1
|
Yan L, He Q, Verma SP, Zhang X, Giel AS, Maj C, Graz K, Naderi E, Chen J, Ali MW, Gharahkhani P, Shu X, Offit K, Shah PM, Gerdes H, Molena D, Srivastava A, MacGregor S, Palles C, Thieme R, Vieth M, Gockel I, Vaughan TL, Schumacher J, Buas MF. Biologically targeted discovery-replication scan identifies G×G interaction in relation to risk of Barrett's esophagus and esophageal adenocarcinoma. HGG ADVANCES 2025; 6:100399. [PMID: 39755942 PMCID: PMC11815673 DOI: 10.1016/j.xhgg.2025.100399] [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: 08/19/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025] Open
Abstract
Inherited genetics represents an important contributor to risk of esophageal adenocarcinoma (EAC), and its precursor Barrett's esophagus (BE). Genome-wide association studies have identified ∼30 susceptibility variants for BE/EAC, yet genetic interactions remain unexamined. To address challenges in large-scale G×G scans, we combined knowledge-guided filtering and machine learning approaches, focusing on genes with (1) known/plausible links to BE/EAC pathogenesis (n = 493) or (2) prior evidence of biological interactions (n = 4,196). Approximately 75 × 106 SNP×SNP interactions were screened via hierarchical group lasso (glinternet) using BEACON GWAS data. The top ∼2,000 interactions retained in each scan were prioritized using p values from single logistic models. Identical scans were repeated among males only (78%), with two independent GWAS datasets used for replication. In overall and male-specific primary replications, 11 of 187 and 20 of 191 interactions satisfied p < 0.05, respectively. The strongest evidence for secondary replication was for rs17744726×rs3217992 among males, with consistent directionality across all cohorts (Pmeta = 2.19 × 10-8); rs3217992 "T" was associated with reduced risk only in individuals homozygous for rs17744726 "G." Rs3217992 maps to the CDKN2B 3' UTR and reportedly disrupts microRNA-mediated repression. Rs17744726 maps to an intronic enhancer region in BLK. Through in silico prioritization and experimental validation, we identified a nearby proxy variant (rs4841556) as a functional modulator of enhancer activity. Enhancer-gene mapping and eQTLs implicated BLK and FAM167A as targets. The first systematic G×G investigation in BE/EAC, this study uncovers differential risk associations for CDKN2B variation by BLK genotype, suggesting novel biological dependency between two risk loci encoding key mediators of tumor suppression and inflammation.
Collapse
Affiliation(s)
- Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Qianchuan He
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shiv P Verma
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xu Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ann-Sophie Giel
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - Carlo Maj
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - Kathryn Graz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elnaz Naderi
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jianhong Chen
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mourad Wagdy Ali
- Department of Genome Sciences, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Puya Gharahkhani
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Xiang Shu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth Offit
- Clinical Genetics, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pari M Shah
- Gastroenterology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hans Gerdes
- Gastroenterology and Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniela Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amitabh Srivastava
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Claire Palles
- Department of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - René Thieme
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Michael Vieth
- Institute of Pathology, Friedrich-Alexander-Universiät Erlangen-Nürnberg, Klinikum Bayreuth, Bayreuth, Germany
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Thomas L Vaughan
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Epidemiology, University of Washington, School of Public Health, Seattle, WA, USA
| | | | - Matthew F Buas
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2
|
Bhatt A, Zaidi HM, Maitra R, Goel S. Infectious Agents and Esophageal Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:1248. [PMID: 40227819 PMCID: PMC11988037 DOI: 10.3390/cancers17071248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Esophageal cancer, primarily comprising the squamous cell carcinoma (ESCC) and adenocarcinoma (EAC) subtypes, is the sixth leading cause of cancer deaths globally. In addition to many well-established endogenous and exogenous risk factors, there is emerging evidence for the etiologic role of infectious agents in esophageal cancer, although these associations are incompletely understood. Here, we review the currently available literature on the relationship between infectious agents and esophageal cancer. By far, human papilloma virus (HPV), particularly HPV 16 and 18, have the strongest etiologic association with ESCC. Less robust is the association of high-risk HPV (hr-HPV) with EAC. Although H. pylori has been implicated in the development of EAC via increased acid reflux, decreased lower esophageal sphincter tone, and the resultant Barrett's metaplasia-dysplasia-adenocarcinoma pathway, some hypothesize based on epidemiological trends that H. pylori may in fact be a protective factor. In rare cases, EBV can cause esophageal lymphoepithelial carcinoma. Several other agents including HSV, polyomaviruses, and Candida are associated with esophageal cancer to varying degrees. In summary, while several studies, including those conflicting with each other, implicate several infectious agents, the evidence is weak, at best. Clearly, further work is needed to help solidify clear etiologies that will help facilitate prevention and treatment.
Collapse
Affiliation(s)
- Ahan Bhatt
- Jacobi Medical Center, Bronx, NY 10461, USA;
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hasan Musanna Zaidi
- Robert Wood Johnson University Hospital, New Brunswick, NJ 08901, USA;
- Department of Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Radhashree Maitra
- Department of Biology, Yeshiva University, Bronx, NY 10461, USA;
- Department of Oncology, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Sanjay Goel
- Robert Wood Johnson University Hospital, New Brunswick, NJ 08901, USA;
- Department of Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Division of Medical Oncology, Rutgers Cancer Institute, New Brunswick, NJ 08901, USA
| |
Collapse
|
3
|
Baik YS, Lee H, Kim YJ, Chung JW, Kim KG. Early detection of esophageal cancer: Evaluating AI algorithms with multi-institutional narrowband and white-light imaging data. PLoS One 2025; 20:e0321092. [PMID: 40184395 PMCID: PMC11970661 DOI: 10.1371/journal.pone.0321092] [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: 10/21/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025] Open
Abstract
Esophageal cancer is one of the most common cancers worldwide, especially esophageal squamous cell carcinoma, which is often diagnosed at a late stage and has a poor prognosis. This study aimed to develop an algorithm to detect tumors in esophageal endoscopy images using innovative artificial intelligence (AI) techniques for early diagnosis and detection of esophageal cancer. We used white light and narrowband imaging data collected from Gachon University Gil Hospital, and applied YOLOv5 and RetinaNet detection models to detect lesions. The models demonstrated high performance, with RetinaNet achieving a precision of 98.4% and sensitivity of 91.3% in the NBI dataset, and YOLOv5 attaining a precision of 93.7% and sensitivity of 89.9% in the WLI dataset. The generalizability of these models was further validated using external data from multiple institutions. This study demonstrates an effective method for detecting esophageal tumors through AI-based esophageal endoscopic image analysis. These efforts are expected to significantly reduce misdiagnosis rates, enhance the effective diagnosis and treatment of esophageal cancer, and promote the standardization of medical services.
Collapse
Affiliation(s)
- Young Seo Baik
- Department of Biomedical Engineering, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hannah Lee
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Young Jae Kim
- Department of Gachon Biomedical & Convergence Institute, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Jun-Won Chung
- Division of Gastroenterology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Kwang Gi Kim
- Department of Biomedical Engineering, College of IT Convergence, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
4
|
Zhang T, Tang X. Untangling immune cell contributions in the progression from GERD to Barrett's esophagus and esophageal cancer: Insights from genetic causal analysis. Int Immunopharmacol 2025; 150:114271. [PMID: 39965389 DOI: 10.1016/j.intimp.2025.114271] [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: 12/09/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) is a rapidly increasing malignancy with significant morbidity and mortality. The progression from gastroesophageal reflux disease (GERD) to Barrett's esophagus (BE) and ultimately to EAC is thought to be influenced by chronic inflammation and immune cell dynamics. Despite the observed correlations in observational studies, the causal relationships between immune cell phenotypes and this disease continuum remain unclear. METHODS This study utilized a two-sample Mendelian Randomization (MR) approach to investigate the causal roles of 731 distinct immune cell phenotypes in the GERD-BE-EAC continuum. The analysis leveraged genome-wide association study (GWAS) data for immune phenotypes from a Sardinian cohort and data for GERD, BE, and EAC from the FinnGen and Open GWAS databases. A comprehensive set of MR methods, including inverse variance weighted (IVW), MR-Egger, and weighted median estimators, was employed to assess causality. Sensitivity analyses were conducted to evaluate heterogeneity and pleiotropy, ensuring the robustness of the findings. RESULTS The study revealed complex and multifaceted roles of immune cells across the GERD-BE-EAC continuum. In GERD, 34 immune phenotypes were found to be causally associated with either increased or decreased risk. Protective effects were observed in phenotypes such as Unswitched Memory B cells, while others like CD45RA- CD4+ T cells were linked to an elevated risk. In the context of BE, 28 immune phenotypes demonstrated significant causal associations, with the majority being protective, including Unswitched Memory B cells and CD62L on Granulocytes. Conversely, certain phenotypes, such as CD24 on Transitional B cells, were identified as risk factors for BE. For EAC, 34 immune phenotypes were implicated, with various B cell subsets, particularly those expressing BAFF-R and CD24, associated with an increased risk, while Switched Memory B cells and specific myeloid cell phenotypes showed protective effects. CONCLUSIONS This study provides novel insights into the complex role of immune cells in the pathogenesis of EAC, revealing a dynamic interplay where certain immune phenotypes may be protective in early stages but become risk-enhancing in later stages of disease progression. These findings highlight the potential of immune cell phenotypes to serve as biomarkers for early detection and targeted therapeutic interventions across the GERD-BE-EAC continuum. Further research is warranted to validate these findings in diverse populations and to explore the underlying mechanisms driving these immune-mediated effects.
Collapse
Affiliation(s)
- Tai Zhang
- Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University Health Science Center, Beijing 100091, China; Peking University Health Science Center, Beijing 100191, China
| | - Xudong Tang
- Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University Health Science Center, Beijing 100091, China; Institute of Digestive Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
5
|
Akhtar S, Al-Shammari A, Al-Huraiti M, Al-Anjery F. Age-period-cohort modeling of oesophageal carcinoma risk in a middle eastern country: 1980-2019. J Public Health (Oxf) 2025; 47:e59-e66. [PMID: 39674677 DOI: 10.1093/pubmed/fdae311] [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: 08/05/2024] [Accepted: 11/29/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Understanding of the factors influencing oesophageal cancer trends is crucial. Therefore, this cross-sectional cohort study sought to disentangle the age, period and cohort effects on the trends of oesophageal cancer in Kuwait. METHODS The data on incident oesophageal carcinoma cases diagnosed between January 1, 1980, through December 31, 2019, and reference population were obtained. Age-period-cohort (APC) analysis was conducted using a loglinear Poisson regression model. RESULTS A total of 496 oesophageal carcinoma cases in 12.8 million person-years (i.e. squamous-cell carcinoma, 269, 54.23%), adenocarcinoma,147, 29.64% and unspecified cases, 80,16.13%) were diagnosed. The overall age-standardized incidence rate (per 105 person-years) of oesophageal carcinoma during the study period was 10.51 (95% CI: 6.62-14.41). The APC analysis results showed that the age and birth cohort effects were the significant determinants of declining, and subsequently steadying the oesophageal carcinoma incidence rates. CONCLUSIONS A substantial decline in oesophageal carcinoma incidence rates was recorded, which significantly varied in all three temporal dimensions. The observed birth cohort patterns suggest changing lifestyle and dietary patterns seem to be responsible for decreasing oesophageal carcinoma risk in Kuwait. Future studies may look for the component causes maintaining the endemicity of oesophageal carcinoma risk in this and similar countries in the region.
Collapse
Affiliation(s)
- Saeed Akhtar
- Department of Community Medicine and Behavioural Sciences, College of Medicine, Kuwait University, PO Box 24923, Safat 13110, Kuwait
| | - Ahmad Al-Shammari
- Department of Surgery, Faculty of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | | | - Fouzan Al-Anjery
- Ministry of Health, Jamal Abdel Nasser Street, Sulaibkhat 13001, Kuwait
| |
Collapse
|
6
|
Miyoshi J, Mannucci A, Scarpa M, Gao F, Toden S, Whitsett T, Inge LJ, Bremner RM, Takayama T, Cheng Y, Bottiglieri T, Nagtegaal ID, Shrubsole MJ, Zaidi AH, Wang X, Coleman HG, Anderson LA, Meltzer SJ, Goel A. Liquid biopsy to identify Barrett's oesophagus, dysplasia and oesophageal adenocarcinoma: the EMERALD multicentre study. Gut 2025; 74:169-181. [PMID: 39562048 PMCID: PMC11869464 DOI: 10.1136/gutjnl-2024-333364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND There is no clinically relevant serological marker for the early detection of oesophageal adenocarcinoma (EAC) and its precursor lesion, Barrett's oesophagus (BE). OBJECTIVE To develop and test a blood-based assay for EAC and BE. DESIGN Oesophageal MicroRNAs of BaRRett, Adenocarcinoma and Dysplasia (EMERALD) was a large, international, multicentre biomarker cohort study involving 792 patient samples from 4 countries (NCT06381583) to develop and validate a circulating miRNA signature for the early detection of EAC and high-risk BE. Tissue-based miRNA sequencing and microarray datasets (n=134) were used to identify candidate miRNAs of diagnostic potential, followed by validation using 42 pairs of matched cancer and normal tissues. The usefulness of the candidate miRNAs was initially assessed using 108 sera (44 EAC, 34 EAC precursors and 30 non-disease controls). We finally trained a machine learning model (XGBoost+AdaBoost) on RT-qPCR results from circulating miRNAs from a training cohort (n=160) and independently tested it in an external cohort (n=295). RESULTS After a strict process of biomarker discovery and selection, we identified six miRNAs that were overexpressed in all sera of patients compared with non-disease controls from three independent cohorts of different nationalities (miR-106b, miR-146a, miR-15a, miR-18a, miR-21 and miR-93). We established a six-miRNA diagnostic signature using the training cohort (area under the receiver operating characteristic curve (AUROC): 97.6%) and tested it in an independent cohort (AUROC: 91.9%). This assay could also identify patients with BE among patients with gastro-oesophageal reflux disease (AUROC: 94.8%, sensitivity: 92.8%, specificity: 85.1%). CONCLUSION Using a comprehensive approach integrating unbiased genome-wide biomarker discovery and several independent experimental validations, we have developed and validated a novel blood test that might complement screening options for BE/EAC. TRIAL REGISTRATION NUMBER NCT06381583.
