1
|
Liu J, Huang B, Ding F, Li Y. Environment factors, DNA methylation, and cancer. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023; 45:7543-7568. [PMID: 37715840 DOI: 10.1007/s10653-023-01749-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023]
Abstract
Today, the rapid development of science and technology and the rapid change in economy and society are changing the way of life of human beings and affecting the natural, living, working, and internal environment on which human beings depend. At the same time, the global incidence of cancer has increased significantly yearly, and cancer has become the number one killer that threatens human health. Studies have shown that diet, living habits, residential environment, mental and psychological factors, intestinal flora, genetics, social factors, and viral and non-viral infections are closely related to human cancer. However, the molecular mechanisms of the environment and cancer development remain to be further explored. In recent years, DNA methylation has become a key hub and bridge for environmental and cancer research. Some environmental factors can alter the hyper/hypomethylation of human cancer suppressor gene promoters, proto-oncogene promoters, and the whole genome, causing low/high expression or gene mutation of related genes, thereby exerting oncogenic or anticancer effects. It is expected to develop early warning markers of cancer environment based on DNA methylation, thereby providing new methods for early detection of cancers, diagnosis, and targeted therapy. This review systematically expounds on the internal mechanism of environmental factors affecting cancer by changing DNA methylation, aiming to help establish the concept of cancer prevention and improve people's health.
Collapse
Affiliation(s)
- Jie Liu
- Department of General Surgery, Second Hospital of Lanzhou University, Lan Zhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lan Zhou, China
| | - Binjie Huang
- Department of General Surgery, Second Hospital of Lanzhou University, Lan Zhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lan Zhou, China
| | - Feifei Ding
- Department of General Surgery, Second Hospital of Lanzhou University, Lan Zhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lan Zhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lan Zhou, China.
- Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lan Zhou, China.
| |
Collapse
|
2
|
Li C, Liu Z, Xu G, Wu S, Peng Y, Wu R, Zhao S, Liao X, Lin R. Aberrant DNA methylation and expression of EYA4 in gastric cardia intestinal metaplasia. Saudi J Gastroenterol 2022; 28:456-465. [PMID: 36453428 PMCID: PMC9843510 DOI: 10.4103/sjg.sjg_228_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Intestinal metaplasia (IM) of the gastric cardia is an important premalignant lesion. However, there is limited information concerning its epidemiological and molecular features. Herein, we aimed to provide an overview of the epidemiological data for gastric cardiac IM and evaluate the role of EYA transcriptional coactivator and phosphatase 4 (EYA4) as an epigenetic biomarker for gastric cardiac IM. METHODS The study was conducted in the context of the gastric cardiac precancerous lesion program in southern China, which included 718 non-cancer participants, who undertook endoscopic biopsy and pathological examination in three endoscopy centers, between November 2018 and November 2021. Pyrosequencing and immunohistochemistry were performed to examine the DNA methylation status and protein expression level of EYA4. RESULTS Gastric cardiac IM presented in 14.1% (101/718) of participants and was more common among older (>50 years; 22.0% [95% CI: 17.8-26.8]) than younger participants (≤50 years; 6.7% [95% CI: 4.5-9.9]; P < 0.001). IM was more common in male participants (16.9% [95% CI: 13.2-21.3] vs. 11.3% [95% CI: 8.3-15.1]; P = 0.04). Pyrosequencing revealed that IM tissues exhibited significantly higher DNA methylation levels in EYA4 gene than normal tissues (P = 0.016). Further, the protein expression level of EYA4 was reduced in IM and absent in intraepithelial neoplasia tissues compared to normal tissues (P < 0.001). CONCLUSIONS Detection rates of gastric cardiac IM increase with age and are higher in men. Our findings highlight the important role of promoter hypermethylation and downregulation of EYA4 in gastric cardiac IM development.
Collapse
Affiliation(s)
- Chenxi Li
- Department of Pathology, Shantou University Medical College, Shantou, P.R. China
| | - Zhaohui Liu
- Department of Gastroenterology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, P.R. China
| | - Guohua Xu
- Department of Gastroenterology, Huiyang Sanhe Hospital, Huizhou, P.R. China
| | - Shibin Wu
- Department of Gastroenterology, Huiyang Sanhe Hospital, Huizhou, P.R. China
| | - Yunhui Peng
- Department of Gastroenterology, Huiyang Sanhe Hospital, Huizhou, P.R. China
| | - Ruinuan Wu
- Department of Pathology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, P.R. China
| | - Shukun Zhao
- Department of Pathology, Shantou University Medical College, Shantou, P.R. China
| | - Xiaoqi Liao
- Department of Pathology, Shantou University Medical College, Shantou, P.R. China
| | - Runhua Lin
- Department of Pathology, Shantou University Medical College, Shantou, P.R. China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou, P.R. China
- Address for correspondence: Dr. Runhua Lin, Department of Pathology, Shantou University Medical College, No. 22, Xinling Road, Shantou 515041, P.R. China. E-mail:
| |
Collapse
|
3
|
Schmidt M, Hackett RJ, Baker AM, McDonald SAC, Quante M, Graham TA. Evolutionary dynamics in Barrett oesophagus: implications for surveillance, risk stratification and therapy. Nat Rev Gastroenterol Hepatol 2022; 19:95-111. [PMID: 34728819 DOI: 10.1038/s41575-021-00531-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Cancer development is a dynamic evolutionary process characterized by marked intratumoural heterogeneity at the genetic, epigenetic and phenotypic levels. Barrett oesophagus, the pre-malignant condition to oesophageal adenocarcinoma (EAC), is an exemplary system to longitudinally study the evolution of malignancy. Evidence has emerged of Barrett oesophagus lesions pre-programmed for progression to EAC many years before clinical detection, indicating a considerable window for therapeutic intervention. In this Review, we explore the mechanisms underlying clonal expansion and contraction that establish the Barrett oesophagus clonal mosaicism over time and space and discuss intrinsic genotypic and extrinsic environmental drivers that direct the evolutionary trajectory of Barrett oesophagus towards a malignant phenotype. We propose that understanding and exploiting the evolutionary dynamics of Barrett oesophagus will identify novel therapeutic targets, improve prognostic tools and offer the opportunity for personalized surveillance programmes geared to prevent progression to EAC.
Collapse
Affiliation(s)
- Melissa Schmidt
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), München, Germany
| | - Richard J Hackett
- Clonal Dynamics in Epithelia Group; Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ann-Marie Baker
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Stuart A C McDonald
- Clonal Dynamics in Epithelia Group; Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Quante
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), München, Germany
- Department of Medicine II, Universitaetsklinikum Freiburg, Freiburg, Germany
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
4
|
Maslyonkina KS, Konyukova AK, Alexeeva DY, Sinelnikov MY, Mikhaleva LM. Barrett's esophagus: The pathomorphological and molecular genetic keystones of neoplastic progression. Cancer Med 2022; 11:447-478. [PMID: 34870375 PMCID: PMC8729054 DOI: 10.1002/cam4.4447] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023] Open
Abstract
Barrett's esophagus is a widespread chronically progressing disease of heterogeneous nature. A life threatening complication of this condition is neoplastic transformation, which is often overlooked due to lack of standardized approaches in diagnosis, preventative measures and treatment. In this essay, we aim to stratify existing data to show specific associations between neoplastic transformation and the underlying processes which predate cancerous transition. We discuss pathomorphological, genetic, epigenetic, molecular and immunohistochemical methods related to neoplasia detection on the basis of Barrett's esophagus. Our review sheds light on pathways of such neoplastic progression in the distal esophagus, providing valuable insight into progression assessment, preventative targets and treatment modalities. Our results suggest that molecular, genetic and epigenetic alterations in the esophagus arise earlier than cancerous transformation, meaning the discussed targets can help form preventative strategies in at-risk patient groups.
Collapse
|
5
|
Wang H, DeFina SM, Bajpai M, Yan Q, Yang L, Zhou Z. DNA methylation markers in esophageal cancer: an emerging tool for cancer surveillance and treatment. Am J Cancer Res 2021; 11:5644-5658. [PMID: 34873485 PMCID: PMC8640794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/30/2021] [Indexed: 06/13/2023] Open
Abstract
Esophageal carcinoma (EC) is one of the most pervasive cancers in the world, with upwards of 500,000 new diagnoses, annually. Despite its prominence, advancements in the detection and treatment of EC have been marginal over the past 30 years and the survival rate continues to stay below 20%. This is due to the uncommonly heterogeneous presentation of EC which presents unprecedented challenges in improving patient survival and quality of care. However, distinct epigenetic alterations to the DNA methylome may provide an avenue to drastically improve the detection and treatment of EC. Specifically, the creation of novel biomarker panels that consist of EC-specific methylation markers have shown promise as a potential alternative to the more invasive, contemporary diagnostic methods. Additionally, growing insight into the biological and clinical properties of EC-specific methylation patterns have opened a window of opportunity for enhanced treatment; of growing interest is the application of "DNMT inhibitors" - a class of drugs which inhibit excessive methylation and have been shown to re-sensitize chemoresistant tumors. Here we provide a comprehensive review of the current advancements in EC DNA methylation to underscore a potential approach to its detection and treatment.
Collapse
Affiliation(s)
- He Wang
- Department of Pathology, Yale School of Medicine, Yale UniversityNew Haven, Connecticut, United States
| | - Samuel M DeFina
- Department of Pathology, Yale School of Medicine, Yale UniversityNew Haven, Connecticut, United States
| | - Manisha Bajpai
- Department of Medicine-Gastroenterology and Hepatology, Rutgers-Robert Wood Johnson Medical School, Rutgers The State University of New JerseyNew Brunswick, NJ, United States
| | - Qin Yan
- Department of Pathology, Yale School of Medicine, Yale UniversityNew Haven, Connecticut, United States
| | - Lei Yang
- Department of Pathology, Yale School of Medicine, Yale UniversityNew Haven, Connecticut, United States
| | - Zhongren Zhou
- Department of Pathology & Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, Rutgers The State University of New JerseyNew Brunswick, NJ, United States
| |
Collapse
|
6
|
Peng W, Tu G, Zhao Z, He B, Cai Q, Zhang P, Peng X, Shi S, Wang X. DNA methylome and transcriptome analysis established a model of four differentially methylated positions (DMPs) as a diagnostic marker in esophageal adenocarcinoma early detection. PeerJ 2021; 9:e11355. [PMID: 34012728 PMCID: PMC8109010 DOI: 10.7717/peerj.11355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/05/2021] [Indexed: 12/14/2022] Open
Abstract
Background Esophageal carcinogenesis involves in alterations of DNA methylation and gene transcription. This study profiled genomic DNA methylome vs. gene expression using transcriptome data on esophageal adenocarcinoma (EAC) tissues from the online databases in order to identify methylation biomarkers in EAC early diagnosis. Materials and Methods The DNA methylome and transcriptome data were downloaded from the UCSC Xena, Gene Expression Omnibus (GEO), and The Cancer Genome Atlas (TCGA) databases and then bioinformatically analyzed for the differentially methylated positions (DMPs) vs. gene expression between EAC and normal tissues. The highly methylated DMPs vs. reduced gene expression in EAC were selected and then stratified with those of the corresponding normal blood samples and other common human cancers to construct an EAC-specific diagnostic model. The usefulness of this model was further verified in other three GEO datasets of EAC tissues. Result A total of 841 DMPs were associated with expression of 320 genes, some of which were aberrantly methylated in EAC tissues. Further analysis showed that four (cg07589773, cg10474350, cg13011388 and cg15208375 mapped to gene IKZF1, HOXA7, EFS and TSHZ3, respectively) of these 841 DMPs could form and establish a diagnostic model after stratified them with the corresponding normal blood samples and other common human cancers. The data were further validated in other three GEO datasets on EAC tissues in early EAC diagnosis. Conclusion This study revealed a diagnostic model of four genes methylation to diagnose EAC early. Further study will confirm the usefulness of this model in a prospective EAC cases.
