1
|
Patil DT, Odze RD. Barrett's Esophagus and Associated Dysplasia. Gastroenterol Clin North Am 2024; 53:1-23. [PMID: 38280743 DOI: 10.1016/j.gtc.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2024]
Abstract
Early detection of dysplasia and effective management are critical steps in halting neoplastic progression in patients with Barrett's esophagus (BE). This review provides a contemporary overview of the BE-related dysplasia, its role in guiding surveillance and management, and discusses emerging diagnostic and therapeutic approaches that might further enhance patient management. Novel, noninvasive techniques for sampling and surveillance, adjunct biomarkers for risk assessment, and their limitations are also discussed.
Collapse
Affiliation(s)
- Deepa T Patil
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Robert D Odze
- Department of Pathology and Lab Medicine, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
2
|
Ooki A, Osumi H, Chin K, Watanabe M, Yamaguchi K. Potent molecular-targeted therapies for advanced esophageal squamous cell carcinoma. Ther Adv Med Oncol 2023; 15:17588359221138377. [PMID: 36872946 PMCID: PMC9978325 DOI: 10.1177/17588359221138377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 10/21/2022] [Indexed: 01/15/2023] Open
Abstract
Esophageal cancer (EC) remains a public health concern with a high mortality and disease burden worldwide. Esophageal squamous cell carcinoma (ESCC) is a predominant histological subtype of EC that has unique etiology, molecular profiles, and clinicopathological features. Although systemic chemotherapy, including cytotoxic agents and immune checkpoint inhibitors, is the main therapeutic option for recurrent or metastatic ESCC patients, the clinical benefits are limited with poor prognosis. Personalized molecular-targeted therapies have been hampered due to the lack of robust treatment efficacy in clinical trials. Therefore, there is an urgent need to develop effective therapeutic strategies. In this review, we summarize the molecular profiles of ESCC based on the findings of pivotal comprehensive molecular analyses, highlighting potent therapeutic targets for establishing future precision medicine for ESCC patients, with the most recent results of clinical trials.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Gastroenterological Chemotherapy,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31
Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Hiroki Osumi
- Department of Gastroenterological Chemotherapy,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo,
Japan
| | - Keisho Chin
- Department of Gastroenterological Chemotherapy,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo,
Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo,
Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy,
Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo,
Japan
| |
Collapse
|
3
|
Jiang J, Mei J, Ma Y, Jiang S, Zhang J, Yi S, Feng C, Liu Y, Liu Y. Tumor hijacks macrophages and microbiota through extracellular vesicles. EXPLORATION (BEIJING, CHINA) 2022; 2:20210144. [PMID: 37324578 PMCID: PMC10190998 DOI: 10.1002/exp.20210144] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/16/2021] [Indexed: 06/17/2023]
Abstract
The tumor microenvironment (TME) is a biological system with sophisticated constituents. In addition to tumor cells, tumor-associated macrophages (TAMs) and microbiota are also dominant components. The phenotypic and functional changes of TAMs are widely considered to be related to most tumor progressions. The chronic colonization of pathogenic microbes and opportunistic pathogens accounts for the generation and development of tumors. As messengers of cell-to-cell communication, tumor-derived extracellular vesicles (TDEVs) can transfer various malignant factors, regulating physiological and pathological changes in the recipients and affecting TAMs and microbes in the TME. Despite the new insights into tumorigenesis and progress brought by the above factors, the crosstalk among tumor cells, macrophages, and microbiota remain elusive, and few studies have focused on how TDEVs act as an intermediary. We reviewed how tumor cells recruit and domesticate macrophages and microbes through extracellular vehicles and how hijacked macrophages and microbiota interact with tumor-promoting feedback, achieving a reciprocal coexistence under the TME and working together to facilitate tumor progression. It is significant to seek evidence to clarify those specific interactions and reveal therapeutic targets to curb tumor progression and improve prognosis.
Collapse
Affiliation(s)
- Jipeng Jiang
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Jie Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing P. R. China
- University of Chinese Academy of Science Beijing P. R. China
| | - Yongfu Ma
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Shasha Jiang
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Jian Zhang
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Shaoqiong Yi
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Changjiang Feng
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Yang Liu
- Postgraduate School Medical School of Chinese PLA Beijing P. R. China
- Department of Thoracic Surgery The First Medical Center of Chinese PLA General Hospital Beijing P. R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Beijing P. R. China
- GBA National Institute for Nanotechnology Innovation Guangdong P. R. China
| |
Collapse
|
4
|
Jiang J, Mei J, Yi S, Feng C, Ma Y, Liu Y, Liu Y, Chen C. Tumor associated macrophage and microbe: The potential targets of tumor vaccine delivery. Adv Drug Deliv Rev 2022; 180:114046. [PMID: 34767863 DOI: 10.1016/j.addr.2021.114046] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023]
Abstract
The occurrence and development of tumors depend on the tumor microenvironment (TME), which is made of various immune cells, activated fibroblasts, basement membrane, capillaries, and extracellular matrix. Tumor associated macrophages (TAMs) and microbes are important components in TME. Tumor cells can recruit and educate TAMs and microbes, and the hijacked TAMs and microbes can promote the progression of tumor reciprocally. Tumor vaccine delivery remodeling TME by targeting TAM and microbes can not only enhance the specificity and immunogenicity of antigens, but also contribute to the regulation of TME. Tumor vaccine design benefits from nanotechnology which is a suitable platform for antigen and adjuvant delivery to catalyze new candidate vaccines applying to clinical therapy at unparalleled speed. In view of the characteristics and mechanisms of TME development, vaccine delivery targeting and breaking the malignant interactions among tumor cells, TAMs, and microbes may serve as a novel strategy for tumor therapy.
Collapse
|
5
|
Shi H, Ju Q, Mao Y, Wang Y, Ding J, Liu X, Tang X, Sun C. TAK1 Phosphorylates RASSF9 and Inhibits Esophageal Squamous Tumor Cell Proliferation by Targeting the RAS/MEK/ERK Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001575. [PMID: 33717835 PMCID: PMC7927628 DOI: 10.1002/advs.202001575] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/20/2020] [Indexed: 05/11/2023]
Abstract
TGF-β-activated kinase 1 (TAK1), a serine/threonine kinase, is a key intermediate in several signaling pathways. However, its role in tumorigenesis is still not understood well. In this study, it is found that TAK1 expression decreases in esophageal tumor tissues and cell lines. In vitro experiments demonstrate that proliferation of esophageal tumor cells is enhanced by knockdown of TAK1 expression and attenuated by elevated expression of TAK1. Using a subcutaneous tumor model, these observations are confirmed in vivo. Based on the results from co-immunoprecipitation coupled with mass spectrometry, Ras association domain family 9 (RASSF9) is identified as a downstream target of TAK1. TAK1 phosphorylates RASSF9 at S284, which leads to reduced RAS dimerization, thereby blocking RAF/MEK/ERK signal transduction. Clinical survey reveals that TAK1 expression is inversely correlated with survival in esophageal cancer patients. Taken together, the data reveal that TAK1-mediated phosphorylation of RASSF9 at Ser284 negatively regulates esophageal tumor cell proliferation via inhibition of the RAS/MEK/ERK axis.
Collapse
Affiliation(s)
- Hui Shi
- Department of Cardiothoracic SurgeryNantong Key Laboratory of Translational Medicine in Cardiothoracic DiseasesNantong Clinical Medical Research Center of Cardiothoracic DiseaseInstitution of Translational Medicine in Cardiothoracic DiseasesAffiliated Hospital of Nantong University20 Xisi RoadNantong226001China
| | - Qianqian Ju
- Department of Cardiothoracic SurgeryNantong Key Laboratory of Translational Medicine in Cardiothoracic DiseasesNantong Clinical Medical Research Center of Cardiothoracic DiseaseInstitution of Translational Medicine in Cardiothoracic DiseasesAffiliated Hospital of Nantong University20 Xisi RoadNantong226001China
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of EducationNantong University19 Qixiu RoadNantong226001China
| | - Yinting Mao
- Department of Cardiothoracic SurgeryNantong Key Laboratory of Translational Medicine in Cardiothoracic DiseasesNantong Clinical Medical Research Center of Cardiothoracic DiseaseInstitution of Translational Medicine in Cardiothoracic DiseasesAffiliated Hospital of Nantong University20 Xisi RoadNantong226001China
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of EducationNantong University19 Qixiu RoadNantong226001China
| | - Yuejun Wang
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of EducationNantong University19 Qixiu RoadNantong226001China
| | - Jie Ding
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of EducationNantong University19 Qixiu RoadNantong226001China
| | - Xiaoyu Liu
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of EducationNantong University19 Qixiu RoadNantong226001China
| | - Xin Tang
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of EducationNantong University19 Qixiu RoadNantong226001China
| | - Cheng Sun
- Department of Cardiothoracic SurgeryNantong Key Laboratory of Translational Medicine in Cardiothoracic DiseasesNantong Clinical Medical Research Center of Cardiothoracic DiseaseInstitution of Translational Medicine in Cardiothoracic DiseasesAffiliated Hospital of Nantong University20 Xisi RoadNantong226001China
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of EducationNantong University19 Qixiu RoadNantong226001China
| |
Collapse
|
6
|
Dhillon VS, Deo P, Bonassi S, Fenech M. Lymphocyte micronuclei frequencies in skin, haematological, prostate, colorectal and esophageal cancer cases: A systematic review and meta-analysis. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 787:108372. [PMID: 34083057 DOI: 10.1016/j.mrrev.2021.108372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/17/2021] [Accepted: 01/31/2021] [Indexed: 01/07/2023]
Abstract
Micronucleus (MN) assay has been widely used as a biomarker of DNA damage, chromosomal instability, cancer risk and accelerated aging in many epidemiological studies. In this narrative review and meta-analysis we assessed the association between lymphocyte micronuclei (MNi) and cancers of the skin, blood, digestive tract, and prostate. The review identified nineteen studies with 717 disease subjects and 782 controls. Significant increases in MRi for MNi were observed in the following groups: subjects with blood cancer (MRi = 3.98; 95 % CI: 1.98-7.99; p = 0.000) and colorectal cancer (excluding IBD) (MRi = 2.69; 95 % CI: 1.82-3.98, p < 0.000). The results of this review suggest that lymphocyte MNi are a biomarker of DNA damage and chromosomal instability in people with haematological or colorectal cancers. However, the MRi for lymphocyte MNi in subjects with cancers of skin, prostate, esophagus was not significantly increased. More case-control and prospective studies are warranted to further verify the observed trends and to better understand the role of lymphocyte MNi as a biomarker of cancer risk in blood, skin, digestive tract and prostate.