Collapse
Affiliation(s)
- Jinsei Miyoshi
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
- Department of Gastroenterology, Kawashima Hospital, Tokushima, Japan
| | - Alessandro Mannucci
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Hospital, Milan, Italy
| | - Marco Scarpa
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padova, Italy
| | - Feng Gao
- Sun Yat-Sen University, The Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Shusuke Toden
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Timothy Whitsett
- Cancer and Cell Biology Division, The Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Landon J Inge
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Ross M Bremner
- Norton Thoracic Institute, St Joseph's Hospital and Medical Center, Phoenix, AZ, USA
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yulan Cheng
- Division of Gastroenterology and Hepatology, Department Of Medicine And Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Teodoro Bottiglieri
- Baylor Scott & White Research Institute, Institute of Metabolic Diseases, Dallas, TX, USA
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Martha J Shrubsole
- Department of Medicine, Division of Epidemiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ali H Zaidi
- Esophageal and Thoracic Research Laboratories, Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China
| | - Helen G Coleman
- Cancer Epidemiology Research Group, Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Lesley A Anderson
- Centre for Health Data Science, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Stephen J Meltzer
- Division of Gastroenterology and Hepatology, Department Of Medicine And Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ajay Goel
- Center for Gastrointestinal Research; Center from Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
7
|
Xie H, Li M, Chen Z, Zheng Y. Association of metformin use with risk and survival outcome of esophageal cancer in patients with diabetes: A systematic review and meta-analysis. PLoS One 2025; 20:e0310687. [PMID: 39774829 PMCID: PMC11706492 DOI: 10.1371/journal.pone.0310687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/04/2024] [Indexed: 01/11/2025] Open
Abstract
The purpose of this study was to conduct a systematic review and meta-analysis to investigate the association between the utilization of metformin and the occurrence and survival rate of esophageal cancer (EC) in individuals with diabetes. METHODS A systematic review and a meta-analysis were performed. Related literature was searched from databases, including PubMed, Embase, Web of Science, and Cochrane Library, covering the period from the inception of these databases until July 2023. RESULTS A total of 16 studies were eligible, including twelve reporting incidences of EC and four reporting OS of EC patients. The combined findings revealed a significant association between the use of metformin and a lower risk of EC (OR, 0.87, P = 0.04). Furthermore, metformin could significantly prolong the OS time (HR, 0.87, P = 0.002). In analyses stratified by treatment modalities, metformin combined with surgery and neoadjuvant chemoradiotherapy presented the strongest protective effect on EC patients with diabetes (HR, 0.38, P = 0.003). CONCLUSION Our meta-analysis indicated that the use of metformin might reduce the EC incidence and improve the OS in EC patients with diabetes.
Collapse
Affiliation(s)
- Hui Xie
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| | - Muhan Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaoqi Chen
- Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yuling Zheng
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
8
|
Gambella A, Fiocca R, Lugaresi M, D'Errico A, Malvi D, Spaggiari P, Tomezzoli A, Albarello L, Ristimäki A, Bottiglieri L, Bonora E, Krishnadath KK, Raulli GD, Rosati R, Romario UF, De Manzoni G, Räsänen J, Mattioli S, Grillo F, Mastracci L. Pre-Surgical Endoscopic Biopsies Are Representative of Esophageal and Esophago-Gastric Junction Adenocarcinoma Histologic Classes and Survival Risk. Cancers (Basel) 2024; 16:4045. [PMID: 39682231 DOI: 10.3390/cancers16234045] [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: 10/21/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Background and Objectives: The Esophageal Adenocarcinoma Study Group Europe (EACSGE) recently proposed a granular histologic classification of esophageal-esophago-gastric junctional adenocarcinomas (EA-EGJAs) based on the study of naïve surgically resected specimens that, when combined with the pTNM stage, is an efficient indicator of prognosis, molecular events, and response to treatment. In this study, we compared histologic classes of endoscopic biopsies taken before surgical resection with those of the surgical specimen, to evaluate the potential of the EACSGE classification at the initial diagnostic workup. Methods: A total of 106 EA-EGJA cases with available endoscopic biopsies and matched surgical resection specimens were retrieved from five Italian institutions. Histologic classification was performed on all specimens to identify well-differentiated glandular adenocarcinoma (WD-GAC), poorly differentiated glandular adenocarcinoma (PD-GAC), mucinous muconodular carcinoma (MMC), infiltrative mucinous carcinoma (IMC), diffuse desmoplastic carcinoma, diffuse anaplastic carcinoma (DAC), and mixed subtypes. Related risk subgroups (low-risk versus high-risk) were also assessed. The correlations of histologic classes and risk subgroups between diagnostic biopsies and surgical resection specimens were explored with Spearman's correlation test. Sensitivity, specificity, accuracy, positive predictive value, negative predictive value, true positives, true negatives, false positives, and false negatives were also calculated. Results: A strong positive correlation between biopsies and surgical specimens occurred for both histologic classes (coefficient: 0.75, p < 0.001) and risk subgroups (coefficient: 0.65, p < 0.001). The highest sensitivities and specificities were observed for MMC, IMC, and DAC (100% and 99% for all), followed by WD-GAC (sensitivity 91%, specificity 79%) and PD-GAC (sensitivity 722%, specificity 86%). The low-risk and high-risk groups presented a sensitivity and specificity of 89% and 76% (low-risk) and 76% and 89% (high-risk). Conclusions: The EACSGE histologic classification of EA-EGJAs and associated prognostic subgroups can be reliably assessed on pre-operative diagnostic biopsies. Further studies on larger and more representative cohorts of EA-EGJAs will allow us to validate our findings and confirm if the EA-EGJA biopsy histomorphology and clinical TNM staging will be as efficient as the surgical specimen histomorphology and pTNM in predicting patient prognoses and tailoring personalized therapeutic approaches.
Collapse
Affiliation(s)
- Alessandro Gambella
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Roberto Fiocca
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Marialuisa Lugaresi
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- Division of Thoracic Surgery, Maria Cecilia Hospital, GVM Care & Research Group, Cotignola, 48022 Ravenna, Italy
| | - Antonietta D'Errico
- Pathology Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Deborah Malvi
- Pathology Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Paola Spaggiari
- Unit of Anatomic Pathology, Humanitas University, 20089 Milan, Italy
| | - Anna Tomezzoli
- Unit of Anatomic Pathology, Azienda Ospedaliera di Verona, 37122 Verona, Italy
| | - Luca Albarello
- Pathology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ari Ristimäki
- Department of Pathology, HUSLAB and HUS Diagnostic Center, University of Helsinki, 00170 Helsinki, Finland
- Helsinki University Hospital, 00170 Helsinki, Finland
| | - Luca Bottiglieri
- Unit of Anatomic Pathology, Istituto Europeo di Oncologia, 20122 Milan, Italy
| | - Elena Bonora
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy
| | - Kausilia K Krishnadath
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Department of Gastroenterology and Hepatology, University Hospital Antwerp, 2650 Antwerp, Belgium
| | | | - Riccardo Rosati
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, Vita Salute San Raffaele University, 20132 Milan, Italy
| | | | - Giovanni De Manzoni
- Department of Surgery, General and Upper G.I. Surgery Division, University of Verona, 37126 Verona, Italy
| | - Jari Räsänen
- Department of General Thoracic and Esophageal Surgery, Helsinki University Hospital, Helsinki University, 00170 Helsinki, Finland
| | - Sandro Mattioli
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- Division of Thoracic Surgery, Maria Cecilia Hospital, GVM Care & Research Group, Cotignola, 48022 Ravenna, Italy
| | - Federica Grillo
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
- IRCCS San Martino Policlinic Hospital of Genoa, 16132 Genoa, Italy
| | - Luca Mastracci
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
- IRCCS San Martino Policlinic Hospital of Genoa, 16132 Genoa, Italy
| |
Collapse
|
9
|
Su SH, Mitani Y, Li T, Sachdeva U, Flashner S, Klein-Szanto A, Dunbar KJ, Abrams J, Nakagawa H, Gabre J. Lactate Suppresses Growth of Esophageal Adenocarcinoma Patient-Derived Organoids through Alterations in Tumor NADH/NAD+ Redox State. Biomolecules 2024; 14:1195. [PMID: 39334961 PMCID: PMC11430592 DOI: 10.3390/biom14091195] [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: 08/26/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Barrett's esophagus (BE) is a common precancerous lesion that can progress to esophageal adenocarcinoma (EAC). There are significant alterations in the esophageal microbiome in the progression from healthy esophagus to BE to EAC, including an increased abundance of a variety of lactate-producing bacteria and an increase of lactate in the tumor microenvironment, as predicted by metabolic modeling. The role of bacterial lactate in EAC is unknown. Here, we utilize patient-derived organoid (PDO) models of EAC and demonstrate that lactate inhibits the growth and proliferation of EAC PDOs through alterations in the tumor NADH/NAD+ redox state. Further RNA sequencing of EAC PDOs identifies ID1 and RSAD2 as potential regulatory molecules crucial in mediating lactate's ability to suppress glycolysis and proliferation. Gene ontology analysis also identifies the activation of inflammatory and immunological pathways in addition to alterations in the metabolic pathways in EAC PDOs exposed to lactate, suggesting a multi-faceted role for lactate in the pathogenesis of EAC.
Collapse
Affiliation(s)
- Steven H. Su
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| | - Yosuke Mitani
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia University Digestive and Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tianxia Li
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia University Digestive and Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Uma Sachdeva
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Samuel Flashner
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia University Digestive and Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andres Klein-Szanto
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Karen J. Dunbar
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia University Digestive and Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Julian Abrams
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia University Digestive and Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hiroshi Nakagawa
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia University Digestive and Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Joel Gabre
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia University Digestive and Liver Diseases Research Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
10
|
Laun S, Pierre F, Kim S, Lunz D, Maddala T, Braun JV, Meltzer SJ, Kann L. Analytical Validation of Esopredict, an Epigenetic Prognostic Assay for Patients with Barrett's Esophagus. Diagnostics (Basel) 2024; 14:2003. [PMID: 39335682 PMCID: PMC11431466 DOI: 10.3390/diagnostics14182003] [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: 07/11/2024] [Revised: 08/27/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
EsopredictTM is a prognostic assay that risk-stratifies Barrett's esophagus patients to predict future progression to high-grade dysplasia (HGD) or esophageal adenocarcinoma (EAC). Established based on foundational studies at Johns Hopkins University, a risk algorithm was developed and clinically validated in two independent studies (n = 320). EsopredictTM is currently offered as a clinical test under the Clinical Laboratory Improvement Amendments (CLIA) guidelines. Here we present the analytical validation by repeated testing of FFPE tissues (n = 26 patients), cell lines, and contrived DNA controls to determine assay performance regarding analytical sensitivity (as defined by the limit of detection (LOD)), analytical specificity (as defined by the limit of blank (LOB)), accuracy as determined from the average positive and negative agreement, repeatability, and reproducibility. The LOD for the assay at 1.5% DNA methylation was significantly higher than the LOB, as determined by an unmethylated DNA control (0% methylated DNA). Inter- and intra-assay average positive agreement (APA) were 88% and 94%, respectively, while average negative agreement (ANA) values were 90% and 94%, respectively. Average inter- and intra-assay precision were <9% and <5% coefficient of variation (CV), respectively. These results confirm that EsopredictTM is a highly reproducible, sensitive, and specific risk categorization assay for the prediction of progression to HGD or EAC within 5 years.
Collapse
Affiliation(s)
| | | | - Suji Kim
- Previse, Halethorpe, MD 21227, USA
| | | | | | | | - Stephen J. Meltzer
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | |
Collapse
|
11
|
Ghosal A, Verma S, Le IT, Lee VT, deGuzman BJ, Aklog L. Analytical Validation of a DNA Methylation Biomarker Test for the Diagnosis of Barrett's Esophagus and Esophageal Adenocarcinoma from Samples Collected Using EsoCheck ®, a Non-Endoscopic Esophageal Cell Collection Device. Diagnostics (Basel) 2024; 14:1784. [PMID: 39202271 PMCID: PMC11354049 DOI: 10.3390/diagnostics14161784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Barrett's esophagus (BE) is a known precursor to esophageal adenocarcinoma (EAC). Guidelines recommend BE screening in populations with multiple risk factors, for which non-endoscopic esophageal cell collection with biomarker testing is considered as an acceptable alternative to esophagogastroduodenoscopy (EGD). The aim of this study was to evaluate analytical performance characteristics of EsoGuard® (EG), a DNA methylation biomarker assay, as a laboratory-developed test (LDT) in esophageal samples collected with the swallowable EsoCheck® (EC) device. EG is a next-generation sequencing (NGS) assay that evaluates methylated vimentin (VIM) and cyclin A1 (CCNA1), clinically validated biomarkers for the detection of BE and EAC. The studies were conducted according to standards of College of American Pathology (CAP), Clinical Laboratory Improvement Amendments (CLIA), and New York (NY) state requirements for the analytical validation of molecular assays. Comparison to Sanger sequencing showed that EG was 100% accurate at all 31 CpG sites evaluated by the assay. The analytical sensitivity, specificity, and accuracy of the assay were 89%, 100%, and 96%, respectively. Intra- and inter-assay precision was 100%. The limit of detection (LOD) was 1 in 400 methylated cells, and the reference range was 84%. In summary, EsoGuard demonstrates high analytical accuracy, repeatability, and reproducibility in samples collected using the EsoCheck device.
Collapse
Affiliation(s)
- Abhisek Ghosal
- Lucid Diagnostics, 360 Madison Ave., Floor 25, New York, NY 10017, USA; (A.G.); (I.T.L.); (V.T.L.); (B.J.d.); (L.A.)
- Lucid Dx Labs, 14 Orchard Road, Lake Forest, CA 92831, USA
| | - Suman Verma
- Lucid Diagnostics, 360 Madison Ave., Floor 25, New York, NY 10017, USA; (A.G.); (I.T.L.); (V.T.L.); (B.J.d.); (L.A.)
- Lucid Dx Labs, 14 Orchard Road, Lake Forest, CA 92831, USA
- PAVmed Inc., 360 Madison Ave., Floor 25, New York, NY 10017, USA
| | - Ivy T. Le
- Lucid Diagnostics, 360 Madison Ave., Floor 25, New York, NY 10017, USA; (A.G.); (I.T.L.); (V.T.L.); (B.J.d.); (L.A.)
- Lucid Dx Labs, 14 Orchard Road, Lake Forest, CA 92831, USA
| | - Victoria T. Lee
- Lucid Diagnostics, 360 Madison Ave., Floor 25, New York, NY 10017, USA; (A.G.); (I.T.L.); (V.T.L.); (B.J.d.); (L.A.)
- PAVmed Inc., 360 Madison Ave., Floor 25, New York, NY 10017, USA
| | - Brian J. deGuzman
- Lucid Diagnostics, 360 Madison Ave., Floor 25, New York, NY 10017, USA; (A.G.); (I.T.L.); (V.T.L.); (B.J.d.); (L.A.)
- Lucid Dx Labs, 14 Orchard Road, Lake Forest, CA 92831, USA
- PAVmed Inc., 360 Madison Ave., Floor 25, New York, NY 10017, USA
| | - Lishan Aklog
- Lucid Diagnostics, 360 Madison Ave., Floor 25, New York, NY 10017, USA; (A.G.); (I.T.L.); (V.T.L.); (B.J.d.); (L.A.)
- Lucid Dx Labs, 14 Orchard Road, Lake Forest, CA 92831, USA
- PAVmed Inc., 360 Madison Ave., Floor 25, New York, NY 10017, USA
| |
Collapse
|
12
|
Laun SE, Kann L, Braun J, Gilbert S, Lunz D, Pierre F, Kalra A, Ma K, Tsai HL, Wang H, Jit S, Cheng Y, Ahmed Y, Wang KK, Leggett CL, Cellini A, Ioffe OB, Zaidi AH, Omstead AN, Jobe B, Korman L, Cornish D, Zellenrath P, Spaander M, Kuipers E, Perpetua L, Greenwald BD, Maddala T, Meltzer SJ. Validation of an Epigenetic Prognostic Assay to Accurately Risk-Stratify Patients with Barrett's Esophagus. Am J Gastroenterol 2024:00000434-990000000-01289. [PMID: 39140473 PMCID: PMC11825890 DOI: 10.14309/ajg.0000000000003030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/20/2024] [Indexed: 08/15/2024]
Abstract
INTRODUCTION:
Esophageal adenocarcinoma (EAC) is the second-most lethal cancer in the United States, with Barrett esophagus (BE) being the strongest risk factor. Assessing the future risk of neoplastic progression in patients with BE is difficult; however, high-grade dysplasia (HGD) and early EAC are treatable by endoscopic eradication therapy (EET), with survival rates of 90%. Thus, it would be beneficial to develop a molecular assay to identify high-risk patients, who merit more frequent endoscopic surveillance or EET, as well as low-risk patients, who can avoid EET and undergo less frequent surveillance.