Collapse
Affiliation(s)
- Weilin Peng
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| | - Guangxu Tu
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| | - Zhenyu Zhao
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| | - Boxue He
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| | - Qidong Cai
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| | - Pengfei Zhang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| | - Xiong Peng
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| | - Shuai Shi
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| | - Xiang Wang
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Vercauteren Drubbel A, Pirard S, Kin S, Dassy B, Lefort A, Libert F, Nomura S, Beck B. Reactivation of the Hedgehog pathway in esophageal progenitors turns on an embryonic-like program to initiate columnar metaplasia. Cell Stem Cell 2021; 28:1411-1427.e7. [PMID: 33882290 DOI: 10.1016/j.stem.2021.03.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 12/18/2020] [Accepted: 03/24/2021] [Indexed: 01/17/2023]
Abstract
Columnar metaplasia of the esophagus is the main risk factor for esophageal adenocarcinoma. There is a lack of evidence to demonstrate that esophageal progenitors can be the source of columnar metaplasia. In this study, using transgenic mouse models, lineage tracing, single-cell RNA sequencing, and transcriptomic and epigenetic profiling, we found that the activation of the Hedgehog pathway in esophageal cells modifies their differentiation status in vivo. This process involves an initial step of dedifferentiation into embryonic-like esophageal progenitors. Moreover, a subset of these cells undergoes full squamous-to-columnar conversion and expresses selected intestinal markers. These modifications of cell fate are associated with remodeling of the chromatin and the appearance of Sox9. Using a conditional knockout mouse, we show that Sox9 is required for columnar conversion but not for the step of dedifferentiation. These results provide insight into the mechanisms by which esophageal cells might initiate columnar metaplasia.
Collapse
Affiliation(s)
| | - Sheleya Pirard
- IRIBHM, ULB/Faculty of Medicine, 808 route de Lennik, 1070 Brussels, Belgium
| | - Simon Kin
- IRIBHM, ULB/Faculty of Medicine, 808 route de Lennik, 1070 Brussels, Belgium
| | - Benjamin Dassy
- IRIBHM, ULB/Faculty of Medicine, 808 route de Lennik, 1070 Brussels, Belgium
| | - Anne Lefort
- IRIBHM, ULB/Faculty of Medicine, 808 route de Lennik, 1070 Brussels, Belgium
| | - Frédérick Libert
- IRIBHM, ULB/Faculty of Medicine, 808 route de Lennik, 1070 Brussels, Belgium
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Benjamin Beck
- IRIBHM, ULB/Faculty of Medicine, 808 route de Lennik, 1070 Brussels, Belgium; WELBIO/FNRS Principal Investigator at IRIBHM, ULB/Faculty of Medicine, 808 route de Lennik, 1070 Brussels, Belgium.
| |
Collapse
|
8
|
Dam AN, Klapman J. A narrative review of Barrett's esophagus in 2020, molecular and clinical update. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1107. [PMID: 33145326 PMCID: PMC7575938 DOI: 10.21037/atm-20-4406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Barrett’s esophagus (BE) is a condition resulting from an acquired metaplastic epithelial change in the esophagus in response to gastroesophageal reflux. BE is the only known precursor lesion to esophageal adenocarcinoma, and can progress from non-dysplastic BE (NDBE) to low grade dysplasia (LGD) and high grade dysplasia (HGD), and ultimately invasive carcinoma. Although the risk of developing esophageal adenocarcinoma (EAC) in NBDE is less than 0.5% per year, there has been a rising incidence of EAC in Western countries, which continue to drive efforts to optimize screening and surveillance methods. The current gold standard for diagnosis is esophagogastroduodenoscopy (EGD), and there has been significant interest in alternative, minimally invasive methods for screening which would be more readily accessible in the primary care setting. Surveillance endoscopy in 3–5 years is recommended for NDBE given the low progression to EAC. The mainstay of treatment for LGD and HGD is endoscopic eradication therapy (EET). Visible lesions are treated with endoscopic mucosal resection (EMR) or endoscopic submucosal dissection (ESD). Radiofrequency ablation (RFA) is considered first line therapy for flat dysplastic BE and cryotherapy has shown promising results as an alternate form of treatment for of dysplasia. The molecular progression of BE to EAC is a complex process involving multiple pathways involving genetic and epigenetic modifications. Genomic studies have further led to the understanding of the complex molecular landscape that occurs early and late in the disease process. Promising biomarker panels have been investigated to help with the diagnosis of BE as well as aid in the risk stratification of BE during surveillance. In addition, clinical prediction models have been developed to categorize BE patients in low, intermediate, and high risk for progression to HGD and EAC. Further clinical and translational research is needed to help refine markers and techniques in diagnosis, screening, and surveillance.
Collapse
Affiliation(s)
- Aamir N Dam
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jason Klapman
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
9
|
Lin R, Li C, Liu Z, Wu R, Lu J. Genome-wide DNA methylation profiling identifies epigenetic signatures of gastric cardiac intestinal metaplasia. J Transl Med 2020; 18:292. [PMID: 32736574 PMCID: PMC7393819 DOI: 10.1186/s12967-020-02453-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Measuring the DNA methylome may offer the opportunity to identify novel disease biomarkers and insights into disease mechanisms. Although aberrant DNA methylation has been investigated in many human cancers and precancerous lesions, the DNA methylation landscape of gastric cardiac intestinal metaplasia (IM) remains unknown. Therefore, we aimed to investigate the genome-wide DNA methylation landscape and to search for potential epigenetic biomarkers of gastric cardiac IM. METHODS Histopathologic profiling was performed on a total of 118 gastric cardiac biopsies from cancer-free individuals. Genome-wide DNA methylation analysis was performed on 11 gastric cardiac mucosal biopsies (IM = 7; normal = 4) using Illumina 850K microarrays. Transcriptional relevance of any candidate epigenetic biomarker was validated by qRT-PCR. RESULTS The detection rate of gastric cardiac IM was 23% (27/118) in cancer-free individuals. Genome-wide DNA methylation profiling showed a global decrease in methylation in IM compared with normal tissues (median methylation = 0.64 and 0.70 for gastric cardiac IM and normal tissues, respectively). Differential methylation analysis between gastric cardiac IM and normal tissues identified 38,237 differentially methylated probes (DMPs) with a majority of sites showing hypermethylation in IM compared with normal tissues (56.3% vs. 43.7%). Subsequent analysis revealed a significant enrichment of hypermethylated DMPs in promoter and CpG islands (p < 0.001 for both, Pearson χ2 test). For DMPs located in promoter CpG islands showing extreme hypermethylation, the candidate gene with the largest number of DMPs (n = 7) was mapped to HOXA5. Accordingly, mRNA expression of HOXA5 was significantly reduced in IM compared to normal tissue. CONCLUSIONS Our results suggest the implication of alterations in DNA methylation in gastric cardiac IM and highlight that HOXA5 hypermethylation may be a promising epigenetic biomarker, emphasizing the role of aberrant HOXA5 expression in the pathogenesis of gastric cardiac IM.
Collapse
Affiliation(s)
- Runhua Lin
- Department of Pathology, Shantou University Medical College, Shantou, 515041, China.
| | - Chenxi Li
- Department of Pathology, Shantou University Medical College, Shantou, 515041, China
| | - Zhaohui Liu
- The Second People's Hospital of Shenzhen/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Ruinuan Wu
- The Second People's Hospital of Shenzhen/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jianghong Lu
- Department of Pathology, Shantou University Medical College, Shantou, 515041, China
| |
Collapse
|
10
|
Caspa Gokulan R, Garcia-Buitrago MT, Zaika AI. From genetics to signaling pathways: molecular pathogenesis of esophageal adenocarcinoma. Biochim Biophys Acta Rev Cancer 2019; 1872:37-48. [PMID: 31152823 PMCID: PMC6692203 DOI: 10.1016/j.bbcan.2019.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
Esophageal adenocarcinoma (EAC) has one of the fastest rising incidence rates in the U.S. and many other Western countries. One of the unique risk factors for EAC is gastroesophageal reflux disease (GERD), a chronic digestive condition in which acidic contents from the stomach, frequently mixed with duodenal bile, enter the esophagus resulting in esophageal tissue injury. At the cellular level, progression to EAC is underlined by continuous DNA damage caused by reflux and chronic inflammatory factors that increase the mutation rate and promote genomic instability. Despite recent successes in cancer diagnostics and treatment, EAC remains a poorly treatable disease. Recent research has shed new light on molecular alterations underlying progression to EAC and revealed novel treatment options. This review focuses on the genetic and molecular studies of EAC. The molecular changes that occur during the transformation of normal Barrett's esophagus to esophageal adenocarcinoma are also discussed.
Collapse
Affiliation(s)
| | | | - Alexander I Zaika
- Department of Surgery, University of Miami, Miami, FL, United States of America; Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, United States of America.
| |
Collapse
|
11
|
Etiology, cancer stem cells and potential diagnostic biomarkers for esophageal cancer. Cancer Lett 2019; 458:21-28. [PMID: 31125642 DOI: 10.1016/j.canlet.2019.05.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/10/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022]
Abstract
Esophageal cancer (EC) has been a leading cause of cancer death worldwide in part due to late detection and lack of precision treatment. EC includes two major malignancies, esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). Recent studies reveal that ESCC and EAC have distinct cell of origin and contain cancer stem cells (also known as tumor initiating cells) expressing different cell surface markers. These biomarkers have potentially important values for both early detection and finding effective therapy. In this review we summarize the updated findings for cell of origin and provide an overview of cancer cell biomarkers that have been tested for ESCC and EAC. In addition, we also discuss recent progress in the study of molecular mechanisms leading to these malignancies.
Collapse
|
12
|
Yu M, Maden S, Stachler M, Kaz AM, Ayers J, Guo Y, Carter KT, Willbanks A, Heinzerling TJ, O’Leary RM, Xu X, Bass A, Chandar AK, Chak A, Elliot R, Willis JE, Markowitz SD, Grady WM. Subtypes of Barrett's oesophagus and oesophageal adenocarcinoma based on genome-wide methylation analysis. Gut 2019; 68:389-399. [PMID: 29884612 PMCID: PMC6565505 DOI: 10.1136/gutjnl-2017-314544] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 04/06/2018] [Accepted: 04/22/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To identify and characterise DNA methylation subtypes in oesophageal adenocarcinoma (EAC) and its precursor Barrett's oesophagus (BE). DESIGN We performed genome-wide DNA methylation profiling on samples of non-dysplastic BE from cancer-free patients (n=59), EAC (n=23), normal squamous oesophagus (n=33) and normal fundus (n=9), and identified methylation subtypes using a recursively partitioned mixture model. We assessed genomic alterations for 9 BE and 22 EAC samples with massively parallel sequencing of 243 EAC-associated genes, and we conducted integrative analyses with transcriptome data to identify epigenetically repressed genes. We also carried out in vitro experiments treating EAC cell lines with 5-Aza-2'-Deoxycytidine (5-Aza-dC), short hairpin RNA knockdown and anticancer therapies. RESULTS We identified and validated four methylation subtypes of EAC and BE. The high methylator subtype (HM) of EAC had the greatest number of activating events in ERBB2 (p<0.05, Student's t-test) and the highest global mutation load (p<0.05, Fisher's exact test). PTPN13 was silenced by aberrant methylation in the HM subtype preferentially and in 57% of EACs overall. In EAC cell lines, 5-Aza-dC treatment restored PTPN13 expression and significantly decreased its promoter methylation in HM cell lines (p<0.05, Welch's t-test). Inhibition of PTPN13 expression in the SK-GT-4 EAC cell line promoted proliferation, colony formation and migration, and increased phosphorylation in ERBB2/EGFR/Src kinase pathways. Finally, EAC cell lines showed subtype-specific responses to topotecan, SN-38 and palbociclib treatment. CONCLUSIONS We identified and characterised methylator subtypes in BE and EAC. We further demonstrated the biological and clinical relevance of EAC methylator subtypes, which may ultimately help guide clinical management of patients with EAC.