Collapse
Affiliation(s)
- Varinderpal S Dhillon
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia.
| | - Permal Deo
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Stefano Bonassi
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy; Unit of Clinical and Molecular Epidemiology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Michael Fenech
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia; Faculty of Health Sciences, National University of Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Grady WM, Yu M, Markowitz SD, Chak A. Barrett's Esophagus and Esophageal Adenocarcinoma Biomarkers. Cancer Epidemiol Biomarkers Prev 2020; 29:2486-2494. [PMID: 33093162 DOI: 10.1158/1055-9965.epi-20-0223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/31/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
Esophageal adenocarcinoma is a major cause of cancer-related morbidity and mortality in Western countries. The incidences of esophageal adenocarcinoma and its precursor Barrett's esophagus have increased substantially in the last four decades. Current care guidelines recommend that endoscopy be used for the early detection and monitoring of patients with Barrett's esophagus; however, the efficacy of this approach is unclear. To prevent the increasing morbidity and mortality from esophageal adenocarcinoma, there is a tremendous need for early detection and surveillance biomarker assays that are accurate, low-cost, and clinically feasible to implement. The last decade has seen remarkable advances in the development of minimally invasive molecular biomarkers, an effort led in large part by the Early Detection Research Network (EDRN). Advances in multi-omics analysis, the development of swallowable cytology collection devices, and emerging technology have led to promising assays that are likely to be implemented into clinical care in the next decade. In this review, an updated overview of the molecular pathology of Barrett's esophagus and esophageal adenocarcinoma and emerging molecular biomarker assays, as well as the role of EDRN in biomarker discovery and validation, will be discussed.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- William M Grady
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington. .,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ming Yu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sanford D Markowitz
- Oncology Division, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Amitabh Chak
- Gastroenterology Division, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
8
|
Xia Y, Fang Y, Zhang H, Shen C, Wang P, Yan W, Li J, Xu Y, Shao S, Zhang Y, Yu X, Peng Z, Peng G, Chen W, Fang D. Role of Kruppel-Like Factor 5 in Deoxycholic Acid-Mediated Intestinal Transdifferentiation of Esophageal Squamous Epithelium. J Cancer 2019; 10:5597-5607. [PMID: 31632504 PMCID: PMC6775683 DOI: 10.7150/jca.30050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 05/26/2019] [Indexed: 12/15/2022] Open
Abstract
Barrett's esophagus (BE) is an acquired condition in which normal squamous epithelium is replaced with metaplastic columnar epithelium as a consequence of gastroesophageal reflux disease. BE is known as a precursor of esophageal adenocarcinoma. Currently, the molecular mechanism underlying epithelial metaplasia in BE patients remains unknown. Therefore, we investigated the role of Krüppel-like factor 5 (KLF5) signaling in the initiation of BE-associated metaplasia. Sprague-Dawley (SD) rats were used to create a surgical model of bile reflux injury. Immunohistochemistry was performed to analyze human and mouse esophageal specimens. Human esophageal squamous epithelial (HET-1A) cells were treated with bile acid and used in transfection experiments. Quantitative real-time PCR and western blot analysis were performed to detect the expression of KLF5, CDX2, MUC2 and villin. Epithelial tissue from both the rat BE model and human BE patients strongly expressed KLF5, CDX2, MUC2, and villin. Bile acid treatment also increased the expression of KLF5, CDX2, MUC2 and villin in esophageal epithelial cells in a time-dependent manner. Moreover, siRNA-mediated knockdown of KLF5 blocked the expression of CDX2, MUC2 and villin, but transfection of a KLF5 expression vector into esophageal epithelial cells promoted their transdifferentiation into columnar-like cells, as demonstrated by increased expression of the intestinal markers CDX2, MUC2 and villin. Thus, in addition to its function as a transcription factor, KLF5 may be linked to an increased risk of BE development.
Collapse
Affiliation(s)
- Yiju Xia
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Yu Fang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Haoxiang Zhang
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Caifei Shen
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Pu Wang
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Wu Yan
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Jingwen Li
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Yin Xu
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Shunzi Shao
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Yafei Zhang
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Xiaona Yu
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Zhihong Peng
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Guiyong Peng
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Wensheng Chen
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| | - Dianchun Fang
- Department of Gastroenterology, Southwest Hospital, Army Medical University, Chongqing, 400038, P.R. China
| |
Collapse
|
9
|
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: 4.7] [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
|
10
|
Chen B, Yu J, Lu L, Dong F, Zhou F, Tao X, Sun E. Upregulated forkhead-box A3 elevates the expression of forkhead-box A1 and forkhead-box A2 to promote metastasis in esophageal cancer. Oncol Lett 2019; 17:4351-4360. [PMID: 30944629 DOI: 10.3892/ol.2019.10078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 02/04/2019] [Indexed: 11/06/2022] Open
Abstract
Esophageal cancer (EC) is one of the most lethal cancers currently known. Members of the forkhead-box A (FOXA) family, including FOXA1 and FOXA2, have been reported to regulate EC progression. However, the role of FOXA3, which is another FOXA member, has not yet been investigated. In the present study, public dataset analyses and immunohistochemistry of 96 samples from patients with EC were performed to determine the potential roles of FOXA3 in EC. The results revealed that FOXA3 was significantly upregulated in EC tumor tissues and Barrett's esophagus tissues. In addition, FOXA3 upregulation was positively associated with tumor invasion, distant metastasis, tumor-node-metastasis stage and shorter overall survival in patients with EC, and multivariate analysis identified FOXA3 as an independent prognostic marker. In vitro experiments demonstrated that the migratory and invasive abilities of EC109 and EC9706 cell lines were inhibited following FOXA3 knockdown. Notably, FOXA3 expression levels were positively correlated with FOXA1 and FOXA2 expression levels according to The Cancer Genome Atlas dataset analysis. Furthermore, FOXA3 knockdown decreased the expression levels of FOXA1 and FOXA2 in EC109 and EC9706 cell lines. Conversely, FOXA1 or FOXA2 overexpression compensated for the effects of FOXA3 knockdown on the migratory and invasive capacities of EC cells. In conclusion, the present study demonstrated that FOXA3 upregulation in EC cells promoted metastasis through regulation of other FOXA members.
Collapse
Affiliation(s)
- Bing Chen
- Department of Pathology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Jiegen Yu
- Department of Management Science, School of Humanities and Management, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Linming Lu
- Department of Pathology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Fangyuan Dong
- Department of Pathology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Fangfang Zhou
- Department of Pathology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xiangxiang Tao
- Department of Pathology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Entao Sun
- Department of Health Inspection and Quarantine, School of Laboratory Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
11
|
Dual role of twist1 in cancer-associated fibroblasts and tumor cells promoted epithelial-mesenchymal transition of esophageal cancer. Exp Cell Res 2019; 375:41-50. [PMID: 30611739 DOI: 10.1016/j.yexcr.2019.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 12/26/2018] [Accepted: 01/02/2019] [Indexed: 02/07/2023]
Abstract
Cancer-associated fibroblasts (CAFs) play critical roles in tumor progression. However, the role and mechanism underlying CAFs in esophageal cancer (EC) remain unclear. In this study, primary CAFs and normal esophageal fibroblasts (NOFs) were isolated and characterized by immunofluorescence, qRT-PCR and western blot. Clinical significance of twist1 in CAFs were evaluated by immunohistochemistry assay. Conditioned medium (CM) was collected from CAFs to evaluate the influence on epithelial-mesenchymal transition (EMT) of EC cells. EC cells were mixed with CAFs and subcutaneously injected into nude mice to assess the in vivo tumor growth. As the result, twist1 was overexpressed in CAFs compared with NOFs and exhibited adverse prognostic significance. In CAFs, twist1 promoted the expression and secretion of CXCL12. In EC cells, activated CXCL12/CXCR4 signaling promoted the EMT process through ERK/AKT - twist1 - MMP1/E-cadherin pathway. In addition, knockdown of twist1 in CAFs also suppressed in vivo tumor growth. In conclusion, our results revealed a dual role of twist1 in CAFs and EC cells to promote the EMT process.