METHODS:
Deidentified endoscopic biopsies were acquired from 240 patients with BE at 6 centers and confirmed as future progressors or nonprogressors. Tissues were analyzed by a set of methylation-specific biomarker assays. Test performance was assessed in an independent validation set using 4 stratification levels: low risks, low-moderate risks, high-moderate risks, and high risks.
RESULTS:
Relative to patients in the low-risk group, high-risk patients were 15.2 times more likely to progress within 5 years to HGD or EAC. For patients in the high-risk category, the average risk of progressing to HGD or EAC within 5 years was 21.5%, 4-fold the BE population prevalence within 5 years, whereas low-risk patients had a progression risk of only 1.85%.
DISCUSSION:
This clinical assay, Esopredict, stratifies future neoplastic progression risk to identify higher-risk patients with BE who can benefit from EET or more frequent surveillance and lower-risk patients who can benefit from reduced surveillance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrew Kalra
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ke Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Biostatistics, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hao Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Biostatistics, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Simran Jit
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yulan Cheng
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yousra Ahmed
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kenneth K. Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Cadman L. Leggett
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Ashley Cellini
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Olga B. Ioffe
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ali H. Zaidi
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ashten N. Omstead
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Blair Jobe
- Esophageal Institute, Department of Surgery, Allegheny Health Network, Pittsburgh, PA, United States
- Department of Surgery, Drexel University, Philadelphia, PA, United States
| | - Louis Korman
- Capital Digestive Care, Chevy Chase, Maryland, USA
| | - Drew Cornish
- Capital Digestive Care, Chevy Chase, Maryland, USA
| | - Pauline Zellenrath
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Manon Spaander
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Ernst Kuipers
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Lorrie Perpetua
- Research Tissue Biorepository Core Facility, University of Connecticut, CT, USA
| | - Bruce D. Greenwald
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Stephen J. Meltzer
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Malick A, Ferris JS, Hur C, Abrams JA, Soroush A. Racial, Ethnic, and Sex Differences in Incidence-Based Mortality of Aggregate Upper Gastrointestinal Cancers. Clin Transl Gastroenterol 2024; 15:e00745. [PMID: 39007497 PMCID: PMC11346873 DOI: 10.14309/ctg.0000000000000745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
INTRODUCTION Current strategies for upper gastrointestinal (UGI) cancer screening primarily target cancer-specific risk, with the strongest focus on esophageal adenocarcinoma (EAC). However, all UGI cancers are amendable to screening and early detection with an upper endoscopic examination. This study assesses and explores incidence-based mortality (IBM) for cumulative UGI cancers, aiming to identify race-based or sex-based disparities. METHODS We used Surveillance, Epidemiology, and End Results Research data to analyze patients diagnosed with EAC, esophageal squamous cell carcinoma, cardia gastric cancer, noncardia gastric cancer, or colorectal adenocarcinoma from 2000 to 2019. Age-adjusted IBM was calculated as a rate per 100,000 population and stratified by sex and race/ethnicity. We also compared UGI cancer IBM with that of colorectal cancer, a cancer with established population-wide endoscopic screening guidelines. RESULTS Cumulative IBM for UGI cancers was 8.40 (95% confidence interval [CI] 8.34-8.46). The highest cancer-specific IBM rates were for EAC (2.26, 95% CI 2.23-2.29), followed by noncardia gastric cancer (2.07, 95% CI 2.04-2.10), cardia gastric cancer (1.60, 95% CI 1.57-1.62), esophageal squamous cell carcinoma (1.21, 95% CI 1.19-1.23), and miscellaneous UGI cancer (1.27, 95% CI 1.13-1.40). UGI cancer IBM was highest among Black men (16.43, 95% CI 15.97-16.89), American Indian/Alaska Native men (15.23, 95% CI 13.75-16.82), and Hispanic men (13.76, 95% CI 13.42-14.11). These rates are significantly greater than among White men (12.81, 95% CI 12.68-12.95). DISCUSSION UGI cancers impose a significantly higher mortality burden on non-White population subgroups that are not currently targeted by any systematic screening approach.
Collapse
Affiliation(s)
- Alyyah Malick
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Jennifer S. Ferris
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Chin Hur
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Julian A. Abrams
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Ali Soroush
- Division of Data Driven and Digital Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
14
|
Hou T, Yang Z, Zhang Q, Zhang X, Liao X, Lin J. Histology Shift in Esophageal Cancer Between Biopsies and Resections After Neoadjuvant Therapy: A Pilot Study. Int J Surg Pathol 2024; 32:920-925. [PMID: 37899731 DOI: 10.1177/10668969231208029] [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] [Indexed: 10/31/2023]
Abstract
Preoperative neoadjuvant therapy followed by resection is the mainstay treatment for locally advanced esophageal adenocarcinoma. We recently observed the histology shift from predominant esophageal adenocarcinoma in the biopsy to neuroendocrine neoplasm with or without adenocarcinoma in the post-treatment resection. The underlying mechanism of this finding is uncertain, and there is limited information in the literature. A total of 11 patients were identified: 10 patients received presurgical chemoradiation and 1 with chemotherapy. All biopsies were diagnosed with adenocarcinoma. When neuroendocrine immunomarkers were retrospectively performed on 5 biopsies, 2 showed focal positivity, although the classic neuroendocrine morphology was not readily appreciated. All resections contained neuroendocrine neoplasm, including 8 of well-differentiated type and 3 of neuroendocrine carcinomas. Two post-treatment esophagectomies consisted of neuroendocrine neoplasm only without residual adenocarcinoma. Upon follow-up, 8 patients died of the disease (median survival = 26 months), and 3 patients were alive after a median follow-up of 14 months. The overall median survival time was better than the reported esophageal neuroendocrine carcinoma (15 months). The 5-year observed survival rate was 11.3%, which was lower than the Surveillance, Epidemiology, and End Results 5-year survival rate of adenocarcinoma (21.8%). We reported a small series of esophageal adenocarcinoma that showed histology shift between biopsy and esophagectomy after neoadjuvant therapy. Our limited data suggest that prognosis of this group is different than the conventional adenocarcinoma. Awareness of this morphological change reminds pathologists to examine the biopsy specimens thoroughly, because recognition of neuroendocrine neoplasm, especially high-grade neuroendocrine component, might potentially affect pre- and post-surgical regimens.
Collapse
Affiliation(s)
- Tieying Hou
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zhaohai Yang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qingzhao Zhang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaoyan Liao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jingmei Lin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
15
|
Uzun DD, Tryjanowski T, Arians N, Mohr S, Schmitt FCF, Michalski CW, Weigand MA, Debus J, Lang K. Impact of Radiotherapy on Endotracheal Intubation Quality Metrics in Patients with Esophageal Cancer: A Challenge for Advanced Airway Management? Cancers (Basel) 2024; 16:2540. [PMID: 39061180 PMCID: PMC11274552 DOI: 10.3390/cancers16142540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: Currently, no data are available in the literature investigating the influence of radiotherapy (RT) on endotracheal intubation success in patients with esophageal cancer. This study aims to evaluate the impact of RT on endotracheal intubation quality metrics in patients with esophageal cancer. (2) Methods: Patients with esophageal cancer who underwent RT followed by surgery between 2012 and 2023 at the University Hospital Heidelberg, Germany, were retrospectively analyzed. (3) Results: Fifty-five patients, predominantly males 65.5% with a mean age of 64 years, were enrolled. Overall, 81.8% of the patients had an ASA class of III, followed by 27.2% ASA II. The mean prescribed cumulative total dose to the primary tumor and lymph node metastasis was 48.2 Gy with a mean single dose of 1.8 Gy. The mean laryngeal total dose was 40.0 Gy. Direct laryngoscopy was performed in 80.0% of cases, followed by 12.1% videolaryngoscopy, and 7.2% required fiberoptic intubation. Overall, 96.4% of patients were successfully intubated on the first attempt. (4) Conclusions: It has been demonstrated that post-RT effects can increase the risk of airway management difficulties and complications. The results of our study did not indicate any evidence of impaired advanced airway management in patients with esophageal cancer who had undergone RT.
Collapse
Affiliation(s)
- Davut D. Uzun
- Department of Anesthesiology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany; (D.D.U.); (S.M.); (F.C.F.S.); (M.A.W.)
| | - Timo Tryjanowski
- Department of Radiation Oncology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany; (T.T.); (N.A.); (J.D.)
| | - Nathalie Arians
- Department of Radiation Oncology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany; (T.T.); (N.A.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Stefan Mohr
- Department of Anesthesiology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany; (D.D.U.); (S.M.); (F.C.F.S.); (M.A.W.)
| | - Felix C. F. Schmitt
- Department of Anesthesiology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany; (D.D.U.); (S.M.); (F.C.F.S.); (M.A.W.)
| | - Christoph W. Michalski
- Department of General, Visceral and Transplantation Surgery, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany;
| | - Markus A. Weigand
- Department of Anesthesiology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany; (D.D.U.); (S.M.); (F.C.F.S.); (M.A.W.)
| | - Juergen Debus
- Department of Radiation Oncology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany; (T.T.); (N.A.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Heidelberg, 69120 Heidelberg, Germany
| | - Kristin Lang
- Department of Radiation Oncology, Medical Faculty Heidelberg, University Heidelberg, 69120 Heidelberg, Germany; (T.T.); (N.A.); (J.D.)
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| |
Collapse
|
16
|
Yoon JY, Lim F, Shah SC, Rubenstein JH, Abrams JA, Katzka D, Inadomi J, Kim MK, Hur C. Endoscopic Surveillance of Intestinal Metaplasia of the Esophagogastric Junction: A Decision Modeling Analysis. Am J Gastroenterol 2024; 119:1289-1297. [PMID: 38275234 PMCID: PMC11222037 DOI: 10.14309/ajg.0000000000002672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/15/2024] [Indexed: 01/27/2024]
Abstract
INTRODUCTION The incidence of esophagogastric junction adenocarcinoma (EGJAC) has been rising. Intestinal metaplasia of the esophagogastric junction (EGJIM) is a common finding in gastroesophageal reflux (irregular Z-line) and may represent an early step in the development of EGJAC in the West. Worldwide, EGJIM may represent progression along the Correa cascade triggered by Helicobacter pylori . We sought to evaluate the cost-effectiveness of endoscopic surveillance of EGJIM. METHODS We developed a decision analytic model to compare endoscopic surveillance strategies for 50-year-old patients after diagnosis of non-dysplastic EGJIM: (i) no surveillance (standard of care), (ii) endoscopy every 3 years, (iii) endoscopy every 5 years, or (iv) 1-time endoscopy at 3 years. We modeled 4 progression scenarios to reflect uncertainty: A (0.01% annual cancer incidence), B (0.05%), C (0.12%), and D (0.22%). RESULTS Cost-effectiveness of endoscopic surveillance depended on the progression rate of EGJIM to cancer. At the lowest progression rate (scenario A, 0.01%), no surveillance strategies were cost-effective. In moderate progression scenarios, 1-time surveillance at 3 years was cost-effective, at $30,989 and $16,526 per quality-adjusted life year for scenarios B (0.05%) and C (0.12%), respectively. For scenario D (0.22%), surveillance every 5 years was cost-effective at $77,695 per quality-adjusted life year. DISCUSSION Endoscopic surveillance is costly and can cause harm; however, low-intensity longitudinal surveillance (every 5 years) is cost-effective in populations with higher EGJAC incidence. No surveillance or 1-time endoscopic surveillance of patients with EGJIM was cost-effective in low-incidence populations. Future studies to better understand the natural history of EGJIM, identify risk factors of progression, and inform appropriate surveillance strategies are required.
Collapse
Affiliation(s)
- Ji Yoon Yoon
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Francesca Lim
- Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Shailja C. Shah
- Division of Gastroenterology, University of California San Diego, La Jolla, CA
- Gastroenterology Section, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Joel H. Rubenstein
- Center for Clinical Management Research, LTC Charles S Kettles Veterans Affairs Medical Center, Ann Arbor, MI
- Barrett’s Esophagus Program, Division of Gastroenterolgy, University of Michigan Medical School, Ann Arbor, MI
| | - Julian A. Abrams
- Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY
| | - David Katzka
- Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY
| | - John Inadomi
- Department of Internal Medicine, The University of Utah School of Medicine, Salt Lake City, UT
| | - Michelle K. Kim
- Department of Gastroenterology, Hepatology and Nutrition, Cleveland Clinic, Cleveland, Ohio
| | - Chin Hur
- Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY
- Herbert Irving Comprehensive Cancer center, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
17
|
Hicheri C, Azimuddin AM, Kortum A, Bailey J, Tang Y, Schwarz RA, Rosen D, Jain S, Mansour NM, Groth S, Vasavada S, Rao A, Maliga A, Gallego L, Carns J, Anandasabapathy S, Richards-Kortum R. Design and Evaluation of ScanCap: A Low-Cost, Reusable Tethered Capsule Endoscope with Blue-Green Illumination Imaging for Unsedated Screening and Early Detection of Barrett's Esophagus. Bioengineering (Basel) 2024; 11:557. [PMID: 38927792 PMCID: PMC11200367 DOI: 10.3390/bioengineering11060557] [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: 04/25/2024] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 06/28/2024] Open
Abstract
Esophageal carcinoma is the sixth-leading cause of cancer death worldwide. A precursor to esophageal adenocarcinoma (EAC) is Barrett's Esophagus (BE). Early-stage diagnosis and treatment of esophageal neoplasia (Barrett's with high-grade dysplasia/intramucosal cancer) increase the five-year survival rate from 10% to 98%. BE is a global challenge; however, current endoscopes for early BE detection are costly and require extensive infrastructure for patient examination and sedation. We describe the design and evaluation of the first prototype of ScanCap, a high-resolution optical endoscopy system with a reusable, low-cost tethered capsule, designed to provide high-definition, blue-green illumination imaging for the early detection of BE in unsedated patients. The tethered capsule (12.8 mm diameter, 35.5 mm length) contains a color camera and rotating mirror and is designed to be swallowed; images are collected as the capsule is retracted manually via the tether. The tether provides electrical power and illumination at wavelengths of 415 nm and 565 nm and transmits data from the camera to a tablet. The ScanCap prototype capsule was used to image the oral mucosa in normal volunteers and ex vivo esophageal resections; images were compared to those obtained using an Olympus CV-180 endoscope. Images of superficial capillaries in intact oral mucosa were clearly visible in ScanCap images. Diagnostically relevant features of BE, including irregular Z-lines, distorted mucosa, and dilated vasculature, were clearly visible in ScanCap images of ex vivo esophageal specimens.