Collapse
Affiliation(s)
- Ming Yu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sean Maden
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Matthew Stachler
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Andrew M. Kaz
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA,Gastroenterology Section, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Jessica Ayers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yuna Guo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kelly T. Carter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Amber Willbanks
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Tai J. Heinzerling
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rachele M O’Leary
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Xinsen Xu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Adam Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA,Eli and Edythe L. Broad Institute, Cambridge, Massachusetts, USA
| | - Apoorva K. Chandar
- Division of Gastroenterology, University Hospitals Cleveland Medical Center, Cleveland, OH,Department of Medicine, Case Western Reserve University, Cleveland, OH; USA
| | - Amitabh Chak
- Division of Gastroenterology, University Hospitals Cleveland Medical Center, Cleveland, OH,Division of Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH; USA
| | - Robin Elliot
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH; USA
| | - Joseph E. Willis
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH; USA
| | - Sanford D. Markowitz
- Department of Medicine, Case Western Reserve University, Cleveland, OH; USA,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH; USA
| | - William M. Grady
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
13
|
Fujii M, Clevers H, Sato T. Modeling Human Digestive Diseases With CRISPR-Cas9-Modified Organoids. Gastroenterology 2019; 156:562-576. [PMID: 30476497 DOI: 10.1053/j.gastro.2018.11.048] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/10/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023]
Abstract
Insights into the stem cell niche have allowed researchers to cultivate adult tissue stem cells as organoids that display structural and phenotypic features of healthy and diseased epithelial tissues. Organoids derived from patients' tissues are used as models of disease and to test drugs. CRISPR-Cas9 technology can be used to genetically engineer organoids for studies of monogenic diseases and cancer. We review the derivation of organoids from human gastrointestinal tissues and how CRISPR-Cas9 technology can be used to study these organoids. We discuss burgeoning technologies that are broadening our understanding of diseases of the digestive system.
Collapse
Affiliation(s)
- Masayuki Fujii
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Hans Clevers
- Hubrecht Institute, University Medical Center Utrecht and Princess Maxima Center, Utrecht, The Netherlands
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
14
|
Talukdar FR, di Pietro M, Secrier M, Moehler M, Goepfert K, Lima SSC, Pinto LFR, Hendricks D, Parker MI, Herceg Z. Molecular landscape of esophageal cancer: implications for early detection and personalized therapy. Ann N Y Acad Sci 2018; 1434:342-359. [PMID: 29917250 DOI: 10.1111/nyas.13876] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/08/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022]
Abstract
Esophageal cancer (EC) is one of the most lethal cancers and a public health concern worldwide, owing to late diagnosis and lack of efficient treatment. Esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) are main histopathological subtypes of EC that show striking differences in geographical distribution, possibly due to differences in exposure to risk factors and lifestyles. ESCC and EAC are distinct diseases in terms of cell of origin, epidemiology, and molecular architecture of tumor cells. Past efforts aimed at translating potential molecular candidates into clinical practice proved to be challenging, underscoring the need for identifying novel candidates for early diagnosis and therapy of EC. Several major international efforts have brought about important advances in identifying molecular landscapes of ESCC and EAC toward understanding molecular mechanisms and critical molecular events driving the progression and pathological features of the disease. In our review, we summarize recent advances in the areas of genomics and epigenomics of ESCC and EAC, their mutational signatures and immunotherapy. We also discuss implications of recent advances in characterizing the genome and epigenome of EC for the discovery of diagnostic/prognostic biomarkers and development of new targets for personalized treatment and prevention.
Collapse
Affiliation(s)
- Fazlur Rahman Talukdar
- Section of Mechanisms of Carcinogenesis, International Agency for Research on Cancer (WHO), Lyon, France
| | | | - Maria Secrier
- Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Markus Moehler
- First Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Katrin Goepfert
- First Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | | | | | - Denver Hendricks
- Division of Medical Biochemistry & Structural Biology, University of Cape Town, Cape Town, South Africa
| | - Mohamed Iqbal Parker
- Division of Medical Biochemistry & Structural Biology, University of Cape Town, Cape Town, South Africa
| | - Zdenko Herceg
- Section of Mechanisms of Carcinogenesis, International Agency for Research on Cancer (WHO), Lyon, France
| |
Collapse
|
15
|
Chettouh H, Mowforth O, Galeano-Dalmau N, Bezawada N, Ross-Innes C, MacRae S, Debiram-Beecham I, O’Donovan M, Fitzgerald RC. Methylation panel is a diagnostic biomarker for Barrett's oesophagus in endoscopic biopsies and non-endoscopic cytology specimens. Gut 2018; 67:1942-1949. [PMID: 29084829 PMCID: PMC6176521 DOI: 10.1136/gutjnl-2017-314026] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Barrett's oesophagus is a premalignant condition that occurs in the context of gastro-oesophageal reflux. However, most Barrett's cases are undiagnosed because of reliance on endoscopy. We have developed a non-endoscopic tool: the Cytosponge, which when combined with trefoil factor 3 immunohistochemistry, can diagnose Barrett's oesophagus. We investigated whether a quantitative methylation test that is not reliant on histopathological analysis could be used to diagnose Barrett's oesophagus. DESIGN Differentially methylated genes between Barrett's and normal squamous oesophageal biopsies were identified from whole methylome data and confirmed using MethyLight PCR in biopsy samples of squamous oesophagus, gastric cardia and Barrett's oesophagus. Selected genes were then tested on Cytosponge BEST2 trial samples comprising a pilot cohort (n=20 cases, n=10 controls) and a validation cohort (n=149 cases, n=129 controls). RESULTS Eighteen genes were differentially methylated in patients with Barrett'soesophagus compared with squamous controls. Hypermethylation of TFPI2, TWIST1, ZNF345 and ZNF569 was confirmed in Barrett's biopsies compared with biopsies from squamous oesophagus and gastric cardia (p<0.05). When tested in Cytosponge samples, these four genes were hypermethylated in patients with Barrett's oesophagus compared with patients with reflux symptoms (p<0.001). The optimum biomarker to diagnose Barrett's oesophagus was TFPI2 with a sensitivity and specificity of 82.2% and 95.7%, respectively. CONCLUSION TFPI2, TWIST1, ZNF345 and ZNF569 methylation have promise as diagnostic biomarkers for Barrett's oesophagus when used in combination with a simple and cost effective non-endoscopic cell collection device.
Collapse
Affiliation(s)
- Hamza Chettouh
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Oliver Mowforth
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Núria Galeano-Dalmau
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Navya Bezawada
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Caryn Ross-Innes
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Shona MacRae
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Irene Debiram-Beecham
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Maria O’Donovan
- Department of Histopathology, Addenbrooke’s Hospital, Cambridge, UK
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
Abstract
The cost-effectiveness of screening and surveillance for Barrett's esophagus continues to evolve as the incidence of esophageal adenocarcinoma increases, biomarkers enhance the identification of individuals at highest risk for developing cancer, and endoscopic eradication of Barrett's esophagus improves. Screening to detect Barrett's esophagus may be cost-effective in selected high-risk groups based on age, race, sex and other factors such as symptoms of heartburn. Currently, endoscopic eradication therapy for Barrett's esophagus and high-grade dysplasia is a cost-effective intervention, while endoscopic therapy for non-dysplastic Barrett's esophagus is not a cost-effective strategy. As diagnosis of low-grade dysplasia improves, endoscopic eradication therapy may also prove to be a cost-effective intervention.
Collapse
Affiliation(s)
- John M Inadomi
- Division of Gastroenterology, Department of Medicine, University of Washington School of Medicine, 1959 NE Pacific Street, Box 356424, Seattle, WA, 98195, USA.
| | - Nina Saxena
- Division of Gastroenterology, Department of Medicine, University of Washington School of Medicine, 1959 NE Pacific Street, Box 356424, Seattle, WA, 98195, USA
| |
Collapse
|
17
|
Abstract
Esophageal adenocarcinoma (EAC) develops from Barrett's esophagus (BE), a condition where the normal squamous epithelia is replaced by specialized intestinal metaplasia in response to chronic gastroesophageal acid reflux. In a minority of individuals, BE can progress to low- and high-grade dysplasia and eventually to intra-mucosal and then invasive carcinoma. BE provides researchers with a unique model to characterize the process by which a carcinoma arises from its precursor lesion. Molecular studies of BE have demonstrated that it is not simply a metaplastic tissue, but rather it harbors frequent alterations that are also present in dysplastic BE and in EAC. Both BE and EAC are characterized by loss of heterozygosity, aneuploidy, specific genetic mutations, and clonal diversity. Epigenetic abnormalities, primary alterations in DNA methylation, are also frequently seen in BE and EAC. Candidate gene and array-based approaches have demonstrated that numerous tumor suppressor genes exhibit aberrant promoter methylation, and some of these altered genes are associated with the neoplastic progression of BE. It has also been shown that the BE and EAC epigenomes are characterized by hypomethylation of intragenic and non-coding regions Recent studies have also provided new insight into the evolutionary forces underlying the molecular alterations seen in BE and EAC and into the molecular pathogenesis of EAC.
Collapse
Affiliation(s)
- William M. Grady
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA,University of Washington School of Medicine, Department of Internal Medicine, Seattle, WA
| | - Ming Yu
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA
| |
Collapse
|
18
|
Li J, Ye D, Wang L, Peng Y, Li Q, Deng H, Zhou C. Role of MLH1 methylation in esophageal cancer carcinogenesis and its clinical significance. Onco Targets Ther 2018; 11:651-663. [PMID: 29440913 PMCID: PMC5798571 DOI: 10.2147/ott.s154999] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The mutL homolog-1 (MLH1) is a DNA mismatch repair gene and has been reported to be frequently methylated in numerous cancers. However, the association between MLH1 methylation and esophageal cancer (EC), as well as its clinical significance, remains unclear. Hence, we conducted a systematic meta-analysis based on 19 articles (including 1384 ECs, 345 premalignant lesions, and 1244 healthy controls). Our analysis revealed that the frequency of MLH1 methylation was significantly elevated during EC carcinogenesis. In addition, we observed that MLH1 promoter methylation was associated with age (odds ratio [OR]=1.79; 95% CI =1.20-2.66), advanced tumor grade (OR=3.7; 95% CI =2.37-5.77), lymph node metastasis (OR=2.65; 95% CI =1.81-3.88), distant metastasis (OR=7.60; 95% CI =1.23-47.19), advanced clinical stage (OR=4.46; 95% CI =2.88-6.91), and poor prognosis in EC patients (hazard ratio =1.64, 95% CI =1.00-2.69). The pooled sensitivity, specificity, and area under the curve of MLH1 methylation in EC patients versus healthy individuals were 0.15, 0.99, and 0.77, respectively. Our findings indicate that MLH1 methylation is involved in the carcinogenesis, progression, and metastasis of EC. Moreover, methylated MLH1 could be a potential diagnostic and prognostic biomarker for EC.