Collapse
|
12
|
Li F, Xu Y, Liu B, Singh PK, Zhao W, Jin J, Han G, Scott AW, Dong X, Huo L, Ma L, Pizzi MP, Wang Y, Li Y, Harada K, Xie M, Skinner HD, Ding S, Wang L, Krishnan S, Johnson RL, Song S, Ajani JA. YAP1-Mediated CDK6 Activation Confers Radiation Resistance in Esophageal Cancer - Rationale for the Combination of YAP1 and CDK4/6 Inhibitors in Esophageal Cancer. Clin Cancer Res 2018; 25:2264-2277. [PMID: 30563933 DOI: 10.1158/1078-0432.ccr-18-1029] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/16/2018] [Accepted: 12/14/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Esophageal cancer is a lethal disease that is often resistant to therapy. Alterations of YAP1 and CDK6 are frequent in esophageal cancer. Deregulation of both molecules may be responsible for therapy resistance. EXPERIMENTAL DESIGN Expressions of YAP1 and CDK6 were examined in esophageal cancer cells and tissues using immunoblotting and immunohistochemistry. YAP1 expression was induced in esophageal cancer cells to examine YAP1-mediated CDK6 activation and its association with radiation resistance. Pharmacologic and genetic inhibitions of YAP1 and CDK6 were performed to dissect the mechanisms and assess the antitumor effects in vitro and in vivo. RESULTS YAP1 expression was positively associated with CDK6 expression in resistant esophageal cancer tissues and cell lines. YAP1 overexpression upregulated CDK6 expression and transcription, and promoted radiation resistance, whereas treatment with the YAP1 inhibitor, CA3, strongly suppressed YAP1 and CDK6 overexpression, reduced Rb phosphorylation, as well as sensitized radiation-resistant/YAP1high esophageal cancer cells to irradiation. CDK4/6 inhibitor, LEE011, and knock down of CDK6 dramatically inhibited expression of YAP1 and sensitized resistant esophageal cancer cells to irradiation indicating a positive feed-forward regulation of YAP1 by CDK6. In addition, suppression of both the YAP1 and CDK6 pathways by the combination of CA3 and LEE011 significantly reduced esophageal cancer cell growth and cancer stem cell population (ALDH1 + and CD133 + ), sensitized cells to irradiation, and showed a strong antitumor effect in vivo against radiation-resistant esophageal cancer cells. CONCLUSIONS Our results document that a positive crosstalk between the YAP1 and CDK6 pathways plays an important role in conferring radiation resistance to esophageal cancer cells. Targeting both YAP1 and CDK6 pathways could be a novel therapeutic strategy to overcome resistance in esophageal cancer.
Collapse
Affiliation(s)
- Fan Li
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas.,Department of General Surgery, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan Xu
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Bovey Liu
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Pankaj Kumar Singh
- Department of Radiation Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Wei Zhao
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Guangchun Han
- Department of Genomic Medicine, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Ailing W Scott
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Xiaochuan Dong
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Longfei Huo
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Lang Ma
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Melissa Pool Pizzi
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Ying Wang
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Yuan Li
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Kazuto Harada
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Min Xie
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Heath D Skinner
- Department of Radiation Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Sheng Ding
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Linghua Wang
- Department of Genomic Medicine, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Sunil Krishnan
- Department of Radiation Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Randy L Johnson
- Department of Cancer Biology, U.T.MD. Anderson Cancer Center, Houston, Texas
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas.
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, U.T.MD. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
13
|
Sawada A, Fujiwara Y, Nagami Y, Tanaka F, Yamagami H, Tanigawa T, Shiba M, Tominaga K, Watanabe T, Gi M, Wanibuchi H, Arakawa T. Alteration of Esophageal Microbiome by Antibiotic Treatment Does Not Affect Incidence of Rat Esophageal Adenocarcinoma. Dig Dis Sci 2016; 61:3161-3168. [PMID: 27461059 DOI: 10.1007/s10620-016-4263-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/20/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent studies suggest that chronic inflammation-associated cancer is relevant to microbiome. Esophageal adenocarcinoma arises from an inflammatory condition called Barrett's esophagus, which is caused by gastroesophageal reflux. We hypothesized that esophageal microbiome plays a role in carcinogenesis of esophageal adenocarcinoma. AIM We investigated whether alteration of microbiome using antibiotics affects the development of esophageal adenocarcinoma in a rat model. METHODS Seven-week-old male Wistar rats which had undergone esophagojejunostomy were divided into control (n = 21) and antibiotic groups (n = 22) at 21 weeks after surgery. Control animals were given drinking water, while the other group was given penicillin G and streptomycin in drinking water until rats were killed at 40 weeks after operation. Incidence rates of Barrett's esophagus and adenocarcinoma in each group were evaluated by histological analysis. DNA was extracted from a portion of the distal esophagus, and the microbiome was investigated using terminal restriction fragment length polymorphism (T-RFLP) analysis. RESULTS All rats in both groups developed Barrett's esophagus. Incidence of esophageal adenocarcinoma was similar between both groups with a trend to reduced incidence in the antibiotics group (89 % in the control group, 71 % in the antibiotics group, P = 0.365). T-RFLP analysis showed that esophageal microbiome was different between two groups such as the proportion of Lactobacillales was lower in the antibiotics group and Clostridium cluster XIVa and XVIII was higher in the antibiotics group. CONCLUSIONS Alteration of microbiome does not affect the incidence of esophageal adenocarcinoma. Microbiome may not contribute to the development of esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Akinari Sawada
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan.
- SAMURAI GI Research Center, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan.
| | - Yasuaki Nagami
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Fumio Tanaka
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
- Department of Premier Preventive Medicine, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Hirokazu Yamagami
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Tetsuya Tanigawa
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
- SAMURAI GI Research Center, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Masatsugu Shiba
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Kazunari Tominaga
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
- SAMURAI GI Research Center, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Toshio Watanabe
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
- SAMURAI GI Research Center, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Min Gi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | - Tetsuo Arakawa
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
- SAMURAI GI Research Center, Osaka City University Graduate School of Medicine, 1-4-7 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| |
Collapse
|
14
|
Kong J, Whelan KA, Laczkó D, Dang B, Caro Monroig A, Soroush A, Falcone J, Amaravadi RK, Rustgi AK, Ginsberg GG, Falk GW, Nakagawa H, Lynch JP. Autophagy levels are elevated in barrett's esophagus and promote cell survival from acid and oxidative stress. Mol Carcinog 2016; 55:1526-1541. [PMID: 26373456 PMCID: PMC4794420 DOI: 10.1002/mc.22406] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 08/11/2015] [Accepted: 08/17/2015] [Indexed: 12/17/2022]
Abstract
Autophagy is a highly conserved mechanism that is activated during cellular stress. We hypothesized that autophagy may be induced by acid reflux, which causes injury, and inflammation, and therefore, contributes to the pathogenesis of Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC). Currently, the role of autophagy in BE and EAC is poorly studied. We quantitatively define autophagy levels in human BE cell lines, a transgenic mouse model of BE, and human BE, and EAC biopsies. Human non-dysplastic BE had the highest basal number of autophagic vesicles (AVs), while AVs were reduced in normal squamous cells and dysplastic BE cells, and nearly absent in EAC. To demonstrate a functional role for autophagy in BE pathogenesis, normal squamous (STR), non-dysplastic BE (CPA), dysplastic BE (CPD), and EAC (OE19) cell lines were exposed to an acid pulse (pH 3.5) followed by incubation in the presence or absence of chloroquine, an autophagy inhibitor. Acid exposure increased reactive oxygen species (ROS) levels in STR and CPA cells. Chloroquine alone had a small impact on intracellular ROS or cell survival. However, combination of chloroquine with the acid pulse resulted in a significant increase in ROS levels at 6 h in STR and CPA cells, and increased cell death in all cell lines. These findings establish increased numbers of AVs in human BE compared to normal squamous or EAC, and suggest that autophagy functions to improve cell survival after acid reflux injury. Autophagy may thus play a critical role in BE pathogenesis and progression. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jianping Kong
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kelly A Whelan
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dorottya Laczkó
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brendan Dang
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Angeliz Caro Monroig
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ali Soroush
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Falcone
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ravi K Amaravadi
- Division of Hematology/Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Medicine, and the Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anil K Rustgi
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gregory G Ginsberg
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gary W Falk
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hiroshi Nakagawa
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John P Lynch
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
15
|
Paulson TG. Studying Cancer Evolution in Barrett’s Esophagus and Esophageal Adenocarcinoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 908:213-36. [DOI: 10.1007/978-3-319-41388-4_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Song S, Honjo S, Jin J, Chang SS, Scott AW, Chen Q, Kalhor N, Correa AM, Hofstetter WL, Albarracin CT, Wu TT, Johnson RL, Hung MC, Ajani JA. The Hippo Coactivator YAP1 Mediates EGFR Overexpression and Confers Chemoresistance in Esophageal Cancer. Clin Cancer Res 2015; 21:2580-90. [PMID: 25739674 DOI: 10.1158/1078-0432.ccr-14-2191] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/13/2015] [Indexed: 01/12/2023]
Abstract
PURPOSE Esophageal cancer is an aggressive malignancy and often resistant to therapy. Overexpression of EGFR has been associated with poor prognosis of patients with esophageal cancer. However, clinical trials using EGFR inhibitors have not provided benefit for patients with esophageal cancer. Failure of EGFR inhibition may be due to crosstalk with other oncogenic pathways. EXPERIMENTAL DESIGN In this study, expression of YAP1 and EGFR were examined in EAC-resistant tumor tissues versus sensitive tissues by IHC. Western blot analysis, immunofluorescence, real-time PCR, promoter analysis, site-directed mutagenesis, and in vitro and in vivo functional assays were performed to elucidate the YAP1-mediated EGFR expression and transcription and the relationship with chemoresistance in esophageal cancer. RESULTS We demonstrate that Hippo pathway coactivator YAP1 can induce EGFR expression and transcription in multiple cell systems. Both YAP1 and EGFR are overexpressed in resistant esophageal cancer tissues compared with sensitive esophageal cancer tissues. Furthermore, we found that YAP1 increases EGFR expression at the level of transcription requiring an intact TEAD-binding site in the EGFR promoter. Most importantly, exogenous induction of YAP1 induces resistance to 5-fluorouracil and docetaxcel, whereas knockdown of YAP1 sensitizes esophageal cancer cells to these cytotoxics. Verteporfin, a YAP1 inhibitor, effectively inhibits both YAP1 and EGFR expression and sensitizes cells to cytotoxics. CONCLUSIONS Our data provide evidence that YAP1 upregulation of EGFR plays an important role in conferring therapy resistance in esophageal cancer cells. Targeting YAP1-EGFR axis may be more efficacious than targeting EGFR alone in esophageal cancer.