Collapse
Affiliation(s)
- Cheima Hicheri
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | - Ahad M. Azimuddin
- Houston Methodist Hospital, Houston, TX 77030, USA;
- Texas A&M School of Medicine, Houston, TX 77030, USA
| | - Alex Kortum
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | - Joseph Bailey
- Rice360 Institute for Global Health Technologies, Rice University, Houston, TX 77030, USA
| | - Yubo Tang
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | - Richard A. Schwarz
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | - Daniel Rosen
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | - Shilpa Jain
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | | | - Shawn Groth
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | | | - Ashwin Rao
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | | | - Leslie Gallego
- Baylor College of Medicine, Houston, TX 77030, USA (L.G.)
| | - Jennifer Carns
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; (C.H.); (R.A.S.)
| | | | | |
Collapse
|
18
|
Dhaliwal L, Kamboj AK, Williams JL, Chandar AK, Sachdeva K, Gibbons E, Lansing R, Passe M, Perez JA, Avenir KLR, Martin SA, Leggett CL, Chak A, Falk GW, Wani S, Shaheen NJ, Kisiel JB, Iyer PG. Prevalence and Predictors of Barrett's Esophagus After Negative Initial Endoscopy: Analysis From Two National Databases. Clin Gastroenterol Hepatol 2024; 22:523-531.e3. [PMID: 37716614 PMCID: PMC10922211 DOI: 10.1016/j.cgh.2023.08.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/17/2023] [Accepted: 08/22/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND & AIMS Guidelines suggest a single screening esophagogastroduodenoscopy (EGD) in patients with multiple risk factors for Barrett's esophagus (BE). We aimed to determine BE prevalence and predictors on repeat EGD after a negative initial EGD, using 2 large national databases (GI Quality Improvement Consortium [GIQuIC] and TriNetX). METHODS Patients who underwent at least 2 EGDs were included and those with BE or esophageal adenocarcinoma detected at initial EGD were excluded. Patient demographics and prevalence of BE on repeat EGD were collected. Multivariate logistic regression was performed to assess for independent risk factors for BE detected on the repeat EGD. RESULTS In 214,318 and 153,445 patients undergoing at least 2 EGDs over a median follow-up of 28-35 months, the prevalence of BE on repeat EGD was 1.7% in GIQuIC and 3.4% in TriNetX, respectively (26%-45% of baseline BE prevalence). Most (89%) patients had nondysplastic BE. The prevalence of BE remained stable over time (from 1 to >5 years from negative initial EGD) but increased with increasing number of risk factors. BE prevalence in a high-risk population (gastroesophageal reflux disease plus ≥1 risk factor for BE) was 3%-4%. CONCLUSIONS In this study of >350,000 patients, rates of BE on repeat EGD ranged from 1.7%-3.4%, and were higher in those with multiple risk factors. Most were likely missed at initial evaluation, underscoring the importance of a high-quality initial endoscopic examination. Although routine repeat endoscopic BE screening after a negative initial examination is not recommended, repeat screening may be considered in carefully selected patients with gastroesophageal reflux disease and ≥2 risk factors for BE, potentially using nonendoscopic tools.
Collapse
Affiliation(s)
- Lovekirat Dhaliwal
- Department of Internal Medicine, Louisiana State University Health, Shreveport, Louisiana
| | - Amrit K Kamboj
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Apoorva K Chandar
- Department of Internal Medicine, Case Western Reserve University Hospitals, Cleveland, Ohio
| | - Karan Sachdeva
- Department of Internal Medicine, Louisiana State University Health, Shreveport, Louisiana
| | - Erin Gibbons
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Ramona Lansing
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Melissa Passe
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jaime A Perez
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Katelin L R Avenir
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Scott A Martin
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Cadman L Leggett
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Amitabh Chak
- Division of Gastroenterology, Case Western Reserve University Hospitals, Cleveland, Ohio
| | - Gary W Falk
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sachin Wani
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicholas J Shaheen
- Division of Gastroenterology and Hepatology, University of North Carolina, Chapel Hill, North Carolina
| | - John B Kisiel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Prasad G Iyer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
19
|
Mithi MT, Sharma M, Puj K, Hazarika P, Pandya SJ, Gandhi J, Parikh A, Shukla S. Neoadjuvant Chemoradiation Followed by Surgery for Locally Advanced Squamous Cell Carcinoma Esophagus: Demographics and Evaluation of Prognostic Factors at a Tertiary Care Center in India. Indian J Surg Oncol 2024; 15:129-135. [PMID: 38511020 PMCID: PMC10948642 DOI: 10.1007/s13193-023-01828-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/28/2023] [Indexed: 03/22/2024] Open
Abstract
Neoadjuvant chemoradiation followed by surgery has been the standard of care for locally advanced carcinoma esophagus. We present our experience and inference of various factors associated with the same treatment and the prognostic influence of the same. A retrospective analysis of a cohort of 132 squamous cell carcinoma esophagus patients post neoadjuvant chemoradiation operated with curative intent was carried out. The 2-year overall survival rate was 64.5%. A pathological complete response was achieved in 32.5% of patients and was the only factor that significantly determined overall survival (p = 0.048). Neoadjuvant chemoradiation before surgery for locally advanced squamous cell cancer of the esophagus remains the standard of care with a pathological complete response being a significant factor in predicting overall survival. More prospective randomized studies are necessary to analyze factors affecting and predicting a pathological complete response which would help organ preservation in patients with a complete response.
Collapse
Affiliation(s)
- Mohamed Taher Mithi
- Department of Surgical Oncology, Gujarat Cancer Research Institute, Ahmedabad, Gujarat 380016 India
| | - Mohit Sharma
- Department of Surgical Oncology, Gujarat Cancer Research Institute, Ahmedabad, Gujarat 380016 India
| | - Ketul Puj
- Department of Surgical Oncology, Gujarat Cancer Research Institute, Ahmedabad, Gujarat 380016 India
| | - Prandweep Hazarika
- Department of Surgical Oncology, Gujarat Cancer Research Institute, Ahmedabad, Gujarat 380016 India
| | - Shashank J. Pandya
- Department of Surgical Oncology, Gujarat Cancer Research Institute, Ahmedabad, Gujarat 380016 India
| | - Jahnavi Gandhi
- Department of Pathology, Gujarat Cancer Research Institute, Ahmedabad, Gujarat 380016 India
| | - Ankita Parikh
- Department of Radiotherapy, Gujarat Cancer Research Institute, Ahmedabad, Gujarat 380016 India
| | - Shivang Shukla
- Department of Surgical Oncology, Gujarat Cancer Research Institute, Ahmedabad, Gujarat 380016 India
| |
Collapse
|
20
|
Solfisburg QS, Baldini F, Baldwin-Hunter B, Austin GI, Lee HH, Park H, Freedberg DE, Lightdale CJ, Korem T, Abrams JA. The Salivary Microbiome and Predicted Metabolite Production Are Associated with Barrett's Esophagus and High-Grade Dysplasia or Adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2024; 33:371-380. [PMID: 38117184 PMCID: PMC10955687 DOI: 10.1158/1055-9965.epi-23-0652] [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: 06/05/2023] [Revised: 09/05/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) is rising in incidence, and established risk factors do not explain this trend. Esophageal microbiome alterations have been associated with Barrett's esophagus (BE) and dysplasia and EAC. The oral microbiome is tightly linked to the esophageal microbiome; this study aimed to identify salivary microbiome-related factors associated with BE, dysplasia, and EAC. METHODS Clinical data and oral health history were collected from patients with and without BE. The salivary microbiome was characterized, assessing differential relative abundance of taxa by 16S rRNA gene sequencing and associations between microbiome composition and clinical features. Microbiome metabolic modeling was used to predict metabolite production. RESULTS A total of 244 patients (125 non-BE and 119 BE) were analyzed. Patients with high-grade dysplasia (HGD)/EAC had a significantly higher prevalence of tooth loss (P = 0.001). There were significant shifts with increased dysbiosis associated with HGD/EAC, independent of tooth loss, with the largest shifts within the genus Streptococcus. Modeling predicted significant shifts in the microbiome metabolic capacities, including increases in L-lactic acid and decreases in butyric acid and L-tryptophan production in HGD/EAC. CONCLUSIONS Marked dysbiosis in the salivary microbiome is associated with HGD and EAC, with notable increases within the genus Streptococcus and accompanying changes in predicted metabolite production. Further work is warranted to identify the biological significance of these alterations and to validate metabolic shifts. IMPACT There is an association between oral dysbiosis and HGD/EAC. Further work is needed to establish the diagnostic, predictive, and causal potential of this relationship.
Collapse
Affiliation(s)
- Quinn S Solfisburg
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Federico Baldini
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - George I Austin
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Harry H Lee
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Microbiome and Pathogen Genomics Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Daniel E Freedberg
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles J Lightdale
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
- CIFAR Azrieli Global Scholars Program, CIFAR, Toronto, Canada
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY USA
| |
Collapse
|
21
|
Saito M, Koike T, Ohara Y, Ogata Y, Kanno T, Jin X, Hatta W, Uno K, Asano N, Imatani A, Masamune A. Linked Color Imaging of Barrett’s Esophageal Adenocarcinoma: Effects on Visibility. GASTROENTEROLOGY INSIGHTS 2024; 15:145-155. [DOI: 10.3390/gastroent15010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Since linked color imaging (LCI) has been reported to increase the color differences in Barrett’s esophageal adenocarcinoma (BA) compared to white light imaging (WLI), a comparison of the visibility scores of various imaging techniques for BA is warranted to determine best practice standards. This study is to clarify the role of LCI, blue light imaging (BLI), and WLI in the evaluation of BA. A group of 19 endoscopists, comprised of 6 experts and 13 trainees, evaluated the visibility of WLI, BLI, and LCI images in 21 superficial BA cases. Visibility scores were compared between WLI, BLI, and LCI. Visibility scores were also evaluated for lesion morphology, background Barrett’s mucosa, and circumferential location. The visibility scores of experts and trainees were analyzed for comparison. The visibility scores of LCI and BLI were 3.83 and 3.31, respectively, compared to three points for WLI. The visibility of LCI was better than that of WLI regardless of lesion morphology, color, background Barrett’s mucosa, and circumferential location. The LCI improved visibility in BA more than the WLI for both experts and trainees. LCI improved the visibility of BA independent of lesion morphology, color, background Barrett’s mucosa, circumferential location, and the endoscopist’s experience.
Collapse
Affiliation(s)
- Masahiro Saito
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Department of Community Medical Supports, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yuki Ohara
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yohei Ogata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Takeshi Kanno
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Xiaoyi Jin
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Kaname Uno
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Naoki Asano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Akira Imatani
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| |
Collapse
|
22
|
Leclercq P, Bisschops R, Bergman JJGHM, Pouw RE. Management of high risk T1 esophageal adenocarcinoma following endoscopic resection. Best Pract Res Clin Gastroenterol 2024; 68:101882. [PMID: 38522880 DOI: 10.1016/j.bpg.2024.101882] [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: 11/04/2023] [Accepted: 01/17/2024] [Indexed: 03/26/2024]
Abstract
High-risk T1 esophageal adenocarcinoma (HR-T1 EAC) is defined as T1 cancer, with one or more of the following histological criteria: submucosal invasion, poorly or undifferentiated cancer, and/or presence of lympho-vascular invasion. Esophagectomy has long been the only available treatment for these HR-T1 EACs and was considered necessary because of a presumed high risk of lymph node metastases up to 46%. However, endoscopic submucosal disscection have made it possible to radically remove HR-T1 EAC, irrespective of size, while leaving the esophageal anatomy intact. Parallel to this development, new publications demonstrated that the risk of lymph node metastases for HR-T1 EAC may be even <24%. Therefore, indications for endoscopic treatment of HR-T1 EAC are being reconsidered and current research aims at finding the optimal management strategy for this indication, where watchful waiting may proof to be an acceptable strategy in selected patients. In this review, we will discuss the latest developments in this field.
Collapse
Affiliation(s)
- Philippe Leclercq
- Departement of Gastroenterology, Universitair Ziekenhuis Leuven, 49 Herestraat, 3000, LEUVEN, Belgium.
| | - Raf Bisschops
- Departement of Gastroenterology, Universitair Ziekenhuis Leuven, 49 Herestraat, 3000, LEUVEN, Belgium.
| | - Jacques J G H M Bergman
- Dept. of Gastroenterology and Hepatology, Amsterdam University Medical Centers, De Boelelaan 1117, Amsterdam, 1081, HV, Netherlands.
| | - Roos E Pouw
- Dept. of Gastroenterology and Hepatology, Amsterdam University Medical Centers, De Boelelaan 1117, Amsterdam, 1081, HV, Netherlands.
| |
Collapse
|
23
|
Uzun DD, Salatzki J, Xynogalos P, Frey N, Debus J, Lang K. Effects of Ionizing Radiation on Cardiac Implantable Electronic Devices (CIEDs) in Patients with Esophageal Cancer Undergoing Radiotherapy: A Pilot Study. Cancers (Basel) 2024; 16:555. [PMID: 38339306 PMCID: PMC10854512 DOI: 10.3390/cancers16030555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
(1) Background: The prevalence of cancer patients relying on cardiac implantable electronic device (CIED) is steadily rising. The aim of this study was to evaluate RT-related malfunctions of CIEDs. (2) Methods: We retrospectively analyze sixteen patients with esophageal cancer who were treated with radiotherapy between 2012 and 2022 at the University Hospital Heidelberg. All patients underwent systemic evaluation including pre-therapeutic cardiological examinations of the CIED functionality and after every single irradiation. (3) Results: Sixteen patients, predominantly male (14) with a mean age of 77 (range: 56-85) years were enrolled. All patients received 28 fractions of radiotherapy with a cumulative total dose 58.8 Gy. The mean maximum dose at the CIEDs was 1.8 Gy. Following radiotherapy and during the one-year post-radiation follow-up period, there were no registered events associated with the treatment in this evaluation. (4) Conclusion: The study did not observe any severe CIED malfunctions following each radiation fraction or after completion of RT. Strict selection of photon energy and alignment with manufacturer-recommended dose limits appear to be important. Our study showed no major differences in the measured values of the pacing threshold, sensing threshold and lead impedance after RT.
Collapse
Affiliation(s)
- Davut D. Uzun
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
- Heidelberg Center for Heart Rhythm Disorders (HCR), 69120 Heidelberg, Germany; (J.S.); (P.X.); (N.F.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Janek Salatzki
- Heidelberg Center for Heart Rhythm Disorders (HCR), 69120 Heidelberg, Germany; (J.S.); (P.X.); (N.F.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Panagiotis Xynogalos
- Heidelberg Center for Heart Rhythm Disorders (HCR), 69120 Heidelberg, Germany; (J.S.); (P.X.); (N.F.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Norbert Frey
- Heidelberg Center for Heart Rhythm Disorders (HCR), 69120 Heidelberg, Germany; (J.S.); (P.X.); (N.F.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Juergen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg University Hospital, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Heidelberg, 69120 Heidelberg, Germany
| | - Kristin Lang
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Li H, Yang X, Zhang A, Liang G, Sun Y, Zhang J. Age-period-cohort analysis of incidence, mortality and disability-adjusted life years of esophageal cancer in global, regional and national regions from 1990 to 2019. BMC Public Health 2024; 24:212. [PMID: 38233775 PMCID: PMC10795420 DOI: 10.1186/s12889-024-17706-8] [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: 07/13/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024] Open
Abstract
OBJECTIVE In view of the high incidence and mortality of esophageal cancer, the latest statistical data on the disease burden of esophageal cancer can provide strategies for cancer screening, early detection and treatment, and help to rationally allocate health resources. This study provides an analysis of the global disease burden and risk factors of esophageal cancer from 1990 to 2019. METHODS Using the 2019 Global Burden of Disease, Injury and Risk Factor (GBD) data, we present the incidence, mortality and disability-adjusted life years (DALY) of esophageal cancer in 21 regions and 204 countries and different sociodemographic index (SDI) regions from 1990 to 2019. The age-period-cohort model was used to estimate the age, period, and cohort trend of esophageal cancer in different SDI regions. The estimated proportion of DALY attributable to each risk factor from 1990 to 2019. RESULTS From 1990 to 2019, the number of new cases of esophageal cancer, the number of deaths and DALY increased by 67.07%, 55.97% and 42.13%, respectively, but age standardized incidence rate (ASIR), age standardized mortality rate (ASMR) and age standardized DALY rate (ASDR) decreased by 19.28%, 25.32% and 88.22%, respectively. Overall, the results of the age-period-cohort model showed that the incidence, mortality, and DALY rates in countries and regions with higher SDI levels showed a downward trend over time and with the passage of time. Conversely, there were no significant changes in incidence and mortality in countries and regions with low SDI levels. In the past 30 years, the incidence and death of esophageal cancer in the world has gradually changed to people over 80 years old, but the population aged 60-79 still accounts for the largest proportion. The global DALY in esophageal cancer is mainly attributable to smoking, followed by alcohol consumption and occupational exposure. CONCLUSIONS Although ASIR, ASMR and ASDR have decreased significantly, esophageal cancer is still the main factor causing the disease burden worldwide. Public health administrators in low SDI and low-middle SDI countries are high-risk areas for esophageal cancer, and preventive control measures should be implemented to raise awareness, screening, and treatment of esophageal cancer in these areas. Tobacco and alcohol control and reduction of occupational hazards are key steps in reducing the burden of esophageal cancer.