Collapse
Affiliation(s)
- Jinyun Li
- Department of Oncology and Hematology, Affiliated Hospital of Ningbo University
| | - Dong Ye
- Department of Otorhinolaryngology – Head and Neck Surgery
| | - Lei Wang
- Department of General Surgery, Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yingying Peng
- Department of General Surgery, Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qun Li
- Department of Otorhinolaryngology – Head and Neck Surgery
| | - Hongxia Deng
- Department of Otorhinolaryngology – Head and Neck Surgery
| | | |
Collapse
|
19
|
Gaur P, Hunt CR, Pandita TK. Emerging therapeutic targets in esophageal adenocarcinoma. Oncotarget 2018; 7:48644-48655. [PMID: 27102294 PMCID: PMC5217045 DOI: 10.18632/oncotarget.8777] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/10/2016] [Indexed: 12/18/2022] Open
Abstract
The incidence of gastro-esophageal disease and associated rate of esophageal adenocarcinoma (EAC) is rising at an exponential rate in the United States. However, research targeting EAC is lagging behind, and much research is needed in the field to identify ways to diagnose EAC early as well as to improve the rate of pathologic complete response (pCR) to systemic therapies. Esophagectomy with subsequent reconstruction is known to be a morbid procedure that significantly impacts a patient's quality of life. If indeed the pCR rate of patients can be improved and those patients destined to be pCR can be identified ahead of time, they may be able to avoid this life-altering procedure. While cancer-specific biological pathways have been thoroughly investigated in other solid malignancies, much remains unexplored in EAC. In this review, we will highlight some of the latest research in the field in regards with EAC, along with new therapeutic targets that are currently being explored. After reviewing conventional treatment and current changes in medical therapy for EAC, we will focus on unchartered grounds such as cancer stem cells, genetics and epigenetics, immunotherapy, and chemoradio-resistant pathways as we simultaneously propose some investigational possibilities that could be applicable to EAC.
Collapse
Affiliation(s)
- Puja Gaur
- Department of General Surgery, Division of Thoracic Surgery, The Houston Methodist Research Institute, Houston, TX, USA
| | - Clayton R Hunt
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX, USA
| | - Tej K Pandita
- Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
20
|
Abstract
Metaplasia is the replacement of one differentiated somatic cell type with another differentiated somatic cell type in the same tissue. Typically, metaplasia is triggered by environmental stimuli, which may act in concert with the deleterious effects of microorganisms and inflammation. The cell of origin for intestinal metaplasia in the oesophagus and stomach and for pancreatic acinar-ductal metaplasia has been posited through genetic mouse models and lineage tracing but has not been identified in other types of metaplasia, such as squamous metaplasia. A hallmark of metaplasia is a change in cellular identity, and this process can be regulated by transcription factors that initiate and/or maintain cellular identity, perhaps in concert with epigenetic reprogramming. Universally, metaplasia is a precursor to low-grade dysplasia, which can culminate in high-grade dysplasia and carcinoma. Improved clinical screening for and surveillance of metaplasia might lead to better prevention or early detection of dysplasia and cancer.
Collapse
Affiliation(s)
- Veronique Giroux
- University of Pennsylvania Perelman School of Medicine, 951 BRB, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| | - Anil K Rustgi
- University of Pennsylvania Perelman School of Medicine, 951 BRB, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
21
|
Lin B, Xie F, Xiao Z, Hong X, Tian L, Liu K. Basal progenitor cells bridge the development, malignant cancers, and multiple diseases of esophagus. J Cell Physiol 2017; 233:3855-3866. [PMID: 28777465 DOI: 10.1002/jcp.26136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/03/2017] [Indexed: 12/19/2022]
Abstract
The esophagus is a pivotal organ originating from anterior foregut that links the mouth and stomach. Moreover, its development involves precise regulation of multiple signal molecules and signal transduction pathways. After abnormal regulation of these molecules in the basal cells of the esophagus occurs, multiple diseases, including esophageal atresia with or without tracheoesophageal fistula, Barrett esophagus, gastroesophageal reflux, and eosinophilic esophagitis, will take place as a result. Furthermore, expression changes of signal molecules or signal pathways in basal cells and the microenvironment around basal cells both can initiate the switch of malignant transformation. In this review, we highlight the molecular events underlying the transition of normal development to multiple esophageal diseases. Additionally, the animal models of esophageal development and related diseases, challenges, and strategies are extensively discussed.
Collapse
Affiliation(s)
- Baoshun Lin
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian, P. R. China
| | - Fuan Xie
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian, P. R. China
| | - Zhangwu Xiao
- Emergency Department of the 476 Hospital, Fuzhou General Hospital, PLA, Fuzhou, Fujian, P. R. China
| | - Xiaoqian Hong
- Dong fang Hospital, Xiamen University, Fuzhou, Fujian, P. R. China
| | - Liming Tian
- Dong fang Hospital, Xiamen University, Fuzhou, Fujian, P. R. China
| | - Kuancan Liu
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian, P. R. China.,Dong fang Hospital, Xiamen University, Fuzhou, Fujian, P. R. China.,Department of Medicine, Columbia University Medical Center, New York, New York
| |
Collapse
|
22
|
Saxena N, Inadomi JM. Effectiveness and Cost-Effectiveness of Endoscopic Screening and Surveillance. Gastrointest Endosc Clin N Am 2017; 27:397-421. [PMID: 28577764 DOI: 10.1016/j.giec.2017.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Guidelines for the screening and surveillance of Barrett's esophagus continue to evolve as the incidence of esophageal adenocarcinoma increases, identification of individuals at highest risk for cancer improves, and management of dysplasia evolves. This article reviews related studies and economic analyses. Advances in diagnosis offer promising strategies to help focus screening efforts on those individuals who are most likely to develop esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Nina Saxena
- Division of Gastroenterology, Department of Medicine, University of Washington School of Medicine, 1959 Northeast Pacific Street, Box 356424, Seattle, WA 98195, USA
| | - John M Inadomi
- Division of Gastroenterology, Department of Medicine, University of Washington School of Medicine, 1959 Northeast Pacific Street, Box 356424, Seattle, WA 98195, USA.
| |
Collapse
|
23
|
Smith E, Palethorpe HM, Hayden AL, Young JP, Underwood TJ, Drew PA. Fibroblasts derived from oesophageal adenocarcinoma differ in DNA methylation profile from normal oesophageal fibroblasts. Sci Rep 2017; 7:3368. [PMID: 28611465 PMCID: PMC5469830 DOI: 10.1038/s41598-017-03501-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 05/02/2017] [Indexed: 12/21/2022] Open
Abstract
Oesophageal adenocarcinoma (OAC) is increasing in incidence and has a poor prognosis. Tumour derived fibroblasts (TDFs) differ functionally from normal fibroblasts (NDFs), and play a pivotal role in cancer. Many of the differences persist through subculture. We measured the DNA methylation profiles of 10 TDFs from OAC with 12 NDF from normal oesophageal mucosa using Infinium HumanMethylation450 Beadchips and found they differed in multidimensional scaling analysis. We identified 4,856 differentially methylated CpGs (DMCs, adjusted p < 0.01 and absolute difference in average β-value > 0.15), of which 3,243 (66.8%) were hypomethylated in TDFs compared to NDFs. Hypermethylated DMCs were enriched at transcription start sites (TSSs) and in CpG islands, and depleted in transcriptional enhancers. Gene ontology analysis of genes with DMCs at TSSs revealed an enrichment of genes involved in development, morphogenesis, migration, adhesion, regulation of processes and response to stimuli. Alpha-smooth muscle actin (α-SMA) is a marker of activated fibroblasts and a poor prognostic indicator in OAC. Hypomethylated DMCs were observed at the TSS of transcript variant 2 of α-SMA, which correlated with an increase in α-SMA protein expression. These data suggest that DNA methylation may contribute to the maintenance of the TDF phenotype.
Collapse
Affiliation(s)
- Eric Smith
- Discipline of Surgical Specialities, Adelaide Medical School, Faculty of Health Sciences, The University of Adelaide, South Australia, 5000, Australia.
- Department of Haematology and Oncology, The Queen Elizabeth Hospital, Woodville, South Australia, 5011, Australia.
| | - Helen M Palethorpe
- Discipline of Surgical Specialities, Adelaide Medical School, Faculty of Health Sciences, The University of Adelaide, South Australia, 5000, Australia
| | - Annette L Hayden
- Cancer Sciences Unit, Somers Cancer Research Building, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - Joanne P Young
- Discipline of Surgical Specialities, Adelaide Medical School, Faculty of Health Sciences, The University of Adelaide, South Australia, 5000, Australia
- Department of Haematology and Oncology, The Queen Elizabeth Hospital, Woodville, South Australia, 5011, Australia
| | - Timothy J Underwood
- Cancer Sciences Unit, Somers Cancer Research Building, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - Paul A Drew
- Discipline of Surgical Specialities, Adelaide Medical School, Faculty of Health Sciences, The University of Adelaide, South Australia, 5000, Australia
- School of Nursing and Midwifery, Flinders University, PO Box 2100, Adelaide, South Australia, 5001, Australia
| |
Collapse
|
24
|
Barnicle A, Seoighe C, Greally JM, Golden A, Egan LJ. Inflammation-associated DNA methylation patterns in epithelium of ulcerative colitis. Epigenetics 2017; 12:591-606. [PMID: 28557546 DOI: 10.1080/15592294.2017.1334023] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aberrant DNA methylation patterns have been reported in inflamed tissues and may play a role in disease. We studied DNA methylation and gene expression profiles of purified intestinal epithelial cells from ulcerative colitis patients, comparing inflamed and non-inflamed areas of the colon. We identified 577 differentially methylated sites (false discovery rate <0.2) mapping to 210 genes. From gene expression data from the same epithelial cells, we identified 62 differentially expressed genes with increased expression in the presence of inflammation at prostate cancer susceptibility genes PRAC1 and PRAC2. Four genes showed inverse correlation between methylation and gene expression; ROR1, GXYLT2, FOXA2, and, notably, RARB, a gene previously identified as a tumor suppressor in colorectal adenocarcinoma as well as breast, lung and prostate cancer. We highlight targeted and specific patterns of DNA methylation and gene expression in epithelial cells from inflamed colon, while challenging the importance of epithelial cells in the pathogenesis of chronic inflammation.