Collapse
Affiliation(s)
- Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Soichiro Honjo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shih-Shin Chang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ailing W Scott
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qiongrong Chen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neda Kalhor
- Department of Pathology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Arlene M Correa
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wayne L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Constance T Albarracin
- Department of Pathology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Tsung-Teh Wu
- Department of Pathology, Mayo Clinic, Rochester, Minnesota
| | - Randy L Johnson
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
17
|
Baruah A, Buttar N, Chandra R, Chen X, Clemons NJ, Compare D, El-Rifai W, Gu J, Houchen CW, Koh SY, Li W, Nardone G, Phillips WA, Sharma A, Singh I, Upton MP, Vega KJ, Wu X. Translational research on Barrett's esophagus. Ann N Y Acad Sci 2014; 1325:170-86. [DOI: 10.1111/nyas.12531] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Anushka Baruah
- Division of Gastroenterology & Hepatology; Mayo Clinic College of Medicine; Rochester Minnesota
| | - Navtej Buttar
- Division of Gastroenterology & Hepatology; Mayo Clinic College of Medicine; Rochester Minnesota
| | - Raghav Chandra
- Division of Gastroenterology & Hepatology; Mayo Clinic College of Medicine; Rochester Minnesota
| | - Xiaoxin Chen
- Cancer Research Program, JLC-BBRI; North Carolina Central University; Durham North Carolina
- Center for Esophageal Disease and Swallowing, Division of Gastroenterology and Hepatology, Department of Medicine; University of North Carolina at Chapel Hill; Chapel Hill North Carolina
| | - Nicholas J. Clemons
- Surgical Oncology Research Laboratory; Peter MacCallum Cancer Centre; East Melbourne Australia
- Department of Surgery (St. Vincent's Hospital); University of Melbourne; Melbourne Australia
- Sir Peter MacCallum Department of Oncology; University of Melbourne; Melbourne Australia
| | - Debora Compare
- Department of Clinical Medicine and Surgery, Gastroenterology Unit; University Federico II; Naples Italy
| | - Wael El-Rifai
- Surgical Oncology Research; Vanderbilt University Medical Center; Nashville Tennessee
| | - Jian Gu
- Department of Epidemiology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Courtney W. Houchen
- Division of Digestive Diseases and Nutrition; University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma
| | - Shze Yung Koh
- Surgical Oncology Research Laboratory; Peter MacCallum Cancer Centre; East Melbourne Australia
- Department of Surgery (St. Vincent's Hospital); University of Melbourne; Melbourne Australia
| | - Wenbo Li
- Cancer Research Program, JLC-BBRI; North Carolina Central University; Durham North Carolina
- Department of Gastroenterology; General Hospital of Jinan Military Command; Jinan China
| | - Gerardo Nardone
- Department of Clinical Medicine and Surgery, Gastroenterology Unit; University Federico II; Naples Italy
| | - Wayne A. Phillips
- Surgical Oncology Research Laboratory; Peter MacCallum Cancer Centre; East Melbourne Australia
- Department of Surgery (St. Vincent's Hospital); University of Melbourne; Melbourne Australia
- Sir Peter MacCallum Department of Oncology; University of Melbourne; Melbourne Australia
| | - Anamay Sharma
- Division of Gastroenterology & Hepatology; Mayo Clinic College of Medicine; Rochester Minnesota
| | - Ishtpreet Singh
- Division of Gastroenterology & Hepatology; Mayo Clinic College of Medicine; Rochester Minnesota
| | - Melissa P. Upton
- Department of Pathology; University of Washington; Seattle Washington
| | - Kenneth J. Vega
- Division of Digestive Diseases and Nutrition; University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma
| | - Xifeng Wu
- Department of Epidemiology; The University of Texas MD Anderson Cancer Center; Houston Texas
| |
Collapse
|
18
|
Wang Z, Da Silva TG, Jin K, Han X, Ranganathan P, Zhu X, Sanchez-Mejias A, Bai F, Li B, Fei DL, Weaver K, Carpio RVD, Moscowitz AE, Koshenkov VP, Sanchez L, Sparling L, Pei XH, Franceschi D, Ribeiro A, Robbins DJ, Livingstone AS, Capobianco AJ. Notch signaling drives stemness and tumorigenicity of esophageal adenocarcinoma. Cancer Res 2014; 74:6364-74. [PMID: 25164006 DOI: 10.1158/0008-5472.can-14-2051] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Esophageal adenocarcinoma ranks sixth in cancer mortality in the world and its incidence has risen dramatically in the Western population over the last decades. Data presented herein strongly suggest that Notch signaling is critical for esophageal adenocarcinoma and underlies resistance to chemotherapy. We present evidence that Notch signaling drives a cancer stem cell phenotype by regulating genes that establish stemness. Using patient-derived xenograft models, we demonstrate that inhibition of Notch by gamma-secretase inhibitors (GSI) is efficacious in downsizing tumor growth. Moreover, we demonstrate that Notch activity in a patient's ultrasound-assisted endoscopic-derived biopsy might predict outcome to chemotherapy. Therefore, this study provides a proof of concept that inhibition of Notch activity will have efficacy in treating esophageal adenocarcinoma, offering a rationale to lay the foundation for a clinical trial to evaluate the efficacy of GSI in esophageal adenocarcinoma treatment.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Thiago G Da Silva
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Ke Jin
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Xiaoqing Han
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Prathibha Ranganathan
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Xiaoxia Zhu
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Avencia Sanchez-Mejias
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Feng Bai
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Bin Li
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Dennis Liang Fei
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Kelly Weaver
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Rodrigo Vasquez-Del Carpio
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Anna E Moscowitz
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Vadim P Koshenkov
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Lilly Sanchez
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Lynne Sparling
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Xin-Hai Pei
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Dido Franceschi
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Afonso Ribeiro
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - David J Robbins
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Alan S Livingstone
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Anthony J Capobianco
- Molecular Oncology Program, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida. Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.
| |
Collapse
|
19
|
Kosovec JE, Zaidi AH, Komatsu Y, Kasi PM, Cothron K, Thompson DV, Lynch E, Jobe BA. Establishing magnetic resonance imaging as an accurate and reliable tool to diagnose and monitor esophageal cancer in a rat model. PLoS One 2014; 9:e93694. [PMID: 24705451 PMCID: PMC3976303 DOI: 10.1371/journal.pone.0093694] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/08/2014] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To assess the reliability of magnetic resonance imaging (MRI) for detection of esophageal cancer in the Levrat model of end-to-side esophagojejunostomy. BACKGROUND The Levrat model has proven utility in terms of its ability to replicate Barrett's carcinogenesis by inducing gastroduodenoesophageal reflux (GDER). Due to lack of data on the utility of non-invasive methods for detection of esophageal cancer, treatment efficacy studies have been limited, as adenocarcinoma histology has only been validated post-mortem. It would therefore be of great value if the validity and reliability of MRI could be established in this setting. METHODS Chronic GDER reflux was induced in 19 male Sprague-Dawley rats using the modified Levrat model. At 40 weeks post-surgery, all animals underwent endoscopy, MRI scanning, and post-mortem histological analysis of the esophagus and anastomosis. With post-mortem histology serving as the gold standard, assessment of presence of esophageal cancer was made by five esophageal specialists and five radiologists on endoscopy and MRI, respectively. RESULTS The accuracy of MRI and endoscopic analysis to correctly identify cancer vs. no cancer was 85.3% and 50.5%, respectively. ROC curves demonstrated that MRI rating had an AUC of 0.966 (p<0.001) and endoscopy rating had an AUC of 0.534 (p = 0.804). The sensitivity and specificity of MRI for identifying cancer vs. no-cancer was 89.1% and 80% respectively, as compared to 45.5% and 57.5% for endoscopy. False positive rates of MRI and endoscopy were 20% and 42.5%, respectively. CONCLUSIONS MRI is a more reliable diagnostic method than endoscopy in the Levrat model. The non-invasiveness of the tool and its potential to volumetrically quantify the size and number of tumors likely makes it even more useful in evaluating novel agents and their efficacy in treatment studies of esophageal cancer.