Collapse
Affiliation(s)
- Huiying Li
- Department of Pathology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin City, Heilongjiang Province, People's Republic of China
| | - Xianzhi Yang
- Emergency Internal Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, People's Republic of China
| | - Aiqi Zhang
- Emergency Internal Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, People's Republic of China
| | - Guanying Liang
- Department of Pathology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin City, Heilongjiang Province, People's Republic of China
| | - Yue Sun
- Academic Department of Science and Technology, Harbin Medical University Cancer Hospital, Harbin City, Heilongjiang Province, People's Republic of China.
| | - Jian Zhang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin City, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
25
|
Agarwal S, Bell MG, Dhaliwal L, Codipilly DC, Dierkhising RA, Lansing R, Gibbons EE, Leggett CL, Kisiel JB, Iyer PG. Population Based Time Trends in the Epidemiology and Mortality of Gastroesophageal Junction and Esophageal Adenocarcinoma. Dig Dis Sci 2024; 69:246-253. [PMID: 37914889 PMCID: PMC10926253 DOI: 10.1007/s10620-023-08126-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/02/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Limited data are available on the epidemiology of gastroesophageal junction adenocarcinoma (GEJAC), particularly in comparison to esophageal adenocarcinoma (EAC). With the advent of molecular non-endoscopic Barrett's esophagus (BE) detection tests which sample the esophagus and gastroesophageal junction, early detection of EAC and GEJAC has become a possibility and their epidemiology has gained importance. AIMS We sought to evaluate time trends in the epidemiology and survival of patients with EAC and GEJAC in a population-based cohort. METHODS EAC and GEJAC patients from 1976 to 2019 were identified using ICD 9 and 10 diagnostic codes from the Rochester Epidemiology Project (REP). Clinical data and survival status were abstracted. Poisson regression was used to calculate incidence rate ratios (IRR). Survival analysis and Cox proportional models were used to assess predictors of survival. RESULTS We included 443 patients (287 EAC,156 GEJAC). The incidence of EAC and GEJAC during 1976-2019 was 1.40 (CI 1.1-1.74) and 0.83 (CI 0.61-1.11) per 100,000 people, respectively. There was an increase in the incidence of EAC (IRR = 2.45, p = 0.011) and GEJAC (IRR = 3.17, p = 0.08) from 2000 to 2004 compared to 1995-1999, plateauing in later time periods. Most patients had associated BE and presented at advanced stages, leading to high 5-year mortality rates (66% in EAC and 59% in GEJAC). Age and stage at diagnosis were predictors of mortality. CONCLUSION The rising incidence of EAC/GEJAC appears to have plateaued somewhat in the last decade. However, both cancers present at advanced stages with persistently poor survival, underscoring the need for early detection.
Collapse
Affiliation(s)
- Siddharth Agarwal
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Matthew G Bell
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lovekirat Dhaliwal
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - D Chamil Codipilly
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Ross A Dierkhising
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Ramona Lansing
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Erin E Gibbons
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Cadman L Leggett
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - John B Kisiel
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Prasad G Iyer
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
26
|
Rubenstein JH, Fontaine S, MacDonald PW, Burns JA, Evans RR, Arasim ME, Chang JW, Firsht EM, Hawley ST, Saini SD, Wallner LP, Zhu J, Waljee AK. Predicting Incident Adenocarcinoma of the Esophagus or Gastric Cardia Using Machine Learning of Electronic Health Records. Gastroenterology 2023; 165:1420-1429.e10. [PMID: 37597631 PMCID: PMC11013733 DOI: 10.1053/j.gastro.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/11/2023] [Accepted: 08/09/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND & AIMS Tools that can automatically predict incident esophageal adenocarcinoma (EAC) and gastric cardia adenocarcinoma (GCA) using electronic health records to guide screening decisions are needed. METHODS The Veterans Health Administration (VHA) Corporate Data Warehouse was accessed to identify Veterans with 1 or more encounters between 2005 and 2018. Patients diagnosed with EAC (n = 8430) or GCA (n = 2965) were identified in the VHA Central Cancer Registry and compared with 10,256,887 controls. Predictors included demographic characteristics, prescriptions, laboratory results, and diagnoses between 1 and 5 years before the index date. The Kettles Esophageal and Cardia Adenocarcinoma predictioN (K-ECAN) tool was developed and internally validated using simple random sampling imputation and extreme gradient boosting, a machine learning method. Training was performed in 50% of the data, preliminary validation in 25% of the data, and final testing in 25% of the data. RESULTS K-ECAN was well-calibrated and had better discrimination (area under the receiver operating characteristic curve [AuROC], 0.77) than previously validated models, such as the Nord-Trøndelag Health Study (AuROC, 0.68) and Kunzmann model (AuROC, 0.64), or published guidelines. Using only data from between 3 and 5 years before index diminished its accuracy slightly (AuROC, 0.75). Undersampling men to simulate a non-VHA population, AUCs of the Nord-Trøndelag Health Study and Kunzmann model improved, but K-ECAN was still the most accurate (AuROC, 0.85). Although gastroesophageal reflux disease was strongly associated with EAC, it contributed only a small proportion of gain in information for prediction. CONCLUSIONS K-ECAN is a novel, internally validated tool predicting incident EAC and GCA using electronic health records data. Further work is needed to validate K-ECAN outside VHA and to assess how best to implement it within electronic health records.
Collapse
Affiliation(s)
- Joel H Rubenstein
- Veterans Affairs Center for Clinical Management Research, Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, Michigan; Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan.
| | - Simon Fontaine
- Department of Statistics, University of Michigan College of Literature, Science, and Arts, Ann Arbor, Michigan
| | - Peter W MacDonald
- Department of Statistics, University of Michigan College of Literature, Science, and Arts, Ann Arbor, Michigan
| | - Jennifer A Burns
- Veterans Affairs Center for Clinical Management Research, Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, Michigan
| | - Richard R Evans
- Veterans Affairs Center for Clinical Management Research, Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, Michigan
| | - Maria E Arasim
- Veterans Affairs Center for Clinical Management Research, Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, Michigan
| | - Joy W Chang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Elizabeth M Firsht
- Veterans Affairs Center for Clinical Management Research, Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, Michigan
| | - Sarah T Hawley
- Veterans Affairs Center for Clinical Management Research, Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan; Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Sameer D Saini
- Veterans Affairs Center for Clinical Management Research, Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, Michigan; Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan
| | - Lauren P Wallner
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan; Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Ji Zhu
- Department of Statistics, University of Michigan College of Literature, Science, and Arts, Ann Arbor, Michigan
| | - Akbar K Waljee
- Veterans Affairs Center for Clinical Management Research, Lieutenant Colonel Charles S. Kettles Veterans Affairs Medical Center, Ann Arbor, Michigan; Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
27
|
Duits LC, Khoshiwal AM, Frei NF, Pouw RE, Smolko C, Arora M, Siegel JJ, Critchley-Thorne RJ, Bergman JJ. An Automated Tissue Systems Pathology Test Can Standardize the Management and Improve Health Outcomes for Patients With Barrett's Esophagus. Am J Gastroenterol 2023; 118:2025-2032. [PMID: 37307529 PMCID: PMC10617665 DOI: 10.14309/ajg.0000000000002363] [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: 01/05/2023] [Accepted: 04/27/2023] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Low-grade dysplasia (LGD) in Barrett's esophagus (BE) is associated with an increased risk of progression to high-grade dysplasia or esophageal adenocarcinoma. However, because of substantial interobserver variability in the diagnosis of LGD, a patient's management plan and health outcome depend largely on which pathologist reviews their case. This study evaluated the ability of a tissue systems pathology test that objectively risk stratifies patients with BE (TissueCypher, TSP-9) to standardize management in a manner consistent with improved health outcomes for patients with BE. METHODS A total of 154 patients with BE with community-based LGD from the prospectively followed screening cohort of the SURF trial were studied. Management decisions were simulated 500 times with varying generalist (n = 16) and expert (n = 14) pathology reviewers to determine the most likely care plan with or without use of the TSP-9 test for guidance. The percentage of patients receiving appropriate management based on the known progression/nonprogression outcomes was calculated. RESULTS The percentage of patients with 100% of simulations resulting in appropriate management significantly increased from 9.1% for pathology alone, to 58.4% when TSP-9 results were used with pathology, and further increased to 77.3% of patients receiving appropriate management when only TSP-9 results were used. Use of the test results also significantly increased the consistency of management decisions for patients when their slides were reviewed by different pathologists ( P < 0.0001). DISCUSSION Management guided by the TSP-9 test can standardize care plans by increasing the early detection of progressors who can receive therapeutic interventions, while also increasing the percentage of nonprogressors who can avoid unnecessary therapy and be managed by surveillance alone.
Collapse
Affiliation(s)
- Lucas C. Duits
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Amir M. Khoshiwal
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Nicola F. Frei
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| | - Roos E. Pouw
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, VUmc, Amsterdam, the Netherlands
| | | | | | | | | | - Jacques J.G.H.M. Bergman
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
28
|
Fernandes A, Li C, French D, Ellsmere J. Ten-year follow-up of endoscopic mucosal resection versus esophagectomy for esophageal intramucosal adenocarcinoma in the setting of Barrett's esophagus: a Canadian experience. Surg Endosc 2023; 37:8735-8741. [PMID: 37563345 DOI: 10.1007/s00464-023-10318-0] [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: 04/18/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Endoscopic mucosal resection (EMR) is an effective treatment for esophageal intramucosal adenocarcinoma (IMC), with similar recurrence and mortality rates versus esophagectomy in up to 5 years of follow-up. Long-term outcomes to 10 years have not been studied. This retrospective study investigates IMC eradication, recurrence, morbidity and mortality at 10 years following EMR versus esophagectomy in a single Canadian institution. METHODS Patients with IMC treated via esophagectomy or EMR from 2006 to 2015 were included. Post-EMR endoscopic follow-up occurred every 3 months for 1 year, every 6 months for 2 years and every 12 months thereafter. Categorical variables were expressed as percentages and continuous variables as mean with standard deviation or median and interquartile range. The student's t-test and Fischer's exact test were used for comparisons. Survival analysis utilized the Kaplan-Meier estimator and log-rank test. RESULTS Twenty-four patients were included. Patient and tumor characteristics were similar between groups. Median follow-up for EMR and esophagectomy were 85.2 months [IQR 64.8] and 126 months [IQR 54] respectively. A mean of 1.3 EMR (SD 1.1) were required for eradication, which was seen in 12 patients (12/14, 86%). No EMR-related complications occurred. Disease progression was seen in two patients (2/14, 14%); local recurrence was seen in 1 patient (1/14, 7%). Esophagectomy eradicated IMC in 10 patients (10/10, 100%); recurrence was seen in 2 (2/10, 20%, metastatic). Major, early esophagectomy-related morbidity affected 3 patients (3/10, 30%), and late morbidity was documented for 9 (9/10, 90%). Esophagectomy and EMR had similar recurrence rates (p = 0.554). Esophagectomy was associated with significantly more procedure-related morbidity (p < 0.001). There was no difference in mortality (p = 0.442) or disease-free survival (p = 0.512) between treatment groups. CONCLUSION EMR and esophagectomy for the treatment of IMC are associated with comparable recurrence rates and disease-free survival in 10-year follow-up. EMR is associated with significantly lower procedure-associated morbidity. EMR can be used to treat T1a distal esophageal adenocarcinoma with minimal procedure-related morbidity, and acceptable oncologic outcomes in long-term follow-up.
Collapse
Affiliation(s)
- Alisha Fernandes
- Division of General and Gastrointestinal Surgery, Department of Surgery, Dalhousie University, Room 8-23/4 Victoria Building, QEII-HSC, VG Site, 1276 South Park St, Halifax, NS, B3H 2Y9, Canada
| | - Chao Li
- Division of General Surgery, Department of Surgery, Centre Intégré Universitaire de Santé et de Services Sociaux de l'Est-de-l'Île-de-Montréal, Montreal, Canada
| | - Daniel French
- Division of Thoracic Surgery, Department of Surgery, Dalhousie University, Halifax, Canada
| | - James Ellsmere
- Division of General and Gastrointestinal Surgery, Department of Surgery, Dalhousie University, Room 8-23/4 Victoria Building, QEII-HSC, VG Site, 1276 South Park St, Halifax, NS, B3H 2Y9, Canada.
| |
Collapse
|
29
|
Valdovinos-Andraca F, Bartnicki-Navarrete I, Bernal-Mendez AR, Rafael Barreto-Zuñiga R, Romano-Munive AF, Gamboa-Domínguez A, Elizondo-Rivera J, Briseño-García D, Tellez-Ávila FI. Clinical and Endoscopic Differences Between Patients With Barrett's Esophagus With and Without Dysplasia/Adenocarcinoma. Cureus 2023; 15:e46323. [PMID: 37916254 PMCID: PMC10617646 DOI: 10.7759/cureus.46323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2023] [Indexed: 11/03/2023] Open
Abstract
INTRODUCTION Barrett's esophagus (BE) is the main precursor of esophageal adenocarcinoma (EAC). This study aimed to identify the risk factors associated with BE progression to dysplasia or EAC in a Latin population. METHODS The study is a retrospective analysis of a single-center cohort of patients with BE, evaluated from 2002 to 2012. RESULTS We identified 420 patients with BE; 281 (66.9%) of them were men with a mean age of 57.2 ± 15.3 years. Among all BE patients evaluated, 81 (19.3%) had progression to some degree of dysplasia/EAC. The mean follow-up was 5.6 years. Multivariate analysis showed that age (OR = 1.03), cigarette smoking (OR = 3.05), long-segment BE (OR = 4.81), and a visible lesion on BE (OR = 6.94) were associated with progression to dysplasia/EAC. CONCLUSION In Latin patients with BE, age, cigarette smoking, long-segment BE, and the presence of lesions were associated with the presence of dysplasia/EAC.