Collapse
Affiliation(s)
- Alan Barnicle
- a Clinical Pharmacology, School of Medicine, National University of Ireland , Galway , Ireland.,b School of Mathematics, Statistics and Applied Mathematics, National University of Ireland , Galway , Ireland
| | - Cathal Seoighe
- b School of Mathematics, Statistics and Applied Mathematics, National University of Ireland , Galway , Ireland
| | - John M Greally
- c Center of Epigenomics and Department of Genetics (Division of Computational Genetics) , Albert Einstein College of Medicine, Morris Park Avenue , Bronx , NY , USA
| | - Aaron Golden
- b School of Mathematics, Statistics and Applied Mathematics, National University of Ireland , Galway , Ireland.,c Center of Epigenomics and Department of Genetics (Division of Computational Genetics) , Albert Einstein College of Medicine, Morris Park Avenue , Bronx , NY , USA.,d Department of Mathematical Sciences , Yeshiva University , 2495 Amsterdam Avenue, New York , NY , USA
| | - Laurence J Egan
- a Clinical Pharmacology, School of Medicine, National University of Ireland , Galway , Ireland
| |
Collapse
|
25
|
Sánchez-Vega F, Gotea V, Chen YC, Elnitski L. CpG island methylator phenotype in adenocarcinomas from the digestive tract: Methods, conclusions, and controversies. World J Gastrointest Oncol 2017; 9:105-120. [PMID: 28344746 PMCID: PMC5348626 DOI: 10.4251/wjgo.v9.i3.105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 10/01/2016] [Accepted: 01/03/2017] [Indexed: 02/05/2023] Open
Abstract
Over the last two decades, cancer-related alterations in DNA methylation that regulate transcription have been reported for a variety of tumors of the gastrointestinal tract. Due to its relevance for translational research, great emphasis has been placed on the analysis and molecular characterization of the CpG island methylator phenotype (CIMP), defined as widespread hypermethylation of CpG islands in clinically distinct subsets of cancer patients. Here, we present an overview of previous work in this field and also explore some open questions using cross-platform data for esophageal, gastric, and colorectal adenocarcinomas from The Cancer Genome Atlas. We provide a data-driven, pan-gastrointestinal stratification of individual samples based on CIMP status and we investigate correlations with oncogenic alterations, including somatic mutations and epigenetic silencing of tumor suppressor genes. Besides known events in CIMP such as BRAF V600E mutation, CDKN2A silencing or MLH1 inactivation, we discuss the potential role of emerging actors such as Wnt pathway deregulation through truncating mutations in RNF43 and epigenetic silencing of WIF1. Our results highlight the existence of molecular similarities that are superimposed over a larger backbone of tissue-specific features and can be exploited to reduce heterogeneity of response in clinical trials.
Collapse
|
26
|
Kaz AM, Wong CJ, Varadan V, Willis JE, Chak A, Grady WM. Global DNA methylation patterns in Barrett's esophagus, dysplastic Barrett's, and esophageal adenocarcinoma are associated with BMI, gender, and tobacco use. Clin Epigenetics 2016; 8:111. [PMID: 27795744 PMCID: PMC5082363 DOI: 10.1186/s13148-016-0273-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/29/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The risk of developing Barrett's esophagus (BE) and/or esophageal adenocarcinoma (EAC) is associated with specific demographic and behavioral factors, including gender, obesity/elevated body mass index (BMI), and tobacco use. Alterations in DNA methylation, an epigenetic modification that can affect gene expression and that can be influenced by environmental factors, is frequently present in both BE and EAC and is believed to play a role in the formation of BE and its progression to EAC. It is currently unknown whether obesity or tobacco smoking influences the risk of developing BE/EAC via the induction of alterations in DNA methylation. To investigate this possibility, we assessed the genome-wide methylation status of 81 esophageal tissues, including BE, dysplastic BE, and EAC epithelia using HumanMethylation450 BeadChips (Illumina). RESULTS We found numerous differentially methylated loci in the esophagus tissues when comparing males to females, obese to lean individuals, and smokers to nonsmokers. Differences in DNA methylation between these groups were seen in a variety of functional genomic regions and both within and outside of CpG islands. Several cancer-related pathways were found to have differentially methylated genes between these comparison groups. CONCLUSIONS Our findings suggest obesity and tobacco smoking may influence DNA methylation in the esophagus and raise the possibility that these risk factors affect the development of BE, dysplastic BE, and EAC through influencing the epigenetic status of specific loci that have a biologically plausible role in cancer formation.
Collapse
Affiliation(s)
- Andrew M. Kaz
- Gastroenterology Section, VA Puget Sound Health Care System, Seattle, WA 98108 USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195 USA
| | - Chao-Jen Wong
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 USA
| | - Vinay Varadan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106 USA
| | - Joseph E. Willis
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
| | - Amitabh Chak
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106 USA
- Division of Gastroenterology, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
| | - William M. Grady
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195 USA
| |
Collapse
|
27
|
DNA methylation profiling of esophageal adenocarcinoma using Methylation Ligation-dependent Macroarray (MLM). Biochem Biophys Res Commun 2016; 479:231-237. [PMID: 27634218 DOI: 10.1016/j.bbrc.2016.09.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/11/2016] [Indexed: 02/08/2023]
Abstract
Most types of cancer cells are characterized by aberrant methylation of promoter genes. In this study, we described a rapid, reproducible, and relatively inexpensive approach allowing the detection of multiple human methylated promoter genes from many tissue samples, without the need of bisulfite conversion. The Methylation Ligation-dependent Macroarray (MLM), an array-based analysis, was designed in order to measure methylation levels of 58 genes previously described as putative biomarkers of cancer. The performance of the design was proven by screening the methylation profile of DNA from esophageal cell lines, as well as microdissected formalin-fixed and paraffin-embedded (FFPE) tissues from esophageal adenocarcinoma (EAC). Using the MLM approach, we identified 32 (55%) hypermethylated promoters in EAC, and not or rarely methylated in normal tissues. Among them, 21promoters were found aberrantly methylated in more than half of tumors. Moreover, seven of them (ADAMTS18, APC, DKK2, FOXL2, GPX3, TIMP3 and WIF1) were found aberrantly methylated in all or almost all the tumor samples, suggesting an important role for these genes in EAC. In addition, dysregulation of the Wnt pathway with hypermethylation of several Wnt antagonist genes was frequently observed. MLM revealed a homogeneous pattern of methylation for a majority of tumors which were associated with an advanced stage at presentation and a poor prognosis. Interestingly, the few tumors presenting less methylation changes had a lower pathological stage. In conclusion, this study demonstrated the feasibility and accuracy of MLM for DNA methylation profiling of FFPE tissue samples.
Collapse
|
28
|
Krause L, Nones K, Loffler KA, Nancarrow D, Oey H, Tang YH, Wayte NJ, Patch AM, Patel K, Brosda S, Manning S, Lampe G, Clouston A, Thomas J, Stoye J, Hussey DJ, Watson DI, Lord RV, Phillips WA, Gotley D, Smithers BM, Whiteman DC, Hayward NK, Grimmond SM, Waddell N, Barbour AP. Identification of the CIMP-like subtype and aberrant methylation of members of the chromosomal segregation and spindle assembly pathways in esophageal adenocarcinoma. Carcinogenesis 2016; 37:356-65. [PMID: 26905591 PMCID: PMC4806711 DOI: 10.1093/carcin/bgw018] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/21/2015] [Accepted: 01/13/2016] [Indexed: 12/11/2022] Open
Abstract
The incidence of esophageal adenocarcinoma (EAC) has risen significantly over recent decades. Although survival has improved, cure rates remain poor, with <20% of patients surviving 5 years. This is the first study to explore methylome, transcriptome and ENCODE data to characterize the role of methylation in EAC. We investigate the genome-wide methylation profile of 250 samples including 125 EAC, 19 Barrett's esophagus (BE), 85 squamous esophagus and 21 normal stomach. Transcriptome data of 70 samples (48 EAC, 4 BE and 18 squamous esophagus) were used to identify changes in methylation associated with gene expression. BE and EAC showed similar methylation profiles, which differed from squamous tissue. Hypermethylated sites in EAC and BE were mainly located in CpG-rich promoters. A total of 18575 CpG sites associated with 5538 genes were differentially methylated, 63% of these genes showed significant correlation between methylation and mRNA expression levels. Pathways involved in tumorigenesis including cell adhesion, TGF and WNT signaling showed enrichment for genes aberrantly methylated. Genes involved in chromosomal segregation and spindle formation were aberrantly methylated. Given the recent evidence that chromothripsis may be a driver mechanism in EAC, the role of epigenetic perturbation of these pathways should be further investigated. The methylation profiles revealed two EAC subtypes, one associated with widespread CpG island hypermethylation overlapping H3K27me3 marks and binding sites of the Polycomb proteins. These subtypes were supported by an independent set of 89 esophageal cancer samples. The most hypermethylated tumors showed worse patient survival.
Collapse
Affiliation(s)
- Lutz Krause
- Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Brisbane, Queensland 4102, Australia, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia, Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia
| | - Kelly A Loffler
- Surgical Oncology Group, School of Medicine, The University of Queensland, Translational Research Institute at the Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia
| | - Derek Nancarrow
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia
| | - Harald Oey
- Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Brisbane, Queensland 4102, Australia
| | - Yue Hang Tang
- Surgical Oncology Group, School of Medicine, The University of Queensland, Translational Research Institute at the Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia
| | - Nicola J Wayte
- Surgical Oncology Group, School of Medicine, The University of Queensland, Translational Research Institute at the Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia
| | - Ann Marie Patch
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia, Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia
| | - Kalpana Patel
- Surgical Oncology Group, School of Medicine, The University of Queensland, Translational Research Institute at the Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia, Mater Medical Research Institute, Level 3 Aubigny Place, Raymond Terrace, Brisbane, Queensland 4101, Australia
| | - Sandra Brosda
- Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Brisbane, Queensland 4102, Australia, Faculty of Technology and Center for Biotechnology (CeBiTec), Bielefeld University, 33615 Bielefeld, Germany
| | - Suzanne Manning
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia
| | - Guy Lampe
- Department of Anatomical Pathology, Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia
| | - Andrew Clouston
- School of Medicine, Centre for Liver Disease Research, The University of Queensland, 1/49 Butterfield Street, Herston, Brisbane, Queensland 4006, Australia
| | - Janine Thomas
- Upper GI Research Unit, Division of Surgery, Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia
| | - Jens Stoye
- Faculty of Technology and Center for Biotechnology (CeBiTec), Bielefeld University, 33615 Bielefeld, Germany
| | - Damian J Hussey
- Department of Surgery, Flinders Medical Centre, Flinders University, Bedford Park, South Australia 5042, Australia
| | - David I Watson
- Department of Surgery, Flinders Medical Centre, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Reginald V Lord
- St. Vincent's Centre for Applied Medical Research, Sydney, New South Wales 2011, Australia, University of Notre Dame, Sydney, New South Wales 2011, Australia, University of New South Wales, Sydney, New South Wales 2011, Australia
| | - Wayne A Phillips
- Peter MacCallum Cancer Centre, East Melbourne, Victoria 3002, Australia, Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - David Gotley
- Department of Surgery, School of Medicine, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia and
| | - B Mark Smithers
- Department of Surgery, School of Medicine, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia and
| | - David C Whiteman
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia
| | - Nicholas K Hayward
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia
| | - Sean M Grimmond
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia, Wolfson Wohl Cancer Research Centre, Institute for Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow Scotland G61 1BD, UK
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, Queensland 4006, Australia, Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia,
| | - Andrew P Barbour
- Surgical Oncology Group, School of Medicine, The University of Queensland, Translational Research Institute at the Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia, Department of Surgery, School of Medicine, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland 4102, Australia and
| |
Collapse
|
29
|
Barnicle A, Seoighe C, Golden A, Greally JM, Egan LJ. Differential DNA methylation patterns of homeobox genes in proximal and distal colon epithelial cells. Physiol Genomics 2016; 48:257-73. [PMID: 26812987 DOI: 10.1152/physiolgenomics.00046.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 01/13/2016] [Indexed: 12/24/2022] Open
Abstract
Region and cell-type specific differences in the molecular make up of colon epithelial cells have been reported. Those differences may underlie the region-specific characteristics of common colon epithelial diseases such as colorectal cancer and inflammatory bowel disease. DNA methylation is a cell-type specific epigenetic mark, essential for transcriptional regulation, silencing of repetitive DNA and genomic imprinting. Little is known about any region-specific variations in methylation patterns in human colon epithelial cells. Using purified epithelial cells and whole biopsies (n= 19) from human subjects, we generated epigenome-wide DNA methylation data (using the HELP-tagging assay), comparing the methylation signatures of the proximal and distal colon. We identified a total of 125 differentially methylated sites (DMS) mapping to transcription start sites of protein-coding genes, most notably several members of the homeobox (HOX) family of genes. Patterns of differential methylation were validated with MassArray EpiTYPER. We also examined DNA methylation in whole biopsies, applying a computational technique to deconvolve variation in methylation within cell types and variation in cell-type composition across biopsies. Including inferred epithelial proportions as a covariate in differential methylation analysis applied to the whole biopsies resulted in greater overlap with the results obtained from purified epithelial cells compared with when the covariate was not included. Results obtained from both approaches highlight region-specific methylation patterns of HOX genes in colonic epithelium. Regional variation in methylation patterns has implications for the study of diseases that exhibit regional expression patterns in the human colon, such as inflammatory bowel disease and colorectal cancer.