Collapse
Affiliation(s)
- Juliann E. Kosovec
- Institute for the Treatment of Esophageal and Thoracic Disease, Allegheny Health Network, Pittsburgh, Pennsylvania, United States of America
| | - Ali H. Zaidi
- Institute for the Treatment of Esophageal and Thoracic Disease, Allegheny Health Network, Pittsburgh, Pennsylvania, United States of America
| | - Yoshihiro Komatsu
- Institute for the Treatment of Esophageal and Thoracic Disease, Allegheny Health Network, Pittsburgh, Pennsylvania, United States of America
| | - Pashtoon M. Kasi
- International Scholars Program, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Kyle Cothron
- Department of Radiology, Allegheny Health Network, Pittsburgh, Pennsylvania, United States of America
| | - Diane V. Thompson
- Department of Medicine, Allegheny Health Network, Pittsburgh, Pennsylvania, United States of America
| | - Edward Lynch
- Department of Pathology, Allegheny Health Network, Pittsburgh, Pennsylvania, United States of America
| | - Blair A. Jobe
- Institute for the Treatment of Esophageal and Thoracic Disease, Allegheny Health Network, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
20
|
Pandilla R, Kotapalli V, Gowrishankar S, Chigurupati M, Patnaik S, Uppin S, Rao S, Kalidindi N, Regulagadda S, Sundaram C, Srinivasulu M, Vasala A, Bashyam MD. Distinct genetic aberrations in oesophageal adeno and squamous carcinoma. Eur J Clin Invest 2013; 43:1233-9. [PMID: 24102414 DOI: 10.1111/eci.12163] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 08/20/2013] [Indexed: 01/07/2023]
Abstract
BACKGROUND The two main oesophageal cancer subtypes namely adenocarcinoma and squamous cell carcinoma exhibit interesting clinical, pathological and geographical variations with the former being more common in the West and the latter in Asia. MATERIALS AND METHODS We evaluated status of p53, EGFR, Wnt and HPV in addition to microsatellite instability and loss of heterozygosity of several chromosomal loci in the two oesophageal cancer subtypes from India. The comparative analysis was extended to two oesophageal adenosquamous mixed cancer samples. RESULTS Our results reveal a high frequency of EGFR overexpression in ESCC as against EAC, while Wnt activation was a significantly more common event in EAC as against ESCC. Frequencies of p53 perturbations were not significantly different in the two subtypes. Interestingly, the EGFR and Wnt status in adenocarcinoma and squamous components of the two oesophageal adenosquamous cancer samples were identical to primary tumours. In addition, no common molecular aberration (including instability and loss of heterozygosity) in several microsatellites was detected in DNA isolated from the two components in both adenosquamous cancer samples. CONCLUSIONS Our results reveal the presence of distinct aberrations in oesophageal adenocarcinoma and squamous cell carcinoma which are replicated in the respective components of adenosquamous cancers. The study therefore suggests perhaps an independent origin of the two components of oesophageal adenosquamous mixed cancer.
Collapse
Affiliation(s)
- Ramaswamy Pandilla
- Laboratory of Molecular Oncology Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Cancer stem cell radioresistance and enrichment: where frontline radiation therapy may fail in lung and esophageal cancers. Cancers (Basel) 2013; 3:1232-52. [PMID: 21603589 PMCID: PMC3095975 DOI: 10.3390/cancers3011232] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Many studies have highlighted the role cancer stem cells (CSC) play in the development and progression of various types of cancer including lung and esophageal cancer. More recently, it has been proposed that the presence of CSCs affects treatment efficacy and patient prognosis. In reviewing this new area of cancer biology, we will give an overview of the current literature regarding lung and esophageal CSCs and radioresistance of CSC, and discuss the potential therapeutic applications of these findings.
Collapse
|
22
|
Bajpai M, Kessel R, Bhagat T, Nischal S, Yu Y, Verma A, Das KM. High resolution integrative analysis reveals widespread genetic and epigenetic changes after chronic in-vitro acid and bile exposure in Barrett's epithelium cells. Genes Chromosomes Cancer 2013; 52:1123-32. [PMID: 24123713 DOI: 10.1002/gcc.22106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 12/12/2022] Open
Abstract
Barrett's epithelium (BE) is a premalignant condition resulting from chronic gastroesophageal reflux that may progress to esophageal adenocarcinoma (EAC). Early intervention holds promise in preventing BE progression. However, identification of high-risk BE patients remains challenging due to inadequate biomarkers for early diagnosis. We investigated the effect of prolonged chronic acid and bile exposure on transcriptome, methylome, and mutatome of cells in an in-vitro BE carcinogenesis (BEC) model. Twenty weeks acid and bile exposed cells from the BEC model (BEC20w) were compared with their naïve predecessors HiSeq Illumina based RNA sequencing was performed on RNA from both the cells for gene expression and mutational analysis. HELP Tagging Assay was performed for DNA methylation analysis. Ingenuity pathway, Gene Ontology, and KEGG PATHWAY analyses were then performed on datasets. Widespread aberrant genetic and epigenetic changes were observed in the BEC20w cells. Combinatorial analyses revealed 433 from a total of 863 downregulated genes had accompanying hypermethylation of promoters. Simultaneously, 690 genes from a total of 1,492 were upregulated with accompanying promoter hypomethylation. In addition, 763 mutations were identified on 637 genes. Ingenuity pathway analysis, Gene Ontology, and KEGG PATHWAY analyses associated the genetic and epigenetic changes in BEC20w cells with cellular and biological functions. Integration of high resolution comparative analyses of naïve BAR-T and BEC20w cells revealed striking genetic and epigenetic changes induced by chronic acid and bile exposure that may disrupt normal cellular functions and promote carcinogenesis. This novel study reveals several potential targets for future biomarkers and therapeutic development.
Collapse
Affiliation(s)
- Manisha Bajpai
- Division of Gastroenterology and Hepatology, Department of Medicine, RUTGERS Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | | | | | | | | | | | | |
Collapse
|
23
|
Lisovsky M, Srivastava A. Barrett Esophagus: Evolving Concepts in Diagnosis and Neoplastic Progression. Surg Pathol Clin 2013; 6:475-96. [PMID: 26839097 DOI: 10.1016/j.path.2013.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Surgical pathologists need to answer 2 questions when evaluating biopsies from the distal esophagus or gastroesophageal junction in patients with a history of gastroesophageal reflux disease: Are the findings consistent with Barrett esophagus? and Is there any evidence of dysplasia? Pathologists should be well informed about the controversy around the definition of Barrett esophagus and the common pitfalls that lead to a false-positive diagnosis of Barrett esophagus or Barrett esophagus-associated dysplasia. A concise description of distinct morphologic types of dysplasia in Barrett esophagus and a summary of recent data on the natural history of BE are provided in this review.
Collapse
Affiliation(s)
- Mikhail Lisovsky
- Department of Pathology, Dartmouth Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| | - Amitabh Srivastava
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Gibson MK, Zaidi AH, Davison JM, Sanz AF, Hough B, Komatsu Y, Kosovec JE, Bhatt A, Malhotra U, Foxwell T, Rotoloni CL, Hoppo T, Jobe BA. Prevention of Barrett esophagus and esophageal adenocarcinoma by smoothened inhibitor in a rat model of gastroesophageal reflux disease. Ann Surg 2013; 258:82-8. [PMID: 23108119 DOI: 10.1097/sla.0b013e318270500d] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Activated hedgehog (Hh) pathway is associated with development of both Barrett esophagus (BE) and esophageal adenocarcinoma (EAC). We hypothesize that blockade of the Hh pathway with smoothened (Smo) inhibitor can prevent the development of BE/EAC in the Levrat model, in which induced gastroduodenoesophageal reflux (GDER) leads to esophageal carcinogenesis. METHODS GDER was induced in 6- to 8-week-old male Sprague-Dawley rats. The Smo inhibitor (10 mg/kg/d) was given orally on postoperative weeks 10 to 16, 18 to 22, and 24 to 28, and rats were killed on week 28. The primary outcome measure was the incidence of BE and EAC. To examine potential therapeutic effects of Smo inhibition on tumor tissue, semiquantitative immunohistochemistry for Ki-67 and caspase 3 was performed. In treated animals that developed cancer, gene expression was analyzed. RESULTS Thirty-eight of 48 controls and 32 of 46 treated animals survived to 28 weeks. messenger ribonucleic acid (mRNA) expression of Indian Hh, a ligand of transmembrane receptor patched 1, was 184× higher in BE and 99× higher in EAC compared with normal esophageal tissue (P = 0.0239 and P = 0.0004, respectively). Compared with controls, the incidence of BE and EAC was decreased in treated animals by 35.7% (relative risk reduction, 36%; P = 0.0015) and 36% (relative risk reduction, 62%; P = 0.0033), respectively. Compared with untreated EAC, Ki-67 was downregulated (P = 0.04) and cleaved caspase 3 was no different in treated EAC (P = 0.398). Of the 84 well-known genes involved in cancer drug resistance, 50 were dysregulated in treated EAC (P < 0.05 for each gene). CONCLUSIONS Smo inhibitor prevents the development of BE and EAC in an in vivo model of GDER.