Collapse
Affiliation(s)
| | | | - Ambrosio R Bernal-Mendez
- Endoscopy, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, MEX
| | | | - Adriana F Romano-Munive
- Endoscopy, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, MEX
| | - Armando Gamboa-Domínguez
- Pathology, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, MEX
| | - Javier Elizondo-Rivera
- Endoscopy, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, MEX
| | - Daniel Briseño-García
- Endoscopy, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, MEX
| | - Felix I Tellez-Ávila
- Gastroenterology and Hepatology, University of Arkansas for Medical Sciences, Little Rock, USA
| |
Collapse
|
30
|
Lin M, Wang L, Liu M, Gu H, Li D, Hou X, Yang H, Shi Y. Risk factors for postoperative infections in esophageal tumor patients. Heliyon 2023; 9:e20741. [PMID: 37842588 PMCID: PMC10568081 DOI: 10.1016/j.heliyon.2023.e20741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/01/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023] Open
Abstract
Postoperative infections (PI) are a serious complication after esophageal cancer surgery, as they might be correlated with an elevated risk of death. While several reports discuss risk factors for PI in esophageal tumor surgery, there is a limited amount of research on overall postoperative infections. Therefore, investigating the factors that influence PI holds great clinical significance. We retrospectively reviewed surgical data from a cohort of 902 patients diagnosed with esophageal tumors. The study included esophageal cancer patients treated in the Department of Thoracic Surgery at Anyang Tumor Hospital from January to December 2021. Preoperative and operative risk factors for PI were evaluated using univariable and multivariable analyses. The overall incidence of PI was 28.3% (255/902). Multivariable logistic regression analysis revealed that smoking and preoperative hospital stays are significant risk factors for PI after esophageal tumor surgery. Smoking and preoperative hospital stays are identified as risk factors for PI following esophageal tumor surgery. Based on our results, we predict that certain groups of patients may have a higher risk of PI following esophageal tumor surgery. Preventive measures or closely monitor of these patients may be required to reduce the incidence of postoperative PI.
Collapse
Affiliation(s)
- Mingzhu Lin
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Lu Wang
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Mengxing Liu
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Huawei Gu
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Dan Li
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Xidong Hou
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Hongye Yang
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Yu Shi
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| |
Collapse
|
31
|
Islam MO, Thangaretnam K, Lu H, Peng D, Soutto M, El-Rifai W, Giordano S, Ban Y, Chen X, Bilbao D, Villarino AV, Schürer S, Hosein PJ, Chen Z. Smoking induces WEE1 expression to promote docetaxel resistance in esophageal adenocarcinoma. Mol Ther Oncolytics 2023; 30:286-300. [PMID: 37732296 PMCID: PMC10507159 DOI: 10.1016/j.omto.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) patients have poor clinical outcomes, with an overall 5-year survival rate of 20%. Smoking is a significant risk factor for EAC. The role of WEE1, a nuclear kinase that negatively regulates the cell cycle in normal conditions, in EAC tumorigenesis and drug resistance is not fully understood. Immunohistochemistry staining shows significant WEE1 overexpression in human EAC tissues. Nicotine, nicotine-derived nitrosamine ketone, or 2% cigarette smoke extract treatment induces WEE1 protein expression in EAC, detected by western blot and immunofluorescence staining. qRT-PCR and reporter assay indicates that smoking induces WEE1 expression through miR-195-5p downregulation in EAC. ATP-Glo cell viability and clonogenic assay confirmed that WEE1 inhibition sensitizes EAC cells to docetaxel treatment in vitro. A TE-10 smoking machine with EAC patient-derived xenograft mouse model demonstrated that smoking induces WEE1 protein expression and resistance to docetaxel in vivo. MK-1775 and docetaxel combined treatment improves EAC patient-derived xenograft mouse survival in vivo. Our findings demonstrate, for the first time, that smoking-induced WEE1 overexpression through miRNA dysregulation in EAC plays an essential role in EAC drug resistance. WEE1 inhibition is a promising therapeutic method to overcome drug resistance and target treatment refractory cancer cells.
Collapse
Affiliation(s)
- Md Obaidul Islam
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Krishnapriya Thangaretnam
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Heng Lu
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Dunfa Peng
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Mohammed Soutto
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Wael El-Rifai
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL 33136, USA
| | - Silvia Giordano
- University of Torino, Candiolo Cancer Institute - FPO, IRCCS, 10060 Candiolo, Italy
| | - Yuguang Ban
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Xi Chen
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Alejandro V. Villarino
- Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Stephan Schürer
- Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
- Institute for Data Science and Computing, University of Miami, Coral Gables, FL 33146, USA
| | - Peter J. Hosein
- Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Zheng Chen
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
32
|
Crowe BR, Krigel A, Li T, Haile R, Al-Ani F, Lebwohl B, Abrams JA, Araujo JL. Veterans with multiple risk factors for Barrett's esophagus are infrequently evaluated with upper endoscopy. Dis Esophagus 2023; 36:doad007. [PMID: 36892169 DOI: 10.1093/dote/doad007] [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: 11/19/2022] [Revised: 01/24/2023] [Accepted: 01/29/2023] [Indexed: 03/10/2023]
Abstract
Recent guidelines recommend screening for patients with chronic gastroesophageal reflux disease who have three or more additional risk factors for Barrett's esophagus (BE). Failure to screen high-risk individuals represents a missed opportunity in esophageal adenocarcinoma prevention and early detection. We aimed to determine the frequency of upper endoscopy and prevalence of BE and esophageal cancer in a cohort of United States veterans who possessed four or more risk factors for BE. All patients at VA New York Harbor Healthcare System with at least four risk factors for BE between 2012 and 2017 were identified. Procedure records were reviewed for upper endoscopies performed between January 2012 and December 2019. Multivariable logistic regression was used to determine risk factors associated with undergoing endoscopy and factors associated with BE and esophageal cancer. 4505 patients with at least four risk factors for BE were included. 828 patients (18.4%) underwent upper endoscopy, of which 42 (5.1%) were diagnosed with BE and 11 (1.3%) with esophageal cancer (10 adenocarcinoma; 1 squamous cell carcinoma). Among individuals who underwent upper endoscopy, risk factors associated with undergoing endoscopy included obesity (OR, 1.79; 95% CI, 1.41-2.30; P < 0.001) and chronic reflux (OR, 3.86; 95% CI, 3.04-4.90; P < 0.001). There were no individual risk factors associated with BE or BE/esophageal cancer. In this retrospective analysis of patients with 4 or more risk factors for BE, fewer than one-fifth of patients underwent upper endoscopy, supporting the need for efforts aimed at improving BE screening rates.
Collapse
Affiliation(s)
- Brooks R Crowe
- Division of Gastroenterology and Hepatology, Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Anna Krigel
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Tian Li
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Rozina Haile
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Firas Al-Ani
- Division of Gastroenterology and Hepatology, Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Benjamin Lebwohl
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Julian A Abrams
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - James L Araujo
- Division of Gastroenterology, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Division of Gastroenterology, Veterans Affairs Puget Sound Healthcare System, Seattle, WA, USA
| |
Collapse
|
33
|
Mozooni Z, Golestani N, Bahadorizadeh L, Yarmohammadi R, Jabalameli M, Amiri BS. The role of interferon-gamma and its receptors in gastrointestinal cancers. Pathol Res Pract 2023; 248:154636. [PMID: 37390758 DOI: 10.1016/j.prp.2023.154636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023]
Abstract
Gastrointestinal malignancies are the most prevalent type of cancer around the world. Even though numerous studies have evaluated gastrointestinal malignancies, the actual underlying mechanism is still unknown. These tumors have a poor prognosis and are frequently discovered at an advanced stage. Globally, there is an increase in the incidence and mortality of gastrointestinal malignancies, including those of the stomach, esophagus, colon, liver, and pancreas. Growth factors and cytokines are signaling molecules that are part of the tumor microenvironment and play a significant role in the development and spread of malignancies. IFN-γ induce its effects by activation of intracellular molecular networks. The main pathway involved in IFN-γ signaling is the JAK/STAT pathway, which regulates the transcription of hundreds of genes and mediates various biological responses. IFN-γ receptor is composed of two IFN-γR1 chains and two IFN-γR2 chains. Binding to IFN-γ, causes the intracellular domains of IFN-γR2 to oligomerize and transphosphorylate with IFN-γR1 which activates downstream signaling components: JAK1 and JAK2. These activated JAKs phosphorylate the receptor, creating binding sites for STAT1. STAT1 is then phosphorylated by JAK, resulting in the formation of STAT1 homodimers (gamma activated factors or GAFs) that translocate to the nucleus and regulate gene expression. The balance between positive and negative regulation of this pathway is crucial for immune responses and tumorigenesis. In this paper, we evaluate the dynamic roles of IFN- γ and its receptors in gastrointestinal cancers and present evidence that inhibiting IFN- γ signaling may be an effective treatment strategy.
Collapse
Affiliation(s)
- Zahra Mozooni
- Institute of Immunology and Infectious Diseases, Antimicrobial Resistance Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Golestani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leyla Bahadorizadeh
- Institute of Immunology and Infectious Diseases, Antimicrobial Resistance Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Internal Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Reyhaneh Yarmohammadi
- Doctoral Student Carolina University Winston, Salem, NC, USA; Skin and Stem Cell Research Center Tehran University of Medical Sciences, Tehran, Iran
| | | | - Bahareh Shateri Amiri
- Department of Internal Medicine, School of Medicine Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Desai M. Ki-67 overexpression for risk stratification of early dysplasia in Barrett's esophagus: Friend or foe? Dis Esophagus 2023; 36:doac079. [PMID: 36336914 DOI: 10.1093/dote/doac079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Madhav Desai
- Department of Gastroenterology, Kansas City VA Medical Center, Kansas City, MO, USA
| |
Collapse
|
35
|
Wu X, Zhu MC, Li GL, Xiong P, Sun W, Zhang N, Zhao B, Li LQ, Fu XN, Zhu M. Treatment and survival analysis for 40-year SEER data on upper esophageal cancer. Front Med (Lausanne) 2023; 10:1128766. [PMID: 37529246 PMCID: PMC10387539 DOI: 10.3389/fmed.2023.1128766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/21/2023] [Indexed: 08/03/2023] Open
Abstract
Background Upper esophageal cancer (UEC) is rare in both Eastern and Western countries. The epidemiological characteristics and long-term survival of UEC patients are less known. In addition, the choice of optimal treatment for UEC has been controversial. Methods Cases of UEC (C15.3 and C15.0) arising during the period from 1973 to 2013 were identified and selected using the SEER database. Student's t-test and Pearson's chi-square test were used to compare the differences in parameters among different groups. Esophageal cancer-specific survival (ECSS) and overall survival (OS) rates were calculated by using the Kaplan-Meier method. Cox proportional hazard regression was used to analyze predictive factors. Results In the past 40 years, the cases of UEC have gradually increased, and the proportion of adenocarcinoma (AD) has gradually increased (from 3.6% to 11.8%, p < 0.001). There has been a significant increase (1973-1982 vs. 2004-2013) in median OS (7 months vs. 10 months, p < 0.001) and median ECSS (7 months vs. 11 months, p < 0.001) among UEC patients from 1973 to 2013. For the impact of different treatments, the results showed that the ECSS and OS of surgery without radiation (SWR) and radiation plus surgery (R+S) were superior to those of radiation without surgery (RWS). Subgroup analysis showed that ECSS and OS were highest among patients treated with SWR compared with R+S and RWS for patients with localized disease. For regional disease, ECSS and OS were highest among patients with R+S compared with SWR or RWS. Among patients with regional-stage squamous cell carcinoma (SCC), OS was higher with neoadjuvant radiotherapy or adjuvant radiotherapy compared with SWR. Multivariate analysis showed that radiotherapy sequence was dependently associated with OS among patients with regional-stage SCC. Conclusion Although the long-term survival of UEC remains poor, it has gradually increased since 1973. This should be closely related to the improvement of medical care over the past 40 years. Different treatment methods have a great influence on the long-term survival of UEC. For localized diseases, surgery may be a better choice. For regional disease, surgery plus adjuvant or neoadjuvant radiotherapy may be more beneficial to improve the long-term prognosis of UEC patients.
Collapse
Affiliation(s)
- Xi Wu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming-Chuang Zhu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Guo-Liang Li
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Peng Xiong
- Intensive Care Unit, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Sun
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Zhao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Le-Qun Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang-Ning Fu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Li A, Cao W. Downregulation of SODD mediates carnosol-induced reduction in cell proliferation in esophageal adenocarcinoma cells. Sci Rep 2023; 13:10580. [PMID: 37386230 PMCID: PMC10310760 DOI: 10.1038/s41598-023-37796-5] [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: 12/14/2022] [Accepted: 06/28/2023] [Indexed: 07/01/2023] Open
Abstract
Esophageal adenocarcinoma carries a poor prognosis associated with a 5-year survival rate of 12.5-20%. Therefore, a new therapeutic modality is needed for this lethal tumor. Carnosol is a phenolic diterpene purified from the herbs such as rosemary and Mountain desert sage and has been shown to have anticancer activities in multiple cancers. In this study we examined the effect of carnosol on cell proliferation in esophageal adenocarcinoma cells. We found that carnosol dose-dependently decreased cell proliferation in FLO-1 esophageal adenocarcinoma cells and significantly increased caspase-3 protein, indicating that carnosol decreases cell proliferation and increases cell apoptosis in FLO-1 cells. Carnosol significantly increased H2O2 production and N-acetyl cysteine, a reactive oxygen species (ROS) scavenger, significantly inhibited carnosol-induced decrease in cell proliferation, indicating that ROS may mediate carnosol-induced decrease in cell proliferation. Carnosol-induced decrease in cell proliferation was partially reversed by NADPH oxidase inhibitor apocynin, suggesting that NADPH oxidases may be partially involved in carnosol's effect. In addition, carnosol significantly downregulated SODD protein and mRNA expression and knockdown of SODD significantly inhibited the carnosol-induced reduction in cell proliferation, suggesting that downregulation of SODD may contribute to carnosol-induced reduction in cell proliferation. We conclude that carnosol dose-dependently decreased cell proliferation and significantly increased caspase-3 protein. Carnosol's effect may be through the overproduction of ROS and the downregulation of SODD. Carnosol might be useful for the treatment of esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Aihua Li
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, 593 Eddy St, APC12, Providence, RI, 02903, USA
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing, China
| | - Weibiao Cao
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, 593 Eddy St, APC12, Providence, RI, 02903, USA.
| |
Collapse
|
37
|
Solfisburg QS, Baldini F, Baldwin-Hunter BL, Lee HH, Park H, Freedberg DE, Lightdale CJ, Korem T, Abrams JA. The Salivary Microbiome and Predicted Metabolite Production are Associated with Progression from Barrett's Esophagus to Esophageal Adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546733. [PMID: 37425673 PMCID: PMC10327009 DOI: 10.1101/2023.06.27.546733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Esophageal adenocarcinoma (EAC) is rising in incidence and associated with poor survival, and established risk factors do not explain this trend. Microbiome alterations have been associated with progression from the precursor Barrett's esophagus (BE) to EAC, yet the oral microbiome, tightly linked to the esophageal microbiome and easier to sample, has not been extensively studied in this context. We aimed to assess the relationship between the salivary microbiome and neoplastic progression in BE to identify microbiome-related factors that may drive EAC development. We collected clinical data and oral health and hygiene history and characterized the salivary microbiome from 250 patients with and without BE, including 78 with advanced neoplasia (high grade dysplasia or early adenocarcinoma). We assessed differential relative abundance of taxa by 16S rRNA gene sequencing and associations between microbiome composition and clinical features and used microbiome metabolic modeling to predict metabolite production. We found significant shifts and increased dysbiosis associated with progression to advanced neoplasia, with these associations occurring independent of tooth loss, and the largest shifts were with the genus Streptococcus. Microbiome metabolic models predicted significant shifts in the metabolic capacities of the salivary microbiome in patients with advanced neoplasia, including increases in L-lactic acid and decreases in butyric acid and L-tryptophan production. Our results suggest both a mechanistic and predictive role for the oral microbiome in esophageal adenocarcinoma. Further work is warranted to identify the biological significance of these alterations, to validate metabolic shifts, and to determine whether they represent viable therapeutic targets for prevention of progression in BE.