Collapse
Affiliation(s)
- Alan Barnicle
- Clinical Pharmacology, School of Medicine, National University of Ireland, Galway, Ireland; School of Mathematics, Statistics and Applied Mathematics, National University of Ireland, Galway, Ireland; and
| | - Cathal Seoighe
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland, Galway, Ireland; and
| | - Aaron Golden
- Center of Epigenomics and Department of Genetics (Division of Computational Genetics), Albert Einstein College of Medicine, Bronx, New York
| | - John M Greally
- Center of Epigenomics and Department of Genetics (Division of Computational Genetics), Albert Einstein College of Medicine, Bronx, New York
| | - Laurence J Egan
- Clinical Pharmacology, School of Medicine, National University of Ireland, Galway, Ireland;
| |
Collapse
|
30
|
Kit O, Vodolazhskiy D, Kolesnikov E, Timoshkina N. Epigenetic markers of esophageal cancer: DNA methylation. ACTA ACUST UNITED AC 2016; 62:520-526. [DOI: 10.18097/pbmc20166205520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Adenocarcinoma and squamous cell carcinoma are the most common types of esophageal cancer with a constant tendency to increase the incidence of growth on the background of the high mortality, which makes particularly the development of new biomarkers that complement and improve the early diagnosis of this disease. Despite the impressive number of studies in routine clinical practice is used only marker of esophageal cancer – ERBB2/HER2 status. This review summarizes data on the identified epigenetic markers of the aberrant methylation of the genome, which may be useful for early detection of esophageal cancer, prognosis estimation and / or prediction of response to treatment. The development of new high-tech genome-wide screening, such as beadarray and immunoprecipitation sequencing method used for the wideband genotyping, but for the analysis of transcriptome and metilom, provides a comprehensive picture of genetic and epigenetic changes during tumorigenesis. Note the need to verify the most biomarkers on large representative samples for the development of valid diagnostic panels, suitable for large-scale screening of risk groups.
Collapse
Affiliation(s)
- O.I. Kit
- Rostov Cancer Research Institute, Rostov-on-Don, Russia
| | | | | | | |
Collapse
|
31
|
Yu M, O'Leary RM, Kaz AM, Morris SM, Carter KT, Chak A, Chandar A, Willis JE, Moinova HR, Markowitz SD, Brenner DE, Anandabapasathy S, Westerhoff M, Wong CJ, Shaheen NJ, Chen Y, Barnholtz-Sloan JS, Grady WM. Methylated B3GAT2 and ZNF793 Are Potential Detection Biomarkers for Barrett's Esophagus. Cancer Epidemiol Biomarkers Prev 2015; 24:1890-7. [PMID: 26545406 DOI: 10.1158/1055-9965.epi-15-0370] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 09/30/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Barrett's esophagus (BE) is a preneoplastic condition in which normal esophageal squamous epithelium (SQ) is replaced by specialized intestinal metaplasia. It is the presumed precursor for esophageal adenocarcinoma (EAC) as well as the strongest risk factor for this cancer. Unfortunately, many patients with BE go undiagnosed under the current BE screening guidelines. The development of noninvasive and accurate BE detection assays could potentially identify many of these undiagnosed BE patients. METHODS DNA methylation is a common epigenetic alteration in BE. Therefore, we conducted a genome-wide methylation screen to identify potential BE biomarkers. Samples from SQ (N = 12), stomach (N = 28), and BE (N = 29) were analyzed and methylation levels at over 485,000 CpG sites were compared. Pyrosequencing assays were used to validate the results and MethyLight assays were developed to detect the methylated alleles in endoscopic brushings. RESULTS We discovered two genes, B3GAT2 and ZNF793, that are aberrantly methylated in BE. Clinical validation studies confirmed B3GAT2 and ZNF793 methylation levels were significantly higher in BE samples (median = 32.5% and 33.1%, respectively) than in control tissues (median = 2.29% and 2.52%, respectively; P < 0.0001 for both genes). Furthermore, gene-specific MethyLight assays could accurately detect BE (P < 0.0001 for both) in endoscopic brushing samples. CONCLUSION B3GAT2 and ZNF793 are hypermethylated in BE, and the methylation status of these genes can be used to detect BE in tissue samples. IMPACT These findings support the development of methylated B3GAT2 and ZNF793 as biomarkers for noninvasive assays for the detection of BE.
Collapse
Affiliation(s)
- Ming Yu
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rachele M O'Leary
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Andrew M Kaz
- Research and Development Service, VA Puget Sound Health Care System, Seattle, Washington. Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Shelli M Morris
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kelly T Carter
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Amitabh Chak
- Division of Gastroenterology, University Hospitals Case Medical Center, Cleveland, Ohio. Division of Oncology, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Apoorva Chandar
- Division of Gastroenterology, University Hospitals Case Medical Center, Cleveland, Ohio. Division of Oncology, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Joseph E Willis
- Department of Pathology, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Helen R Moinova
- Department of Medicine, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Sanford D Markowitz
- Department of Medicine, University Hospitals Case Medical Center, Cleveland, Ohio. Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, Ohio
| | - Dean E Brenner
- University of Michigan Medical Center, Ann Arbor, Michigan
| | | | - Maria Westerhoff
- Department of Anatomic Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Chao-Jen Wong
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Nicholas J Shaheen
- Division of Gastroenterology & Hepatology, University of North Carolina Hospitals, Chapel Hill, North Carolina
| | - Yanwen Chen
- Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, Ohio
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, Ohio
| | - William M Grady
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Medicine, University of Washington School of Medicine, Seattle, Washington.
| |
Collapse
|
32
|
Zeb MH, Baruah A, Kossak SK, Buttar NS. Chemoprevention in Barrett's Esophagus: Current Status. Gastroenterol Clin North Am 2015; 44:391-413. [PMID: 26021201 DOI: 10.1016/j.gtc.2015.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chemoprevention in Barrett's esophagus is currently applied only in research settings. Identifying pathways that can be targeted by safe, pharmaceutical or natural compounds is key to expanding the scope of chemoprevention. Defining meaningful surrogate markers of cancer progression is critical to test the efficacy of chemopreventive approaches. Combinatorial chemoprevention that targets multiple components of the same pathway or parallel pathways could reduce the risk and improve the efficacy of chemoprevention. Here we discuss the role of chemoprevention as an independent or an adjuvant management option in BE-associated esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Muhammad H Zeb
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Anushka Baruah
- Department of Internal Medicine, John H. Stroger, Jr. Hospital of Cook County, 1901 W. Harrison Street, Chicago, IL 60612, USA
| | - Sarah K Kossak
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Navtej S Buttar
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
33
|
Kaz AM, Grady WM, Stachler MD, Bass AJ. Genetic and Epigenetic Alterations in Barrett's Esophagus and Esophageal Adenocarcinoma. Gastroenterol Clin North Am 2015; 44:473-89. [PMID: 26021206 PMCID: PMC4449457 DOI: 10.1016/j.gtc.2015.02.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Esophageal adenocarcinoma (EAC) develops from Barrett's esophagus (BE), wherein normal squamous epithelia is replaced by specialized intestinal metaplasia in response to chronic gastroesophageal acid reflux. BE can progress to low- and high-grade dysplasia, intramucosal, and invasive carcinoma. Both BE and EAC are characterized by loss of heterozygosity, aneuploidy, specific genetic mutations, and clonal diversity. Given the limitations of histopathology, genomic and epigenomic analyses may improve the precision of risk stratification. Assays to detect molecular alterations associated with neoplastic progression could be used to improve the pathologic assessment of BE/EAC and to select high-risk patients for more intensive surveillance.
Collapse
Affiliation(s)
- Andrew M. Kaz
- VA Puget Sound Health Care System, R&D Department, Seattle, WA,Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA,University of Washington School of Medicine, Department of Internal Medicine, Seattle, WA
| | - William M. Grady
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA,University of Washington School of Medicine, Department of Internal Medicine, Seattle, WA
| | - Matthew D. Stachler
- Department of Pathology, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
| | - Adam J. Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
34
|
Bansal A, Fitzgerald RC. Biomarkers in Barrett's Esophagus: Role in Diagnosis, Risk Stratification, and Prediction of Response to Therapy. Gastroenterol Clin North Am 2015; 44:373-90. [PMID: 26021200 DOI: 10.1016/j.gtc.2015.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Esophageal adenocarcinoma (EAC) has increased dramatically in the past 3 decades, making its precursor lesion Barrett's esophagus (BE) an important clinical problem. Effective interventions are available, but overall outcomes remain unchanged. Most of the BE population remains undiagnosed; most EACs are diagnosed late, and most BE patients will never progress to cancer. These epidemiologic factors make upper endoscopy an inefficient and ineffective strategy for BE diagnosis and risk stratification. In the current review, biomarkers for diagnosis, risk stratification, and predictors of response to therapy in BE are discussed.
Collapse
Affiliation(s)
- Ajay Bansal
- Division of Gastroenterology and Hepatology, Department of Veterans Affairs Medical Center and the University of Kansas Medical Center, 4801 East Linwood Boulevard, Kansas City, MO 64128-2295, USA.
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Hills Road, Cambridge CB2 0XZ, UK
| |
Collapse
|
35
|
Inadomi JM. Cost considerations in implementing a screening and surveillance strategy for Barrett's oesophagus. Best Pract Res Clin Gastroenterol 2015; 29:51-63. [PMID: 25743456 DOI: 10.1016/j.bpg.2014.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/27/2014] [Accepted: 12/11/2014] [Indexed: 01/31/2023]
Abstract
There is increasing concern over the rising costs of healthcare leading to debate regarding the use of resources to implement preventive strategies. Oesophageal adenocarcinoma and its precursor, Barrett's oesophagus provides an excellent opportunity to highlight this issue since cancer is uncommon even among individuals with documented Barrett's oesophagus. This review provides a brief introduction to economic analysis in healthcare and summarizes published studies of the cost-effectiveness of strategies to reduce mortality from cancer. Current best estimates highlight the cost-effectiveness of endoscopic ablation among patients with Barrett's oesophagus and high-grade dysplasia and the low cost-effectiveness of ablation among patients without dysplasia. The cost-effectiveness of ablation among patients with Barrett's and low-grade dysplasia is poorly defined due to the ambiguity of diagnosing dysplasia, the unknown risk of cancer among patients with low-grade dysplasia, and the uncertain durability of ablation to maintain remission from metaplasia and dysplasia and prevent cancer.