Collapse
Affiliation(s)
- Michael K Gibson
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Fassan M, Volinia S, Palatini J, Pizzi M, Fernandez-Cymering C, Balistreri M, Realdon S, Battaglia G, Souza R, Odze RD, Zaninotto G, Croce CM, Rugge Md Facg M. MicroRNA Expression Profiling in the Histological Subtypes of Barrett's Metaplasia. Clin Transl Gastroenterol 2013; 4:e34. [PMID: 23677165 PMCID: PMC3671360 DOI: 10.1038/ctg.2013.5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES: The histological definition of Barrett's esophagus (BE) is debated, particularly regarding the phenotype of its metaplastic columnar epithelium. Histologically proven intestinal metaplasia (IM) was the sine qua non condition for a diagnosis of BE but, more recently, non-intestinalized (i.e., cardiac gastric-type; GM) columnar metaplasia has been re-included in the spectrum of Barrett's histology. MicroRNAs modulate cell commitment, and are also reportedly dysregulated in Barrett's carcinogenesis. This study investigates miRNA expression in the histological spectrum of esophageal columnar metaplastic changes, specifically addressing the biological profile of GM vs. IM. METHODS: A study was performed to discover microRNA microarray in 30 matching mucosa samples obtained from 10 consecutive BE patients; for each patient, biopsy tissue samples were obtained from squamous, GM and intestinalized epithelium. Microarray findings were further validated by qRT-PCR analysis in another bioptic series of 75 mucosa samples. RESULTS: MicroRNA profiling consistently disclosed metaplasia-specific microRNA signatures. Six microRNAs were significantly dysregulated across the histological phenotypes considered; five of them (two overexpressed (hsa-miR-192; -miR-215) and three under-expressed (hsa-miR-18a* -miR-203, and -miR-205)) were progressively dysregulated in the phenotypic sequence from squamous to gastric-type, to intestinal-type mucosa samples. CONCLUSIONS: A consistent microRNA expression signature underlies both gastric- and intestinal-type esophageal metaplasia. The pattern of microRNA dysregulation suggests that GM may further progress to IM. The clinico-pathological implications of these molecular profiles prompt further study on the “personalized” cancer risk associated with each of these metaplastic transformations.
Collapse
Affiliation(s)
- Matteo Fassan
- 1] Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, University of Padua, Padua, Italy [2] Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), General Oncology Unit, University of Padua, Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Song S, Maru DM, Ajani JA, Chan CH, Honjo S, Lin HK, Correa A, Hofstetter WL, Davila M, Stroehlein J, Mishra L. Loss of TGF-β adaptor β2SP activates notch signaling and SOX9 expression in esophageal adenocarcinoma. Cancer Res 2013; 73:2159-69. [PMID: 23536563 DOI: 10.1158/0008-5472.can-12-1962] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TGF-β and Notch signaling pathways play important roles in regulating self-renewal of stem cells and gastrointestinal carcinogenesis. Loss of TGF-β signaling components activates Notch signaling in esophageal adenocarcinoma, but the basis for this effect has been unclear. Here we report that loss of TGF-β adapter β2SP (SPNB2) activates Notch signaling and its target SOX9 in primary fibroblasts or esophageal adenocarcinoma cells. Expression of the stem cell marker SOX9 was markedly higher in esophageal adenocarcinoma tumor tissues than normal tissues, and its higher nuclear staining in tumors correlated with poorer survival and lymph node invasion in esophageal adenocarcinoma patients. Downregulation of β2SP by lentivirus short hairpin RNA increased SOX9 transcription and expression, enhancing nuclear localization for both active Notch1 (intracellular Notch1, ICN1) and SOX9. In contrast, reintroduction into esophageal adenocarcinoma cells of β2SP and a dominant-negative mutant of the Notch coactivator mastermind-like (dnMAN) decreased SOX9 promoter activity. Tumor sphere formation and invasive capacity in vitro and tumor growth in vivo were increased in β2SP-silenced esophageal adenocarcinoma cells. Conversely, SOX9 silencing rescued the phenotype of esophageal adenocarcinoma cells with loss of β2SP. Interaction between Smad3 and ICN1 via Smad3 MH1 domain was also observed, with loss of β2SP increasing the binding between these proteins, inducing expression of Notch targets SOX9 and C-MYC, and decreasing expression of TGF-β targets p21(CDKN1A), p27 (CDKN1B), and E-cadherin. Taken together, our findings suggest that loss of β2SP switches TGF-β signaling from tumor suppression to tumor promotion by engaging Notch signaling and activating SOX9.
Collapse
Affiliation(s)
- Shumei Song
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Barrett's esophagus (BE), a well-known complication of gastroesophageal reflux disease (GERD), constitutes a precancerous condition for adenocarcinoma of the distal esophagus. The so-called Barrett's carcinoma shows increasing incidences in countries of the western hemisphere; new data, however, indicate that the rise in incidence is not quite as dramatic as previously assumed. The definition of BE is currently changing: despite good reasons for a purely endoscopic definition of BE, goblet cells are still mandatory for this diagnosis in Germany and the USA. Dysplastic changes in the epithelium are the most important risk factor for the development of Barrett's adenocarcinoma and recently dysplasia was subclassified into a more frequent adenomatous (intestinal) and a non-adenomatous (gastric-foveolar) types. The gold standard for diagnosing dysplasia is still H&E staining. The histological diagnosis of dysplasia is still encumbered by a significant interobserver variability, especially regarding the differentiation between low grade dysplasia and inflammatory/reactive changes and the discrimination between high grade dysplasia and adenocarcinoma. Current data, however, show much higher interobserver agreement in endoscopic resection specimens than in biopsies. Nevertheless, the histological diagnosis of dysplasia should be corroborated by an external second opinion because of its clinical consequences. In endoscopic resections of early Barrett's adenocarcinoma, the pathological report has to include a risk stratification for the likelihood of lymphogenic metastases.
Collapse
Affiliation(s)
- G B Baretton
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.
| | | |
Collapse
|
28
|
Notch signaling pathway and Cdx2 expression in the development of Barrett's esophagus. J Transl Med 2012; 92:896-909. [PMID: 22449796 DOI: 10.1038/labinvest.2012.56] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cdx2 expression in esophageal stem cells induced by reflux bile acids may be an important factor for development of Barrett's esophagus, whereas Notch signaling is a molecular signaling pathway that plays an important role in the determination of cell differentiation. ATOH1 (a factor associated with Notch signaling) plays an important role in differentiation of stem cells into goblet cells. However, the relationship between the Notch signaling pathway and Cdx2 expression in the development of Barrett's esophagus has not been explored. The aim of this study was to investigate the interrelationship between Notch signaling and Cdx2 in esophageal epithelial cells. The expressions of Cdx2, MUC2, and intracellular signaling molecules related to Notch signaling (Notch1, Hes1, and ATOH1) were examined using real-time polymerase chain reaction (PCR) and immunohistochemical staining with biopsy specimens obtained from esophageal intestinal metaplasia (IM) with goblet cells (IM⁺) and columnar epithelium not accompanied by goblet cells (IM⁻). For in vitro experiments, we employed human esophageal epithelial cell lines (OE33, OE19, and Het-1A). After forced Cdx2 expression by applying a Cdx2 expression vector to the cells, changes in the expressions of Notch1, Hes1, ATOH1, Cdx2, and MUC2 were analyzed by real-time PCR and western blot analysis. Changes in expressions of Notch1, Hes1, ATOH1, Cdx2, and MUC2 in cells were analyzed following stimulation with bile acids in the presence or absence of Cdx2 blocking with Cdx2-siRNA. Suppressed Hes1 and enhanced ATOH1 and MUC2 expressions were identified in IM⁺ specimens. Forced expression of Cdx2 in cells suppressed Hes1, and enhanced ATOH1 and MUC2 expressions, whereas bile acids suppressed Hes1, and enhanced ATOH1, Cdx2, and MUC2 expressions. On the other hand, these effects were blocked by siRNA-based Cdx2 downregulation. Enhanced expression of Cdx2 by stimulation with bile acids may induce intestinal differentiation of esophageal columnar cells by interaction with the Notch signaling pathway.
Collapse
|
29
|
Pizzi M, Fassan M, Realdon S, Balistreri M, Battaglia G, Giacometti C, Zaninotto G, Zagonel V, De Boni M, Rugge M. Anterior gradient 2 profiling in Barrett columnar epithelia and adenocarcinoma. Hum Pathol 2012; 43:1839-44. [PMID: 22521076 DOI: 10.1016/j.humpath.2012.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Revised: 01/03/2012] [Accepted: 01/04/2012] [Indexed: 01/10/2023]
Abstract
Barrett esophagus is the precancerous lesion leading to Barrett adenocarcinoma. The natural history of Barrett metaplasia and its neoplastic progression are still controversial. Anterior gradient 2 is up-regulated in both Barrett intestinal metaplasia and Barrett adenocarcinoma, but no information is available on anterior gradient 2 expression in the spectrum of the phenotypic changes occurring in the natural history of Barrett adenocarcinoma (Barrett esophagus cardiac-type metaplasia, Barrett esophagus intestinal metaplasia, low-grade intraepithelial neoplasia [formerly called low-grade dysplasia], and high-grade intraepithelial neoplasia [formerly called high-grade dysplasia]). Applying immunohistochemistry and reverse transcription and quantitative real-time polymerase chain reaction, this study addressed the role of anterior gradient 2 in Barrett carcinogenesis. Anterior gradient 2 expression was assessed semiquantitatively in 125 consecutive biopsy samples in the adenocarcinoma spectrum arising in Barrett esophagus (Barrett esophagus cardiac-type metaplasia, 25; Barrett esophagus intestinal metaplasia, 25; low-grade intraepithelial neoplasia, 25; high-grade intraepithelial neoplasia, 25; Barrett adenocarcinoma, 25). Additional biopsy samples of esophageal squamous mucosa (n=25) served as controls. Anterior gradient 2 messenger RNA expression was also tested (reverse transcription and quantitative real-time polymerase chain reaction) in a different series of 40 samples (esophageal squamous mucosa, 10; Barrett esophagus cardiac-type metaplasia, 10; Barrett esophagus intestinal metaplasia, 10; Barrett adenocarcinoma, 10). Anterior gradient 2 was never expressed in squamous esophageal epithelium but consistently overexpressed (to much the same degree) in the whole spectrum of Barrett disease (Barrett esophagus cardiac-type metaplasia, Barrett esophagus intestinal metaplasia, low-grade intraepithelial neoplasia, high-grade intraepithelial neoplasia, and Barrett adenocarcinoma). Anterior gradient 2 messenger RNA was expressed significantly more in Barrett esophagus cardiac-type metaplasia, Barrett esophagus intestinal metaplasia, and Barrett adenocarcinoma than in native squamous epithelium (P<.001), with no significant differences between the 3 groups. Anterior gradient 2 overexpression affects the whole spectrum of the metaplastic/neoplastic lesions involved in Barrett carcinogenesis. This study supports the biological similarity of the nonintestinal and intestinal types of Barrett metaplasia as precursors of Barrett adenocarcinoma.