Collapse
Affiliation(s)
- Quinn S Solfisburg
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Federico Baldini
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Harry H Lee
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Microbiome and Pathogen Genomics Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Daniel E Freedberg
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles J Lightdale
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
- CIFAR Azrieli Global Scholars Program, CIFAR, Toronto, Canada
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY USA
| |
Collapse
|
38
|
Strasser MK, Gibbs DL, Gascard P, Bons J, Hickey JW, Schürch CM, Tan Y, Black S, Chu P, Ozkan A, Basisty N, Sangwan V, Rose J, Shah S, Camilleri-Broet S, Fiset PO, Bertos N, Berube J, Djambazian H, Li R, Oikonomopoulos S, Fels-Elliott DR, Vernovsky S, Shimshoni E, Collyar D, Russell A, Ragoussis I, Stachler M, Goldenring JR, McDonald S, Ingber DE, Schilling B, Nolan GP, Tlsty TD, Huang S, Ferri LE. Concerted epithelial and stromal changes during progression of Barrett's Esophagus to invasive adenocarcinoma exposed by multi-scale, multi-omics analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544265. [PMID: 37333362 PMCID: PMC10274886 DOI: 10.1101/2023.06.08.544265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Esophageal adenocarcinoma arises from Barrett's esophagus, a precancerous metaplastic replacement of squamous by columnar epithelium in response to chronic inflammation. Multi-omics profiling, integrating single-cell transcriptomics, extracellular matrix proteomics, tissue-mechanics and spatial proteomics of 64 samples from 12 patients' paths of progression from squamous epithelium through metaplasia, dysplasia to adenocarcinoma, revealed shared and patient-specific progression characteristics. The classic metaplastic replacement of epithelial cells was paralleled by metaplastic changes in stromal cells, ECM and tissue stiffness. Strikingly, this change in tissue state at metaplasia was already accompanied by appearance of fibroblasts with characteristics of carcinoma-associated fibroblasts and of an NK cell-associated immunosuppressive microenvironment. Thus, Barrett's esophagus progresses as a coordinated multi-component system, supporting treatment paradigms that go beyond targeting cancerous cells to incorporating stromal reprogramming.
Collapse
|
39
|
Akhtar S, Al-Shammari A, Al-Huraiti M, Al-Anjery F, Al-Sabah S, Memon A, Siddique I. Disparities in oesophageal cancer risk by age, sex, and nativity in Kuwait:1980-2019. BMC Cancer 2023; 23:293. [PMID: 37004010 PMCID: PMC10064499 DOI: 10.1186/s12885-023-10770-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND This cross-sectional cohort study assessed the inequalities in oesophageal carcinoma risk by age, sex and nativity in Kuwait: 1980-2019. METHODS Using oesophageal cancer incidence data from the Kuwait National Cancer Registry, relevant Kuwaiti population data and World Standard Population as a reference, age-standardized incidence rates (ASIR) (per 100,000 person-years) overall and by subcohorts were computed. The incident oesophageal cancer cases count was overdispersed with excessive structural zeros, therefore, it was analyzed using multivariable zero-inflated negative binomial (ZINB) model. RESULTS Overall ASIR of oesophageal cancer was 10.51 (95% CI: 6.62-14.41). The multivariable ZINB model showed that compared with the younger age category (< 30 years), the individuals in higher age groups showed a significant (p < 0.001) increasing tendency to develop the oesophageal cancer. Furthermore, compared with the non-Kuwaiti residents, the Kuwaiti nationals were significantly (p < 0.001) more likely to develop oesophageal cancer during the study period. Moreover, compared with 1980-84 period, ASIRs steadily and significantly (p < 0.005) declined in subsequent periods till 2015-19. CONCLUSIONS A high incidence of oesophageal cancer was recorded in Kuwait, which consistently declined from 1980 to 2019. Older adults (aged ≥ 60 years) and, Kuwaiti nationals were at high risk of oesophageal cancer. Focused educational intervention may minimize oesophageal cancer incidence in high-risk groups in this and other similar settings. Future studies may contemplate to evaluate such an intervention.
Collapse
Affiliation(s)
- Saeed Akhtar
- Department of Community Medicine and Behavioural Sciences, College of Medicine, Kuwait University, PO Box 24923, Safat, 13110, Kuwait.
| | - Ahmad Al-Shammari
- Department of Surgery, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | | | | | - Salman Al-Sabah
- Department of Surgery, College of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Anjum Memon
- Department of Primary Care and Public Health, Brighton and Sussex Medical School, Brighton, UK
| | - Iqbal Siddique
- Department of Medicine, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
40
|
Sharma P, Falk GW, Bhor M, Ozbay AB, Latremouille-Viau D, Guérin A, Shi S, Elvekrog MM, Limburg P. Real-world upper endoscopy utilization patterns among patients with gastroesophageal reflux disease, Barrett esophagus, and Barrett esophagus-related esophageal neoplasia in the United States. Medicine (Baltimore) 2023; 102:e33072. [PMID: 36961193 PMCID: PMC10036066 DOI: 10.1097/md.0000000000033072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/02/2023] [Indexed: 03/25/2023] Open
Abstract
This study fills a gap in literature by providing contemporary real-world evidence on the prevalence of patients with gastroesophageal reflux disease (GERD), Barrett esophagus (BE), and Barrett esophagus-related neoplasia (BERN) and their upper endoscopy utilization patterns in the United States. A retrospective cohort study design was used: adults with GERD, nondysplastic Barrett esophagus (NDBE), and BERN (indefinite for dysplasia [IND], low-grade dysplasia [LGD], high-grade dysplasia [HGD], or esophageal adenocarcinoma [EAC]) were identified from the MarketScan databases (January 01, 2015-December 31, 2019). For each disease stage, prevalence of adults in commercial claims by calendar year, annual number of upper endoscopies per patient and time between upper endoscopies were reported. In 2019, in commercial claims (N = 12,363,227), the annual prevalence rate of GERD was 13.7% and 0.70% for BE/BERN, among which, 87.1% had NDBE, 6.8% had IND, 2.3% had LGD, 1.0% had HGD, and 2.8% had EAC. From 2015-2019, the study included 3,310,385 patients with GERD, 172,481 with NDBE, 11,516 with IND, 4332 with LGD, 1549 with HGD, and 11,676 with EAC. Annual mean number of upper endoscopies was 0.20 per patient for GERD, 0.37 per patient for NDBE, 0.43 for IND, 0.58 for LGD, and 0.87 for HGD. Median time (months) to second upper endoscopy was 38.10 for NDBE, 36.63 for IND, 22.63 for LGD, and 11.90 for HGD. Upper endoscopy utilization increased from GERD to BE to BERN, and time between upper endoscopies decreased as the disease stage progressed from BE to BERN, with less frequent utilization in BERN than what would be expected from guideline recommendations for surveillance.
Collapse
Affiliation(s)
- Prateek Sharma
- Department of Gastroenterology, University of Kansas School of Medicine and VA Medical Center, Kansas City, MO
| | - Gary W. Falk
- Perelman School of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sharma P, Falk GW, Bhor M, Ozbay AB, Latremouille-Viau D, Guerin A, Shi S, Elvekrog MM, Limburg P. Healthcare Resource Utilization and Costs Among Patients With Gastroesophageal Reflux Disease, Barrett's Esophagus, and Barrett's Esophagus-Related Neoplasia in the United States. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2023; 10:51-58. [PMID: 36883055 PMCID: PMC9985944 DOI: 10.36469/001c.68191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Background: Gastroesophageal reflux disease (GERD) is a risk factor for Barrett's esophagus (BE) and BE-related neoplasia (BERN). Objectives: This study aimed to evaluate healthcare resource utilization (HRU) and costs associated with GERD, BE, and BERN in the United States. Methods: Adult patients with GERD, nondysplastic BE (NDBE), and BERN (including indefinite for dysplasia [IND], low-grade dysplasia [LGD], high-grade dysplasia [HGD] or esophageal adenocarcinoma [EAC]), were identified from a large US administrative claims database, the IBM Truven Health MarketScan® databases (Q1/2015-Q4/2019). Patients were categorized into the corresponding mutually exclusive EAC-risk/diagnosis cohorts based on the most advanced stage from GERD to EAC using diagnosis codes in medical claims. Disease-related HRU and costs (2020 USD) were calculated for each cohort. Results: Patients were categorized into the following EAC-risk/diagnosis cohorts: 3 310 385 into GERD, 172 481 into NDBE, 11 516 into IND, 4332 into LGD, 1549 into HGD, and 11 676 into EAC. Disease-related annual mean number of inpatient admissions, office visits, and emergency department visits by cohort were 0.09, 1.45, and 0.19 for GERD; 0.08, 1.55, and 0.10 for NDBE; 0.10, 1.92, and 0.13 for IND; 0.09, 2.05, and 0.10 for LGD; 0.12, 2.16, and 0.14 for HGD; and 1.43, 6.27, and 0.87 for EAC. Disease-related annual mean total healthcare costs by cohort were $6955 for GERD, $8755 for NDBE, $9675 for IND, $12 241 for LGD, $24 239 for HGD, and $146 319 for EAC. Discussion: Patients with GERD, BE, and BERN had important HRU and costs, including inpatient admissions and office visits. As patients progressed to more advanced stages, there was substantially higher disease-related resource utilization, with associated costs being 16 times higher in patients with EAC than those with NDBE. Conclusions: Findings suggest the need for early identification of high-risk individuals prior to progression to EAC to potentially improve clinical and economic outcomes in this population.
Collapse
Affiliation(s)
- Prateek Sharma
- University of Kansas School of Medicine and VA Medical Center, Kansas City, Missouri
| | - Gary W Falk
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | | | | | | | | | - Sherry Shi
- Analysis Group, Montréal, Québec, Canada
| | | | | |
Collapse
|
42
|
Endoscopic therapy replaces surgery for clinical T1 oesophageal cancer in the Netherlands: a nationwide population-based study. Surg Endosc 2023:10.1007/s00464-023-09914-x. [PMID: 36849563 DOI: 10.1007/s00464-023-09914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/28/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND Endoscopic resection for early oesophageal cancer was introduced around 2000 in the Netherlands. The scientific question was how the treatment and survival of early oesophageal and gastro-oesophageal junction cancer has changed over time in the Netherlands. METHODS Data were obtained from the nationwide population-based Netherlands Cancer Registry. All patients diagnosed with clinical in situ or T1 oesophageal or GOJ cancer without lymph node or distance metastasis during the study period (2000-2014) were extracted. Primary outcome parameters were the trends in treatment modalities over time and relative survival of each treatment regime. RESULTS A total of 1020 patients were diagnosed with a clinical in situ or T1 oesophageal or gastro-oesophageal junction cancer without lymph node or distance metastasis. The proportion of patients who received endoscopic treatment increased from 2.5% in 2000 to 58.1% in 2014. During the same period the proportion of patients who received surgery decreased from 57.5 to 23.1%. Five-year relative survival of all patients was 69%. Five-year relative survival after endoscopic therapy was 83% and after surgery 80%. Relative excess risk analyses showed no significant difference in survival between patients in the endoscopic therapy group and patients in the surgery group after adjustment for age, sex, clinical TNM classification, morphology and tumour location (RER 1.15; CI 0.76-1.75; p 0.76). CONCLUSION Our results demonstrate an increase in endoscopic treatment and a decrease of surgical treatment for in situ and T1 oesophageal/GOJ cancer between 2000-2014 in the Netherlands. The relative 5-year survival after endoscopic treatment is high (83%) and comparable with surgery (80%).
Collapse
|
43
|
Thawani R, Agrawal N, Taflin NF, Kardosh A, Chen EY. Application of Value Framework to Phase III Trials of Immune Checkpoint Inhibitors in Esophageal and Gastric Cancer. Oncologist 2023; 28:40-47. [PMID: 36130326 PMCID: PMC9847562 DOI: 10.1093/oncolo/oyac187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/10/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Recent trials testing immune-checkpoint inhibitors in esophago-gastric malignancies have shown mixed results. We aim to assess key subgroups using the ASCO Net Health Benefit Score (NHBS) and ESMO Magnitude of Clinical Benefit Scale (MCBS). MATERIALS AND METHODS A search for phase III trials of FDA-approved anti-PD-1 or anti-PD-L1 drugs in esophago-gastric cancer trials was identified using www.clinicaltrials.gov. These published studies were scored using the ASCO NHBS and ESMO MCBS. The ASCO NHBS scores were compared by primary site of cancer (esophageal vs gastric) and PD-L1 expression using the Mann-Whitney test and the ESMO-MCBS grading, by Fisher's Exact test. RESULTS Fifteen of 45 clinical trials were included. Of them, 6 were primarily esophageal cancer trials, and 9 were primarily gastric cancer trials. Ten stratified their analysis based on PD-L1 expression. The ASCO NHBS score was higher (mean 40, range 20 to 56.6 vs. mean 12, range -1.1 to 18.4, P < .01) for esophageal cancer than gastric cancer. No difference was observed in survival and response endpoints between the 2 groups. Similarly, the ESMO MCBS scored higher for esophageal cancer group than gastric cancer (P < .05). Additionally, the scores were higher in those with high PD-L1 expression vs. low PD-L1 (mean 36, range 11.2-66.6 vs. mean 14, range -19.5 to 43.6, P < .05). CONCLUSION The ASCO NHB and ESMO scores were consistently higher among esophageal cancer trials than gastric cancer trials and in those with high PD-L1 expression than low expression. Histology and PD-L1 expression should be considered when discussing value of immunotherapy to patients.
Collapse
Affiliation(s)
- Rajat Thawani
- Knight Cancer Institute, Division of Hematology and Oncology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Neha Agrawal
- Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Nicholas F Taflin
- Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Adel Kardosh
- Knight Cancer Institute, Division of Hematology and Oncology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Emerson Y Chen
- Corresponding author: Emerson Y. Chen, MD, MCR, 3181 SW Sam Jackson Park Road, OC14HO, Portland, OR 97239, USA. Tel: +1 503 418 1297;
| |
Collapse
|
44
|
Grasa L, Chueca E, Arechavaleta S, García-González MA, Sáenz MÁ, Valero A, Hördnler C, Lanas Á, Piazuelo E. Antitumor effects of lactate transport inhibition on esophageal adenocarcinoma cells. J Physiol Biochem 2023; 79:147-161. [PMID: 36342616 PMCID: PMC9905156 DOI: 10.1007/s13105-022-00931-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/25/2022] [Indexed: 11/09/2022]
Abstract
As a consequence of altered glucose metabolism, cancer cell intake is increased, producing large amounts of lactate which is pumped out the cytosol by monocarboxylate transporters (MCTs). MCT 1 and MCT4 are frequently overexpressed in tumors, and recently, MCT inhibition has been reported to exert antineoplastic effects. In the present study, MCT1 and MCT4 levels were assessed in esophageal adenocarcinoma (EAC) cells and the effects of the MCT-1 selective inhibitor AZD3965, hypoxia, and a glucose overload were evaluated in vitro. Two EAC cell lines (OE33 and OACM5.1C) were treated with AZD3965 (10-100 nM) under different conditions (normoxia/hypoxia) and also different glucose concentrations, and parameters of cytotoxicity, oxidative stress, intracellular pH (pHi), and lactate levels were evaluated. MCT1 was present in both cell lines whereas MCT4 was expressed in OE33 cells and only in a small proportion of OACM5.1C cells. Glucose addition did not have any effect on apoptosis nor cell proliferation. AZD3965 increased apoptosis and reduced proliferation of OACM5.1C cells, effects which were abrogated when cells were growing in hypoxia. MCT1 inhibition increased intracellular lactate levels in all the cells evaluated, but this increase was higher in cells expressing only MCT1 and did not affect oxidative stress. AZD3965 induced a decrease in pHi of cells displaying low levels of MCT4 and also increased the sodium/hydrogen exchanger 1 (NHE-1) expression on these cells. These data provide in vitro evidence supporting the potential of MCT inhibitors as novel antineoplastic drugs for EAC and highlight the importance of achieving a complete MCT inhibition.