Collapse
Affiliation(s)
- John M Inadomi
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of Washington, United States; Department of Health Services, School of Public Health, University of Washington, United States.
| |
Collapse
|
36
|
Kailasam A, Mittal SK, Agrawal DK. Epigenetics in the Pathogenesis of Esophageal Adenocarcinoma. Clin Transl Sci 2014; 8:394-402. [PMID: 25388215 DOI: 10.1111/cts.12242] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epigenetic influences, such as DNA methylation, histone acetylation, and up-regulation/down-regulation of genes by microRNAs, change the genetic makeup of an individual without affecting DNA base-pair sequences. Indeed, epigenetic changes play an integral role in the progression from normal esophageal mucosa to Barrett's esophagus to esophageal adenocarcinoma via dysplasia-metaplasia-neoplasia sequence. Many genes involved in esophageal adenocarcinoma display hypermethylation, leading to their down-regulation. The classes of these genes include cell cycle control, DNA and growth factor repair, tumor suppressors, antimetastasis, Wnt-related genes, and proapoptotic genes. Histone acetylation in the pathophysiology of esophageal diseases has not been thoroughly investigated, and its critical role in the development of esophageal adenocarcinoma is less defined. Many microRNAs have been associated with the development of Barrett's esophagus and esophageal adenocarcinoma. Here, we critically addressed the specific steps most closely influenced by microRNAs in the progression from Barrett's esophagus to esophageal adenocarcinoma. However, microRNAs can target up to hundreds of genes, making it difficult to correlate directly with a given phenotype of the disease. Esophageal adenocarcinoma progressing from premalignant condition of Barrett's esophagus carries an extremely poor prognosis. Risk stratification for patients based on their epigenetic profiles may be useful in providing more targeted and directed treatment to patients.
Collapse
Affiliation(s)
- Aparna Kailasam
- School of Medicine, Center for Clinical & Translational Science, Creighton University, Omaha, NE, USA
| | - Sumeet K Mittal
- School of Medicine, Center for Clinical & Translational Science, Creighton University, Omaha, NE, USA
| | - Devendra K Agrawal
- School of Medicine, Center for Clinical & Translational Science, Creighton University, Omaha, NE, USA
| |
Collapse
|
37
|
di Pietro M, Alzoubaidi D, Fitzgerald RC. Barrett's esophagus and cancer risk: how research advances can impact clinical practice. Gut Liver 2014; 8:356-70. [PMID: 25071900 PMCID: PMC4113043 DOI: 10.5009/gnl.2014.8.4.356] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/15/2014] [Indexed: 12/18/2022] Open
Abstract
Barrett’s esophagus (BE) is the only known precursor to esophageal adenocarcinoma (EAC), whose incidence has increased sharply in the last 4 decades. The annual conversion rate of BE to cancer is significant, but small. The identification of patients at a higher risk of cancer therefore poses a clinical conundrum. Currently, endoscopic surveillance is recommended in BE patients, with the aim of diagnosing either dysplasia or cancer at early stages, both of which are curable with minimally invasive endoscopic techniques. There is a large variation in clinical practice for endoscopic surveillance, and dysplasia as a marker of increased risk is affected by sampling error and high interobserver variability. Screening programs have not yet been formally accepted, mainly due to the economic burden that would be generated by upper gastrointestinal endoscopy. Screening programs have not yet been formally accepted, mainly due to the economic burden that would be generated by widespread indication to upper gastrointestinal endoscopy. In fact, it is currently difficult to formulate an accurate algorithm to confidently target the population at risk, based on the known clinical risk factors for BE and EAC. This review will focus on the clinical and molecular factors that are involved in the development of BE and its conversion to cancer and on how increased knowledge in these areas can improve the clinical management of the disease.
Collapse
Affiliation(s)
| | - Durayd Alzoubaidi
- Department of Gastroenterology, Basildon and Thurrock University Hospital, Basildon, UK
| | | |
Collapse
|
38
|
Kaz AM, Wong CJ, Dzieciatkowski S, Luo Y, Schoen RE, Grady WM. Patterns of DNA methylation in the normal colon vary by anatomical location, gender, and age. Epigenetics 2014; 9:492-502. [PMID: 24413027 DOI: 10.4161/epi.27650] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Alterations in DNA methylation have been proposed to create a field cancerization state in the colon, where molecular alterations that predispose cells to transformation occur in histologically normal tissue. However, our understanding of the role of DNA methylation in field cancerization is limited by an incomplete characterization of the methylation state of the normal colon. In order to determine the colon's normal methylation state, we extracted DNA from normal colon biopsies from the rectum, sigmoid, transverse, and ascending colon and assessed the methylation status of the DNA by pyrosequencing candidate loci as well as with HumanMethylation450 arrays. We found that methylation levels of repetitive elements LINE-1 and SAT-α showed minimal variability throughout the colon in contrast to other loci. Promoter methylation of EVL was highest in the rectum and progressively lower in the proximal segments, whereas ESR1 methylation was higher in older individuals. Genome-wide methylation analysis of normal DNA revealed 8388, 82, and 93 differentially methylated loci that distinguished right from left colon, males from females, and older vs. younger individuals, respectively. Although variability in methylation between biopsies and among different colon segments was minimal for repetitive elements, analyses of specific cancer-related genes as well as a genome-wide methylation analysis demonstrated differential methylation based on colon location, individual age, and gender. These studies advance our knowledge regarding the variation of DNA methylation in the normal colon, a prerequisite for future studies aimed at understanding methylation differences indicative of a colon field effect.
Collapse
Affiliation(s)
- Andrew M Kaz
- Division of Clinical Research; Fred Hutchinson Cancer Research Center; Seattle, WA USA; Research and Development Service; VA Puget Sound Health Care System; Seattle, WA USA; Department of Medicine; University of Washington School of Medicine; Seattle, WA USA
| | - Chao-Jen Wong
- Division of Clinical Research; Fred Hutchinson Cancer Research Center; Seattle, WA USA
| | | | - Yanxin Luo
- Division of Clinical Research; Fred Hutchinson Cancer Research Center; Seattle, WA USA; Department of Colorectal Surgery; the Sixth Affiliated Hospital; Sun Yat-Sen University; Guangzhou, PR China; Gastrointestinal Institute; Sun Yat-Sen University; Guangzhou, PR China
| | - Robert E Schoen
- Department of Medicine; University of Pittsburgh Medical Center; Pittsburgh, PA USA
| | - William M Grady
- Division of Clinical Research; Fred Hutchinson Cancer Research Center; Seattle, WA USA; Department of Medicine; University of Washington School of Medicine; Seattle, WA USA
| |
Collapse
|
39
|
Abstract
Barrett's esophagus (BE) is defined as the metaplastic conversion of the distal esophageal squamous epithelium to intestinalized columnar epithelium. It is a premalignant condition associated with esophageal adenocarcinoma (EAC) and is the major risk factor for EAC. Recent studies suggest that the molecular mechanisms responsible for the pathogenesis of BE are closely related to transcription factors, signaling proteins and microRNAs (miRNAs). MiRNAs are expected to be used as novel biomarkers for the diagnosis, prognosis assessment and targeted treatment of EAC. This article summarizes recent results involving stem cells, immune factors, transcription factors, DNA methylation, nitric oxide, signaling pathways, microRNAs in the development of BE. Understanding of the molecular mechanisms behind the pathogenesis of BE has important implications for improved management of BE and EAC.
Collapse
|
40
|
Xu E, Gu J, Hawk ET, Wang KK, Lai M, Huang M, Ajani J, Wu X. Genome-wide methylation analysis shows similar patterns in Barrett's esophagus and esophageal adenocarcinoma. Carcinogenesis 2013; 34:2750-6. [PMID: 23996928 DOI: 10.1093/carcin/bgt286] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Barrett's esophagus (BE) is a precursor of esophageal adenocarcinoma (EAC). To identify novel tumor suppressors involved in esophageal carcinogenesis and potential biomarkers for the malignant progression of BE, we performed a genome-wide methylation profiling of BE and EAC tissues. Using Illumina's Infinium HumanMethylation27 BeadChip microarray, we examined the methylation status of 27 578 CpG sites in 94 normal esophageal (NE), 77 BE and 117 EAC tissue samples. The overall methylation of CpG sites within the CpG islands was higher, but outside of the CpG islands was lower in BE and EAC tissues than in NE tissues. Hierarchical clustering analysis showed an excellent separation of NE tissues from BE and EAC tissues; however, the clustering of BE and EAC tissues was less clear, suggesting that methylation occurs early during the progression of EAC. We confirmed many previously reported hypermethylated genes and identified a large number of novel hypermethylated genes in BE and EAC tissues, particularly genes encoding ADAM (A Disintegrin And Metalloproteinase) peptidase proteins, cadherins and protocadherins, and potassium voltage-gated channels. Pathway analysis showed that a number of channel and transporter activities were enriched for hypermethylated genes. We used pyrosequencing to validate selected candidate genes and found high correlations between the array and pyrosequencing data (rho > 0.8 for each validated gene). The differentially methylated genes and pathways may provide biological insights into the development and progression of BE and become potential biomarkers for the prediction and early detection of EAC.
Collapse
Affiliation(s)
- Enping Xu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Luo Y, Kaz AM, Kanngurn S, Welsch P, Morris SM, Wang J, Lutterbaugh JD, Markowitz SD, Grady WM. NTRK3 is a potential tumor suppressor gene commonly inactivated by epigenetic mechanisms in colorectal cancer. PLoS Genet 2013; 9:e1003552. [PMID: 23874207 PMCID: PMC3708790 DOI: 10.1371/journal.pgen.1003552] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 04/23/2013] [Indexed: 12/03/2022] Open
Abstract
NTRK3 is a member of the neurotrophin receptor family and regulates cell survival. It appears to be a dependence receptor, and thus has the potential to act as an oncogene or as a tumor suppressor gene. NTRK3 is a receptor for NT-3 and when bound to NT-3 it induces cell survival, but when NT-3 free, it induces apoptosis. We identified aberrantly methylated NTRK3 in colorectal cancers through a genome-wide screen for hypermethylated genes. This discovery led us to assess whether NTRK3 could be a tumor suppressor gene in the colon. NTRK3 is methylated in 60% of colon adenomas and 67% of colon adenocarcinomas. NTRK3 methylation suppresses NTRK3 expression. Reconstitution of NTRK3 induces apoptosis in colorectal cancers, if NT-3 is absent. Furthermore, the loss of NTRK3 expression associates with neoplastic transformation in vitro and in vivo. We also found that a naturally occurring mutant NTRK3 found in human colorectal cancer inhibits the tumor suppressor activity of NTRK3. In summary, our findings suggest NTRK3 is a conditional tumor suppressor gene that is commonly inactivated in colorectal cancer by both epigenetic and genetic mechanisms whose function in the pathogenesis of colorectal cancer depends on the expression status of its ligand, NT-3. NTRK3 is a neurotrophin receptor and appears to be a dependence receptor in certain tissues. NTRK3 has been previously shown to be an oncogene in breast cancer and possibly hepatocellular carcinoma. Through a genome-wide methylation screen, we unexpectedly found that NTRK3 is commonly methylated in colorectal cancers but not in normal colon samples, which led us to assess whether NTRK3 could be a tumor suppressor gene in the colon. We now demonstrate that NTRK3 is frequently methylated in colorectal adenomas and cancers. Induced NTRK3 expression in the absence of its ligand, NT-3, causes apoptosis and suppresses in vitro anchorage-independent colony formation and in vivo tumor growth. Reintroduction of NT-3 releases colon cancer cells from NTRK3-mediated apoptosis, which is consistent with NTRK3 being a dependence receptor in the colon. Finally, somatic mutations of NTRK3 have been observed in primary human colorectal cancer. We provide evidence that a subset of these mutations inactivate tumor suppressor activities of NTRK3. These findings suggest that NTRK3 is a conditional tumor suppressor gene in the colon that is inactivated by both genetic and epigenetic mechanisms and whose function in the pathogenesis of colorectal cancer depends on the expression status of its ligand, NT-3.