Collapse
Affiliation(s)
- Marco Pizzi
- Department of Medical Diagnostic Sciences and Special Therapies, Surgical Pathology and Cytopathology Unit, University of Padova, 35100 Padova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Math1/Atoh1 contributes to intestinalization of esophageal keratinocytes by inducing the expression of Muc2 and Keratin-20. Dig Dis Sci 2012; 57:845-57. [PMID: 22147253 PMCID: PMC3407817 DOI: 10.1007/s10620-011-1998-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Accepted: 11/22/2011] [Indexed: 12/15/2022]
Abstract
BACKGROUND Esophageal intestinal metaplasia, also known as Barrett's esophagus, is the replacement of the normal epithelium with one that resembles the intestine morphologically. Generally, this includes intestinal mucin-secreting goblet cells. Barrett's esophagus is an important risk factor for adenocarcinoma development. In-vitro models for Barrett's esophagus have not, to date, focused on the induction of goblet cells in Barrett's epithelium. AIMS To explore the contribution of Math1/Atoh1 to induction of Barrett's esophagus and intestinal mucin-secreting goblet cells from normal human esophageal epithelium. METHODS We explored the level and pattern of Math1/Atoh1 mRNA and protein expression in human Barrett's esophagus. Then, using retroviral-mediated gene expression, we induced Math1 mRNA and protein expression in a human esophageal keratinocyte cell line. We evaluated the effects of this ectopic Math1 expression on cell proliferation and gene expression patterns in cells cultured under two-dimensional and three-dimensional tissue-engineering conditions. RESULTS Math1/Atoh1 mRNA and protein are detected in human Barrett's esophagus specimens, but the mRNA levels vary substantially. In the keratinocyte expression studies, we observed that Math1/Atoh1 ectopic expression significantly reduced cell proliferation and altered cell morphology. Moreover, Math1/Atoh1 expression is associated with a more intestinalized gene expression pattern that is distinct from that reported in after studies using other intestinal transcription factors. Most significantly, we observe the induction of the Barrett's esophagus markers Mucin-2 and Keratin-20 with Math1/Atoh1 expression. CONCLUSIONS We conclude that ectopic Math1/Atoh1 expression makes unique contributions to intestinalization of the esophageal epithelium in Barrett's esophagus.
Collapse
|
31
|
Cox2 and β-catenin/T-cell factor signaling intestinalize human esophageal keratinocytes when cultured under organotypic conditions. Neoplasia 2012; 13:792-805. [PMID: 21969813 DOI: 10.1593/neo.11788] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 07/14/2011] [Accepted: 07/18/2011] [Indexed: 12/12/2022] Open
Abstract
The incidence of esophageal adenocarcinoma (EAC) is rising in the United States. An important risk factor for EAC is the presence of Barrett esophagus (BE). BE is the replacement of normal squamous esophageal epithelium with a specialized columnar epithelium in response to chronic acid and bile reflux. However, the emergence of BE from squamous keratinocytes has not yet been demonstrated. Our research has focused on this. Wnt and cyclooxygenase 2 (Cox2) are two pathways whose activation has been associated with BE and progression to EAC, but their role has not been tested experimentally. To explore their contribution, we engineered a human esophageal keratinocyte cell line to express either a dominant-active Wnt effector CatCLef or a Cox2 complementary DNA. In a two-dimensional culture environment, Cox2 expression increases cell proliferation and migration, but neither transgene induces known BE markers. In contrast, when these cells were placed into three-dimensional organotypic culture conditions, we observed more profound effects. CatCLef-expressing cells were more proliferative, developed a thicker epithelium, and upregulated Notch signaling and several BE markers including NHE2. Cox2 expression also increased cell proliferation and induced a thicker epithelium. More importantly, we observed cysts form within the epithelium, filled with intestinal mucins including Muc5B and Muc17. This suggests that Cox2 expression in a three-dimensional culture environment induces a lineage of mucin-secreting cells and supports an important causal role for Cox2 in BE pathogenesis. We conclude that in vitro modeling of BE pathogenesis can be improved by enhancing Wnt signaling and Cox2 activity and using three-dimensional organotypic culture conditions.
Collapse
|
32
|
Fassan M, Ludwig K, Pizzi M, Castoro C, Guzzardo V, Balistreri M, Zaninotto G, Ruol A, Giacomelli L, Ancona E, Rugge M. Human epithelial growth factor receptor 2 (HER2) status in primary and metastatic esophagogastric junction adenocarcinomas. Hum Pathol 2012; 43:1206-12. [PMID: 22217477 DOI: 10.1016/j.humpath.2011.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 09/03/2011] [Accepted: 09/07/2011] [Indexed: 12/15/2022]
Abstract
Differences in human epithelial growth factor receptor 2 dysregulation in primary solid tumors and metastases may (at least partially) explain human epithelial growth factor receptor 2-targeted therapeutic inconsistencies. Human epithelial growth factor receptor 2 status was tested in a series of 47 radically treated consecutive esophagogastric junction adenocarcinomas (male/female, 38/9; mean age, 67.9 years) in both primary cancers and paired synchronous nodal metastases. None of the patients received neoadjuvant therapy. For each case, 2 nonadjacent tissue samples from primary esophagogastric junction adenocarcinoma and 2 different metastatic nodes were considered (188 tissue samples in all). Human epithelial growth factor receptor 2 status was assessed by immunohistochemistry (PATHWAY-HER2/neu [4B5]; Ventana Medical Systems, Milan, Italy) and dual chromogenic in situ hybridization (duoCISH; DAKO, Glostrup, Denmark). Immunohistochemistry staining scores were nil in 22 tumors (47%), 1 (21%) in 10, 2 (13%) in 6, and 3 (19%) in 9. Human epithelial growth factor receptor 2 gene amplification (25.5%) was associated with more differentiated phenotype (Fisher exact test, P = .039) and advanced tumor stage (Fisher exact test, P = .015). Significant agreement was observed between human epithelial growth factor receptor 2 protein expression (immunohistochemistry) and human epithelial growth factor receptor 2 gene's amplification (chromogenic in situ hybridization) (κ = 0.84, P < .001). Both immunohistochemistry and chromogenic in situ hybridization documented an excellent intratumor agreement in human epithelial growth factor receptor 2 status (κ = 0.75, P < .001; κ = 0.88, P < .001, respectively). Human epithelial growth factor receptor 2 status was comparable in primary versus metastatic nodal cancers by both immunohistochemistry and chromogenic in situ hybridization (Cohen Φ, both P < .001). In esophagogastric junction adenocarcinomas, human epithelial growth factor receptor 2 status (as assessed by immunohistochemistry and/or chromogenic in situ hybridization) is virtually unaffected by intratumor variability; it is consistent with findings in nodal metastases, and it reliably identifies patients with esophagogastric junction adenocarcinoma eligible for anti-human epithelial growth factor receptor 2 therapy.
Collapse
Affiliation(s)
- Matteo Fassan
- Department of Medical Diagnostic Sciences and Special Therapies (Pathology and Cytopathology Unit), University of Padova, 35100 Padova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Macke RA, Nason KS, Mukaisho KI, Hattori T, Fujimura T, Sasaki S, Oyama K, Miyashita T, Ohta T, Miwa K, Gibson MK, Zaidi A, Malhotra U, Atasoy A, Foxwell T, Jobe B. Barrett's esophagus and animal models. Ann N Y Acad Sci 2011; 1232:392-400. [PMID: 21950831 DOI: 10.1111/j.1749-6632.2011.06061.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The following on Barrett's esophagus (BE) and animal models contains commentaries on the factors of BE carcinogenesis; a duodenoesophageal reflux model; translation of targeted therapies for esophageal adenocarcinoma; and novel target regimens selected through a proteomics screen.
Collapse
Affiliation(s)
- Ryan A Macke
- Esophageal Cancer Program, University of Pittsburgh and University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wiseman EF, Ang YS. Risk factors for neoplastic progression in Barrett’s esophagus. World J Gastroenterol 2011; 17:3672-83. [PMID: 21990948 PMCID: PMC3181452 DOI: 10.3748/wjg.v17.i32.3672] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 10/11/2010] [Accepted: 10/18/2010] [Indexed: 02/06/2023] Open
Abstract
Barrett’s esophagus (BE) confers a significant increased risk for development of esophageal adenocarcinoma (EAC), with the pathogenesis appearing to progress through a “metaplasia-dysplasia-carcinoma” (MDC) sequence. Many of the genetic insults driving this MDC sequence have recently been characterized, providing targets for candidate biomarkers with potential clinical utility to stratify risk in individual patients. Many clinical risk factors have been investigated, and associations with a variety of genetic, specific gastrointestinal and other modifiable factors have been proposed in the literature. This review summarizes the current understanding of the mechanisms involved in neoplastic progression of BE to EAC and critically appraises the relative roles and contributions of these putative risk factors from the published evidence currently available.