Collapse
Affiliation(s)
- Laura Grasa
- IIS Aragón, Instituto de Investigación Sanitaria Aragón, Avenida San Juan Bosco 13, 50009, Saragossa, Spain.
- Faculty of Veterinary Medicine, University of Zaragoza, Calle Miguel Servet, 177, 50013, Saragossa, Spain.
| | - Eduardo Chueca
- IIS Aragón, Instituto de Investigación Sanitaria Aragón, Avenida San Juan Bosco 13, 50009, Saragossa, Spain
- CIBERehd, Instituto de Salud Carlos III, Calle Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Samantha Arechavaleta
- IIS Aragón, Instituto de Investigación Sanitaria Aragón, Avenida San Juan Bosco 13, 50009, Saragossa, Spain
- CIBERehd, Instituto de Salud Carlos III, Calle Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - María Asunción García-González
- IIS Aragón, Instituto de Investigación Sanitaria Aragón, Avenida San Juan Bosco 13, 50009, Saragossa, Spain
- CIBERehd, Instituto de Salud Carlos III, Calle Monforte de Lemos 3-5, 28029, Madrid, Spain
- IACS Aragón, Instituto Aragonés de Ciencias de La Salud, Avenida San Juan Bosco 13, 50009, Saragossa, Spain
| | - María Ángeles Sáenz
- Faculty of Medicine, University of Zaragoza, Calle de Pedro Cerbuna, 12, 50009, Saragossa, Spain
| | - Alberto Valero
- Servicio de Patología, Hospital Universitario Miguel Servet, Paseo Isabel La Católica 1-3, 50009, Saragossa, Spain
| | - Carlos Hördnler
- Servicio de Patología, Hospital Universitario Miguel Servet, Paseo Isabel La Católica 1-3, 50009, Saragossa, Spain
| | - Ángel Lanas
- IIS Aragón, Instituto de Investigación Sanitaria Aragón, Avenida San Juan Bosco 13, 50009, Saragossa, Spain
- CIBERehd, Instituto de Salud Carlos III, Calle Monforte de Lemos 3-5, 28029, Madrid, Spain
- Faculty of Medicine, University of Zaragoza, Calle de Pedro Cerbuna, 12, 50009, Saragossa, Spain
| | - Elena Piazuelo
- IIS Aragón, Instituto de Investigación Sanitaria Aragón, Avenida San Juan Bosco 13, 50009, Saragossa, Spain
- CIBERehd, Instituto de Salud Carlos III, Calle Monforte de Lemos 3-5, 28029, Madrid, Spain
- IACS Aragón, Instituto Aragonés de Ciencias de La Salud, Avenida San Juan Bosco 13, 50009, Saragossa, Spain
| |
Collapse
|
45
|
Zyla RE, Kalimuthu SN. Barrett’s Esophagus and Esophageal Adenocarcinoma: A Histopathological Perspective. Thorac Surg Clin 2022; 32:413-424. [DOI: 10.1016/j.thorsurg.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
46
|
Yadlapati R, Hubscher E, Pelletier C, Jacob R, Brackley A, Shah S. Induction and maintenance of healing in erosive esophagitis in the United States. Expert Rev Gastroenterol Hepatol 2022; 16:967-980. [PMID: 36254610 DOI: 10.1080/17474124.2022.2134115] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Erosive esophagitis (EE) occurs when refluxate from the stomach causes T-lymphocyte infiltration of the esophageal mucosa, resulting in mucosal breaks. Currently, therapy with proton-pump inhibitors (PPIs) is the standard treatment for EE in the United States, but few comprehensive reviews exist on the efficacy of PPIs in US populations. Here, we present the most contemporary, thorough analysis of PPI efficacy rates, and identify and characterize patient subgroups at risk for poor healing outcomes. AREAS COVERED We searched the literature to identify studies reporting rates of endoscopic healing and maintained healing of EE with PPI therapies in the US and found a paucity of recent evidence and real-world evidence. Twenty-two studies from 2009 and earlier were included in the final dataset. EXPERT OPINION Rates of EE healing with PPIs were highest after 8 weeks of treatment, with over 80% of patients in most treatment arms demonstrating endoscopic healing, compared to lower efficacy (<80%) at 4 weeks. Rates of maintained healing with PPIs at 6 and 12 months were mostly lower than 80%, although the data were limited. Symptomatic patients and those with severe EE were less likely to achieve healing. Obese patients experienced similar healing rates as non-obese patients.
Collapse
Affiliation(s)
- Rena Yadlapati
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | | | - Corey Pelletier
- Health Economics and Outcomes Research, Phathom Pharmaceuticals, Florham Park, New Jersey, USA
| | - Rinu Jacob
- Health Economics and Outcomes Research, Phathom Pharmaceuticals, Florham Park, New Jersey, USA
| | - Allison Brackley
- Real-World Advanced Analytics, Cytel, Inc, Waltham, Massachusetts, USA
| | - Shailja Shah
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
47
|
Esophageal OCT Imaging Using a Paddle Probe Externally Attached to Endoscope. Dig Dis Sci 2022; 67:4805-4812. [PMID: 35084606 PMCID: PMC10015416 DOI: 10.1007/s10620-021-07372-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/18/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Endoscopic surveillance of Barrett's esophagus (BE) by white light examination is insufficient to diagnose dysplastic change. In this work, we describe an optical imaging method to obtain high-resolution cross-sectional imaging using a paddle-shaped probe affixed to the endoscope tip. METHODS We integrated Optical Coherence Tomography (OCT), an optical imaging method that produces cross-sectional images, into a paddle probe attached to video endoscope. We acquired images of esophageal epithelium from patients undergoing routine upper GI endoscopy. Images were classified by a reviewer blinded to patient identity and condition, and these results were compared with clinical diagnosis. RESULTS We successfully captured epithelial OCT images from 30 patients and identified features consistent with both squamous epithelium and Barrett's esophagus. Our blinded image reviewer classified BE versus non-BE with 91.5% accuracy (65/71 image regions), including sensitivity of 84.6% for BE (11/13) and a specificity of 93.1% (54/58). However, in 16 patients, intubation of the probe into the esophagus could not be achieved. CONCLUSIONS A paddle probe is a feasible imaging format for acquiring cross-sectional OCT images from the esophagus and can provide a structural assessment of BE and non-BE tissue. Probe form factor is the current limiting obstacle, but could be addressed by further miniaturization.
Collapse
|
48
|
Yu M, Moinova HR, Willbanks A, Canon VK, Wang T, Carter K, Kaz A, Reddi D, Inadomi J, Luebeck G, Iyer PG, Canto MI, Wang JS, Shaheen NJ, Thota PN, Willis JE, LaFramboise T, Chak A, Markowitz SD, Grady WM. Novel DNA Methylation Biomarker Panel for Detection of Esophageal Adenocarcinoma and High-Grade Dysplasia. Clin Cancer Res 2022; 28:3761-3769. [PMID: 35705525 PMCID: PMC9444948 DOI: 10.1158/1078-0432.ccr-22-0445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/10/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Current endoscopy-based screening and surveillance programs have not been proven effective at decreasing esophageal adenocarcinoma (EAC) mortality, creating an unmet need for effective molecular tests for early detection of this highly lethal cancer. We conducted a genome-wide methylation screen to identify novel methylation markers that distinguish EAC and high-grade dysplasia (HGD) from normal squamous epithelium (SQ) or nondysplastic Barrett's esophagus (NDBE). EXPERIMENTAL DESIGN DNA methylation profiling of samples from SQ, NDBE, HGD, and EAC was performed using HM450 methylation arrays (Illumina) and reduced-representation bisulfate sequencing. Ultrasensitive methylation-specific droplet digital PCR and next-generation sequencing (NGS)-based bisulfite-sequencing assays were developed to detect the methylation level of candidate CpGs in independent esophageal biopsy and endoscopic brushing samples. RESULTS Five candidate methylation markers were significantly hypermethylated in HGD/EAC samples compared with SQ or NDBE (P < 0.01) in both esophageal biopsy and endoscopic brushing samples. In an independent set of brushing samples used to construct biomarker panels, a four-marker panel (model 1) demonstrated sensitivity of 85.0% and 90.8% for HGD and EACs respectively, with 84.2% and 97.9% specificity for NDBE and SQ respectively. In a validation set of brushing samples, the panel achieved sensitivity of 80% and 82.5% for HGD and EAC respectively, at specificity of 67.6% and 96.3% for NDBE and SQ samples. CONCLUSIONS A novel DNA methylation marker panel differentiates HGD/EAC from SQ/NDBE. DNA-methylation-based molecular assays hold promise for the detection of HGD/EAC using esophageal brushing samples.
Collapse
Affiliation(s)
- Ming Yu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Helen R. Moinova
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Amber Willbanks
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Victoria K. Canon
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ting Wang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kelly Carter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Andrew Kaz
- Gastroenterology Section, VA Puget Sound Health Care System, WA, USA,Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Deepti Reddi
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - John Inadomi
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Georg Luebeck
- Public Heath Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Prasad G. Iyer
- Barrett’s Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Marcia I. Canto
- Division of Gastroenterology and Hepatology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jean S. Wang
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Nicholas J. Shaheen
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina, Chapel Hill, NC
| | | | - Joseph E. Willis
- Department of Pathology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Thomas LaFramboise
- Department of Genetics and Genome Sciences, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA,Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Amitabh Chak
- Gastroenterology Division, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Sanford D. Markowitz
- Seidman Cancer Center, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - William M. Grady
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
49
|
Liu J, Liu L, Su Y, Wang Y, Zhu Y, Sun X, Guo Y, Shan J. IL-33 Participates in the Development of Esophageal Adenocarcinoma. Pathol Oncol Res 2022; 28:1610474. [PMID: 36110250 PMCID: PMC9469785 DOI: 10.3389/pore.2022.1610474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022]
Abstract
Background: The progression from chronic gastroesophageal reflux disease (GERD) to Barrett esophagus (BE) and esophageal adenocarcinoma (EAC) is an inflammatory-driven neoplastic change. Interleukin-33 (IL-33) has identified as a crucial factor in several inflammatory disorders and malignancies.Methods: The high-density tissue microarray of the human EAC was analyzed with IL-33 immunohistochemistry staining (IHC). By anastomosing the jejunum with the esophagus, the rat model of EAC with mixed gastroduodenal reflux was established. The expression of IL-33 was determined using quantitative real-time polymerase chain reaction (RT-qPCR), western blot (WB), IHC and enzyme-linked immunosorbent assay (ELISA). Esophageal adenocarcinoma cells (OE19 and OE33) and human esophageal epithelial cells (HEECs) were used.Results: In the cytoplasm of human EAC tissue, IL-33 expression was substantially greater than in adjacent normal tissue. In rat model, the expression of IL-33 in the EAC group was considerably greater than in the control group, and this expression increased with the upgrade of pathological stage. In in vitro experiment, the mRNA and protein levels of IL-33 were considerably greater in OE19 and OE33 than in HEECs. The stimulation of IL-33 enhanced the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of OE19 and OE33, but soluble ST2 (sST2) inhibited these effects. IL-33 stimulated the release of IL-6 by OE19 and OE33 cells.Conclusion: This study demonstrated the overexpression of IL-33 in the transition from GERD to EAC and that IL-33 promoted carcinogenesis in EAC cells through ST2. IL-33 might be a possible preventive target for EAC.
Collapse
Affiliation(s)
- Jia Liu
- School of Medicine, Southwest Jiaotong University, Chengdu, China
- The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Lei Liu
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Yang Su
- School of Medicine, Southwest Jiaotong University, Chengdu, China
- The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Yi Wang
- North Sichuan Medical College, Nanchong, China
| | - Yuchun Zhu
- North Sichuan Medical College, Nanchong, China
| | - Xiaobin Sun
- Department of Gastroenterology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Yuanbiao Guo
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Jing Shan
- Department of Gastroenterology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
- *Correspondence: Jing Shan,
| |
Collapse
|
50
|
Tanaka I, Hirasawa D, Suzuki K, Unno S, Inoue S, Ito S, Togashi J, Akahira J, Fujishima F, Matsuda T. Which factors make Barrett's esophagus lesions difficult to diagnose? Endosc Int Open 2022; 10:E1045-E1052. [PMID: 35979028 PMCID: PMC9377827 DOI: 10.1055/a-1843-0334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 05/03/2022] [Indexed: 11/06/2022] Open
Abstract
Background and study aims Although the Japan Esophageal Society's magnifying endoscopic classification for Barrett's epithelium (JES-BE) offers high diagnostic accuracy, some cases are challenging to diagnose as dysplastic or non-dysplastic in daily clinical practice. Therefore, we investigated the diagnostic accuracy of this classification and the clinicopathological features of Barrett's esophagus cases that are difficult to diagnose correctly. Patients and methods Five endoscopists with experience with fewer than 10 cases of magnifying observation for superficial Barrett's esophageal carcinoma reviewed 132 images of Barrett's mucosa or carcinoma (75 dysplastic and 57 non-dysplastic cases) obtained using high-definition magnification endoscopy with narrow-band imaging (ME-NBI). They diagnosed each image as dysplastic or non-dysplastic according to the JES-BE classification, and the diagnostic accuracy was calculated. To identify risk factors for misdiagnosed images, images with a correct rate of less than 40 % were defined as difficult-to-diagnose, and those with 60 % or more were defined as easy-to-diagnose. Logistic regression analysis was performed to identify risk factors for difficult-to-diagnose images. Results The sensitivity, specificity and overall accuracy were 67 %, 80 % and 73 %, respectively. Of the 132 ME-NBI images, 34 (26 %) were difficult-to-diagnose and 99 (74 %) were easy-to-diagnose. Logistic regression analysis showed low-grade dysplasia (LGD) and high-power magnification images were each significant risk factors for difficult-to-diagnose images (OR: 6.80, P = 0.0017 and OR: 3.31, P = 0.0125, respectively). Conclusions This image assessment study suggested feasibility of the JES-BE classification for diagnosis of Barrett's esophagus by non-expert endoscopists and risk factors for difficult diagnosis as high-power magnification and LGD histology. For non-experts, high-power magnification images are better evaluated in combination with low-power magnification images.
Collapse
Affiliation(s)
- Ippei Tanaka
- Department of Gastroenterology, Sendai Kousei Hospital, Miyagi, Japan
| | - Dai Hirasawa
- Department of Gastroenterology, Sendai Kousei Hospital, Miyagi, Japan
| | - Kenjiro Suzuki
- Department of Gastroenterology, Sendai Kousei Hospital, Miyagi, Japan
| | - Syuhei Unno
- Department of Gastroenterology, Sendai Kousei Hospital, Miyagi, Japan
| | - Shin Inoue
- Department of Gastroenterology, Sendai Kousei Hospital, Miyagi, Japan
| | - Satoshi Ito
- Department of Gastroenterology, Sendai Kousei Hospital, Miyagi, Japan
| | - Jyunichi Togashi
- Department of Gastroenterology, Sendai Kousei Hospital, Miyagi, Japan
| | - Junichi Akahira
- Department of Anatomic Pathology, Sendai Kousei Hospital, Miyagi, Japan
| | - Fumiyoshi Fujishima
- Department of Anatomic Pathology, Tohoku University School of Medicine, Miyagi, Japan
| | - Tomoki Matsuda
- Department of Gastroenterology, Sendai Kousei Hospital, Miyagi, Japan
| |
Collapse
|