Collapse
Affiliation(s)
- Yanxin Luo
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Andrew M. Kaz
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Research and Development Service, VA Puget Sound Health Care System, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Samornmas Kanngurn
- Tumor Biology Research Unit and Department of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Piri Welsch
- Division of Medical Genetics, University of Washington Medical School, Seattle, Washington, United States of America
| | - Shelli M. Morris
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jianping Wang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - James D. Lutterbaugh
- Department of Medicine and Ireland Cancer Center, Case Western Reserve University School of Medicine and Case Medical Center, Cleveland, Ohio, United States of America
| | - Sanford D. Markowitz
- Department of Medicine and Ireland Cancer Center, Case Western Reserve University School of Medicine and Case Medical Center, Cleveland, Ohio, United States of America
| | - William M. Grady
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
42
|
Clemons NJ, Phillips WA, Lord RV. Signaling pathways in the molecular pathogenesis of adenocarcinomas of the esophagus and gastroesophageal junction. Cancer Biol Ther 2013; 14:782-95. [PMID: 23792587 PMCID: PMC3909547 DOI: 10.4161/cbt.25362] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Esophageal adenocarcinoma develops in response to severe gastroesophageal reflux disease through the precursor lesion Barrett esophagus, in which the normal squamous epithelium is replaced by a columnar lining. The incidence of esophageal adenocarcinoma in the United States has increased by over 600% in the past 40 years and the overall survival rate remains less than 20% in the community. This review highlights some of the signaling pathways for which there is some evidence of a role in the development of esophageal adenocarcinoma. An increasingly detailed understanding of the biology of this cancer has emerged recently, revealing that in addition to the well-recognized alterations in single genes such as p53, p16, APC, and telomerase, there are interactions between the components of the reflux fluid, the homeobox gene Cdx2, and the Wnt, Notch, and Hedgehog signaling pathways.
Collapse
Affiliation(s)
- Nicholas J Clemons
- Surgical Oncology Research Laboratory; Peter MacCallum Cancer Centre; East Melbourne, Australia; Sir Peter MacCallum Department of Oncology; University of Melbourne, Melbourne, Australia; Department of Surgery (St. Vincent's Hospital); University of Melbourne; Melbourne, Australia
| | - Wayne A Phillips
- Surgical Oncology Research Laboratory; Peter MacCallum Cancer Centre; East Melbourne, Australia; Sir Peter MacCallum Department of Oncology; University of Melbourne, Melbourne, Australia; Department of Surgery (St. Vincent's Hospital); University of Melbourne; Melbourne, Australia
| | - Reginald V Lord
- St. Vincent's Centre for Applied Medical Research; Sydney, Australia; Notre Dame University School of Medicine; Sydney, Australia
| |
Collapse
|
43
|
Shah AK, Saunders NA, Barbour AP, Hill MM. Early diagnostic biomarkers for esophageal adenocarcinoma--the current state of play. Cancer Epidemiol Biomarkers Prev 2013; 22:1185-209. [PMID: 23576690 DOI: 10.1158/1055-9965.epi-12-1415] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is one of the two most common types of esophageal cancer with alarming increase in incidence and very poor prognosis. Aiming to detect EAC early, currently high-risk patients are monitored using an endoscopic-biopsy approach. However, this approach is prone to sampling error and interobserver variability. Diagnostic tissue biomarkers related to genomic and cell-cycle abnormalities have shown promising results, although with current technology these tests are difficult to implement in the screening of high-risk patients for early neoplastic changes. Differential miRNA profiles and aberrant protein glycosylation in tissue samples have been reported to improve performance of existing tissue-based diagnostic biomarkers. In contrast to tissue biomarkers, circulating biomarkers are more amenable to population-screening strategies, due to the ease and low cost of testing. Studies have already shown altered circulating glycans and DNA methylation in BE/EAC, whereas disease-associated changes in circulating miRNA remain to be determined. Future research should focus on identification and validation of these circulating biomarkers in large-scale trials to develop in vitro diagnostic tools to screen population at risk for EAC development.
Collapse
Affiliation(s)
- Alok Kishorkumar Shah
- The University of Queensland Diamantina Institute; and School of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia
| | | | | | | |
Collapse
|
44
|
GERD-Barrett-Adenocarcinoma: Do We Have Suitable Prognostic and Predictive Molecular Markers? Gastroenterol Res Pract 2013; 2013:643084. [PMID: 23573078 PMCID: PMC3615572 DOI: 10.1155/2013/643084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/18/2013] [Indexed: 02/07/2023] Open
Abstract
Due to unfavorable lifestyle habits (unhealthy diet and tobacco abuse) the incidence of gastroesophageal reflux disease (GERD) in western countries is increasing. The GERD-Barrett-Adenocarcinoma sequence currently lacks well-defined diagnostic, progressive, predictive, and prognostic biomarkers (i) providing an appropriate screening method identifying the presence of the disease, (ii) estimating the risk of evolving cancer, that is, the progression from Barrett's esophagus (BE) to esophageal adenocarcinoma (EAC), (iii) predicting the response to therapy, and (iv) indicating an overall survival—prognosis for EAC patients. Based on histomorphological findings, detailed screening and therapeutic guidelines have been elaborated, although epidemiological studies could not support the postulated increasing progression rates of GERD to BE and EAC. Additionally, proposed predictive and prognostic markers are rather heterogeneous by nature, lack substantial proofs, and currently do not allow stratification of GERD patients for progression, outcome, and therapeutic effectiveness in clinical practice. The aim of this paper is to discuss the current knowledge regarding the GERD-BE-EAC sequence mainly focusing on the disputable and ambiguous status of proposed biomarkers to identify promising and reliable markers in order to provide more detailed insights into pathophysiological mechanisms and thus to improve prognostic and predictive therapeutic approaches.
Collapse
|
45
|
Alvi MA, Liu X, O'Donovan M, Newton R, Wernisch L, Shannon NB, Shariff K, di Pietro M, Bergman JJGHM, Ragunath K, Fitzgerald RC. DNA methylation as an adjunct to histopathology to detect prevalent, inconspicuous dysplasia and early-stage neoplasia in Barrett's esophagus. Clin Cancer Res 2013; 19:878-88. [PMID: 23243219 PMCID: PMC4998953 DOI: 10.1158/1078-0432.ccr-12-2880] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Endoscopic surveillance of Barrett's esophagus is problematic because dysplasia/early-stage neoplasia is frequently invisible and likely to be missed because of sampling bias. Molecular abnormalities may be more diffuse than dysplasia. The aim was therefore to test whether DNA methylation, especially on imprinted and X-chromosome genes, is able to detect dysplasia/early-stage neoplasia. EXPERIMENTAL DESIGN 27K methylation arrays were used to find genes best able to differentiate between 22 Barrett's esophagus and 24 esophageal adenocarcinoma (EAC) samples. These were validated using pyrosequencing on a retrospective cohort (60 Barrett's esophagus, 36 dysplastic, and 90 EAC) and then in a prospective multicenter study (98 Barrett's esophagus patients, including 28 dysplastic and 9 early EAC) designed to utilize biomarkers to stratify patients according to their prevalent dysplasia/EAC status. RESULTS Genes (23%) on the array, including 7% of X-linked and 69% of imprinted genes, have shown statistically significant changes in methylation in EAC versus Barrett's esophagus (Wilcoxon P < 0.05). 6/7 selected candidate genes were successfully internally (Pearson's P < 0.01) and externally validated (ANOVA P < 0.001). Four genes (SLC22A18, PIGR, GJA12, and RIN2) showed the greatest area under curve (0.988) to distinguish between Barrett's esophagus and dysplasia/EAC in the retrospective cohort. This methylation panel was able to stratify patients from the prospective cohort into three risk groups based on the number of genes methylated (low risk: <2 genes, intermediate: 2, and high: >2). CONCLUSION Widespread DNA methylation changes were observed in Barrett's carcinogenesis including ≈70% of known imprinted genes. A four-gene methylation panel stratified patients with Barrett's esophagus into three risk groups with potential clinical utility.
Collapse
Affiliation(s)
- Muhammad A Alvi
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Chisholm JA, Mayne GC, Hussey DJ, Watson DI. Molecular biomarkers and ablative therapies for Barrett's esophagus. Expert Rev Gastroenterol Hepatol 2012; 6:567-81. [PMID: 23061708 DOI: 10.1586/egh.12.39] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Barrett's esophagus is the major risk factor for esophageal adenocarcinoma. Endoscopic interventions that ablate Barrett's esophagus mucosa lead to replacement with a new squamous (neosquamous) mucosa, but it can be difficult to achieve complete ablation. Knowing whether cancer is less likely to develop in neosquamous mucosa or residual Barrett's esophagus after ablation is critical for determining the efficacy of treatment. This issue can be informed by assessing biomarkers that are associated with an increased risk of progression to adenocarcinoma. Although there are few postablation biomarker studies, evidence suggests that neosquamous mucosa may have a reduced risk of adenocarcinoma in patients who have been treated for dysplasia or cancer, but some patients who do not have complete eradication of nondysplastic Barrett's esophagus may still be at risk. Biomarkers could be used to optimize endoscopic surveillance strategies following ablation, but this needs to be assessed by clinical studies and economic modeling.
Collapse
Affiliation(s)
- Jacob A Chisholm
- Flinders University Department of Surgery, Flinders Medical Centre, Bedford Park, South Australia 5042, Australia
| | | | | | | |
Collapse
|
47
|
Kaz AM, Grady WM. Epigenetic biomarkers in esophageal cancer. Cancer Lett 2012; 342:193-9. [PMID: 22406828 DOI: 10.1016/j.canlet.2012.02.036] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 02/25/2012] [Accepted: 02/29/2012] [Indexed: 12/13/2022]
Abstract
The aberrant DNA methylation of tumor suppressor genes is well documented in esophageal cancer, including adenocarcinoma (EAC) and squamous cell carcinoma (ESCC) as well as in Barrett's esophagus (BE), a pre-malignant condition that is associated with chronic acid reflux. BE is a well-recognized risk factor for the development of EAC, and consequently the standard of care is for individuals with BE to be placed in endoscopic surveillance programs aimed at detecting early histologic changes that associate with an increased risk of developing EAC. Yet because the absolute risk of EAC in individuals with BE is minimal, a clinical need in the management of BE is the identification of additional risk markers that will indicate individuals who are at a significant absolute risk of EAC so that they may be subjected to more intensive surveillance. The best currently available risk marker is the degree of dysplasia in endoscopic biopsies from the esophagus; however, this marker is suboptimal for a variety of reasons. To date, there are no molecular biomarkers that have been translated to widespread clinical practice. The search for biomarkers, including hypermethylated genes, for either the diagnosis of BE, EAC, or ESCC or for risk stratification for the development of EAC in those with BE is currently an area of active research. In this review, we summarize the status of identified candidate epigenetic biomarkers for BE, EAC, and ESCC. Most of these aberrantly methylated genes have been described in the context of early detection or diagnostic markers; others might prove useful for estimating prognosis or predicting response to treatment. Finally, special attention will be paid to some of the challenges that must be overcome in order to develop clinically useful esophageal cancer biomarkers.
Collapse
Affiliation(s)
- Andrew M Kaz
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, United States; Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States; Research and Development Service, VA Puget Sound Health Care System, Seattle, WA, United States.
| | | |
Collapse
|