoscopy, or cytosponge. In order to prevent the development of BO or its neoplastic progression, there are modifiable risk factors like obesity or smoking that can be influenced. In addition, several drugs like proton pump inhibitors, aspirin, nonsteroidal anti-inflammatory drugs and statins have shown promising effects in mostly observational studies. However, data from prospective randomized trials are scarce in order to draw final conclusions.
Collapse
Affiliation(s)
- Oliver Pech
- Department of Gastroenterology and Interventional Endoscopy, Krankenhaus Barmherzige Brüder, Regensburg, Germany
| |
Collapse
|
46
|
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
|
47
|
Affiliation(s)
- Sri G Thrumurthy
- Department of Surgery, Epsom and St Helier University Hospitals, Sutton SM5 1AA, UK
- Department of Surgery, Royal Marsden Hospital, London SW3 6JJ, UK
| | - M Asif Chaudry
- Department of Surgery, Royal Marsden Hospital, London SW3 6JJ, UK
| | | | - Muntzer Mughal
- Department of Surgery, University College Hospital London, London, UK
| |
Collapse
|
48
|
Cell-Free DNA in the Liquid Biopsy Context: Role and Differences Between ctDNA and CTC Marker in Cancer Management. Methods Mol Biol 2019; 1909:47-73. [PMID: 30580422 DOI: 10.1007/978-1-4939-8973-7_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Liquid biopsy is a new diagnostic concept to investigate the molecular features of solid tumors by blood, saliva, urine, and any other body fluids which show a source of potential biomarkers. In cancer patients, it is a simple and less invasive mean, representing a sustainable alternative to interrogate all tumor cells longitudinally, quantifying and characterizing the biological materials (DNAs, RNAs, proteins) which originate from cancer tissues. Circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) analysis from a simple blood draw received enormous attention for the related clinical research results. A rich scientific literature demonstrates that liquid biopsy is a valid instrument to assess the tumor biomarkers in real time and profile the cancer genotype in diagnostic and prognostic field, as well to quantify minimal residual disease, during patient follow-up. This could be a breakthrough for a companion diagnostic and personalized medicine. Liquid biopsy needs further implementation in the methodological aspects as well as cost-based assessment. The number of new molecular diagnostic assays increases day by day, but the standards for their adoption and clinical validation are still to be achieved.
Collapse
|
49
|
Shah AK, Joshi V, Hartel G, Barbour AP, Hill MM. To BE or not to BE: non-invasive screening for Barrett's esophagus, dysplasia and adenocarcinoma. Transl Gastroenterol Hepatol 2019; 4:31. [PMID: 31231698 PMCID: PMC6556700 DOI: 10.21037/tgh.2019.04.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 04/28/2019] [Indexed: 01/03/2023] Open
Affiliation(s)
- Alok K. Shah
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Virendra Joshi
- Advanced Digestive Institute, University Medical Center (UMC), New Orleans, Louisiana, USA
| | - Gunter Hartel
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andrew P. Barbour
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Michelle M. Hill
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
50
|
Fitzgerald RC, Corley DA. Will a Proton Pump Inhibitor and an Aspirin Keep the Doctor Away for Patients With Barrett's Esophagus? Gastroenterology 2019; 156:1228-1231. [PMID: 30849313 DOI: 10.1053/j.gastro.2019.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | - Douglas A Corley
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| |
Collapse
|