Collapse
|
35
|
Mendelson J, Song S, Li Y, Maru DM, Mishra B, Davila M, Hofstetter WL, Mishra L. Dysfunctional transforming growth factor-β signaling with constitutively active Notch signaling in Barrett's esophageal adenocarcinoma. Cancer 2011; 117:3691-702. [PMID: 21305538 PMCID: PMC3236645 DOI: 10.1002/cncr.25861] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 11/02/2010] [Accepted: 11/09/2010] [Indexed: 02/03/2023]
Abstract
BACKGROUND Esophageal adenocarcinoma is often considered to arise from a clonal stem-like population of cells, which is potentially responsible for its poor prognosis. Transforming growth factor β (TGF-β) and Notch signaling pathways play important roles in regulating self-renewal of stem cells and cell-fate determination. Both pathways are frequently implicated in gastrointestinal carcinogenesis. However, their contributions to esophageal adenocarcinoma remain unclear. METHODS We evaluated TGF-β and Notch signaling components in normal esophagus, Barrett's esophagus, and adenocarcinoma tissues and cell lines via immunohistochemical analysis and immunoblotting; Hes-1 transcription was assayed using a Hes-1 luciferase reporter. RESULTS We observed loss of Smad4 (P<.05) and β2 spectrin (β2SP) (P<.01) in 5/10 Barrett's esophagus and 17/22 adenocarcinoma tissue sections. Concomitantly, dramatically raised levels of Notch signaling components Hes1 and Jagged1 occurred in adenocarcinoma tissues and cell lines compared with normal tissues. In normal esophagus, Oct3/4-positive cells are located in the basal layer (2-3 per cluster), representing a pool of progenitor cells. We observed an expansion of this pool of Oct3/4 positive cells in esophageal adenocarcinoma (15 per cluster). Furthermore, a panel of SOXs proteins documented for stem cell markers exhibit increased expression in tumor cells, indicating expansion of putative cancer stem cells. Finally, we observed growth inhibition in BE3 cells with a γ-secretase inhibitor, but not in SKGT-4 cells. Unlike SKGT-4 cells, BE3 cells have activated Notch signaling with disruption of TGF-β signaling. CONCLUSIONS Our findings demonstrated a potential therapeutic value for targeted therapy in esophageal adenocarcinoma in the setting of loss of β2SP/TGF-β with concomitant constitutively active Notch signaling.
Collapse
Affiliation(s)
| | - Shumei Song
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Li
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dipen M Maru
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bibhuti Mishra
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marta Davila
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wayne L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lopa Mishra
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Correspondence to: Lopa Mishra, MD, Del&Dennis McCarthy Distinguished Professor & Chair, Department of Gastroenterology, Hepatology and Nutrition, The University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, Tel: 713-794-3221, Fax: 713-745-1886,
| |
Collapse
|
36
|
Nandakumar V, Kelbauskas L, Johnson R, Meldrum D. Quantitative characterization of preneoplastic progression using single-cell computed tomography and three-dimensional karyometry. Cytometry A 2011; 79:25-34. [PMID: 21182180 DOI: 10.1002/cyto.a.20997] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The development of morphological biosignatures to precisely characterize preneoplastic progression necessitates high-resolution three-dimensional (3D) cell imagery and robust image processing algorithms. We report on the quantitative characterization of nuclear structure alterations associated with preneoplastic progression in human esophageal epithelial cells using single-cell optical tomography and fully automated 3D karyometry. We stained cultured cells with hematoxylin and generated 3D images of individual cells by mathematically reconstructing 500 projection images acquired using optical absorption tomographic imaging. For 3D karyometry, we developed novel, fully automated algorithms to robustly segment the cellular, nuclear, and subnuclear components in the acquired cell images, and computed 41 quantitative morphological descriptors from these segmented volumes. In addition, we developed algorithms to quantify the spatial distribution and texture of the nuclear DNA. We applied our methods to normal, metaplastic, and dysplastic human esophageal epithelial cell lines, analyzing 100 cells per line. The 3D karyometric descriptors elucidated quantitative differences in morphology and enabled robust discrimination between cell lines on the basis of extracted morphological features. The morphometric hallmarks of cancer progression such as increased nuclear size, elevated nuclear content, and anomalous chromatin texture and distribution correlated with this preneoplastic progression model, pointing to the clinical use of our method for early cancer detection.
Collapse
Affiliation(s)
- Vivek Nandakumar
- School of Electrical, Computer, and Energy Engineering, Arizona State University, Tempe, Arizona, USA
| | | | | | | |
Collapse
|
37
|
Fassan M, Volinia S, Palatini J, Pizzi M, Baffa R, De Bernard M, Battaglia G, Parente P, Croce CM, Zaninotto G, Ancona E, Rugge M. MicroRNA expression profiling in human Barrett's carcinogenesis. Int J Cancer 2011; 129:1661-70. [PMID: 21128279 DOI: 10.1002/ijc.25823] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 11/19/2010] [Indexed: 12/19/2022]
Abstract
Barrett's esophagus (BE) is characterized by the native stratified squamous epithelium (N) lining the esophagus being replaced by a columnar epithelium with intestinal differentiation (Barrett's mucosa; BM). BM is considered as the main risk factor for esophageal adenocarcinoma (Barrett's adenocarcinoma; BAc). MicroRNAs (miRNAs) are a class of small noncoding RNAs that control gene expression by targeting messenger RNAs and they are reportedly dysregulated in BM. To test the hypothesis that a specific miRNA expression signature characterizes BM development and progression, we performed miRNA microarray analysis comparing native esophageal mucosa with all the phenotypic lesions seen in the Barrett's carcinogenic process. Specimens were collected from 14 BE patients who had undergone esophagectomy, including: 14 with N, 14 with BM, 7 with low-grade intraepithelial neoplasia, 5 with high-grade intra-epithelial neoplasia and 11 with BAc. Microarray findings were further validated by quantitive real-time polymerase chain reaction and in situ hybridization analyses using a different series of consecutive cases (162 biopsy samples and 5 esophagectomies) of histologically proven, long-segment BE. We identified a miRNA signature of Barrett's carcinogenesis consisting of an increased expression of 6 miRNAs and a reduced expression of 7 miRNAs. To further support these results, we investigated target gene expression using the Oncomine database and/or immunohistochemical analysis. We found that target gene expression correlated significantly with miRNA dysregulation. Specific miRNAs are directly involved in BE progression to cancer. miRNA profiling significantly expands current knowledge on the molecular history of Barrett's carcinogenesis, also identifying molecular markers of cancer progression.
Collapse
Affiliation(s)
- Matteo Fassan
- Department of Medical Diagnostic Sciences and Special Therapies, Surgical Pathology and Cytopathology Unit, University of Padova, Padova (PD), Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fassan M, Pizzi M, Battaglia G, Giacomelli L, Parente P, Bocus P, Ancona E, Rugge M. Programmed cell death 4 (PDCD4) expression during multistep Barrett's carcinogenesis. J Clin Pathol 2011; 63:692-6. [PMID: 20702469 PMCID: PMC2976066 DOI: 10.1136/jcp.2010.078253] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Aim To test the contribution of programmed cell death 4 (PDCD4) tumour suppressor gene in Barrett's carcinogenesis. Methods PDCD4 immunohistochemical expression was assessed in 88 biopsy samples obtained from histologically proven long-segment Barrett's mucosa (BM; 25 non-intestinal columnar metaplasia, 25 intestinal metaplasia (IM), 16 low-grade intraepithelial neoplasia (LG-IEN), 12 high-grade IEN (HG-IEN) and 10 Barrett's adenocarcinoma (BAc)). As controls, 25 additional samples of native oesophageal mucosa (N) were obtained from patients with dyspepsia. To further support the data, the expression levels of miR-21, an important PDCD4 expression regulator, in 14 N, 5 HG-IEN and 11 BAc samples were determined by quantitative real-time PCR analysis. Results PDCD4 immunostaining decreased progressively and significantly with the progression of the phenotypic changes occurring during Barrett's carcinogenesis (p<0.001). Normal basal squamous epithelial layers featured strong PDCD4 nuclear immunoreaction (mostly coexisting with weak–moderate cytoplasmic staining). Non-intestinal columnar metaplasia and intestinal metaplasia preserved a strong nuclear immunostaining; conversely, a significant decrease in PDCD4 nuclear expression was seen in dysplastic (LG-IEN and HG-IEN) and neoplastic lesions. Weak–moderate cytoplasmic immunostaining was evident in cases of LG-IEN, while HG-IEN and BAc samples showed weak cytoplasmic or no protein expression. As expected, miR-21 expression was significantly upregulated in HG-IEN and BAc samples, consistently with PDCD4 dysregulation. Conclusions These data support a significant role for PDCD4 downregulation in the progression of BM to BAc, and confirm miR-21 as a negative regulator of PDCD4 in vivo. Further efforts are needed to validate PDCD4 as a potential prognostic marker in patients with Barrett's oesophagus.
Collapse
Affiliation(s)
- Matteo Fassan
- Department of Medical Diagnostic Sciences, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Molecular response prediction in multimodality treatment for adenocarcinoma of the esophagus and esophagogastric junction. Recent Results Cancer Res 2010; 182:179-91. [PMID: 20676881 DOI: 10.1007/978-3-540-70579-6_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancers arising from the esophagus are becoming more common in the United States and Europe. In 2009, an estimate of 14,530 new cases will be diagnosed and more than 90% will die of their disease. Esophageal cancer is currently the most rapidly increasing cancer in the western world and is coinciding with a shift in histological type and primary tumor location. Despite recent improvements in the detection, surgical resection, and (radio-) chemotherapy, the overall survival (OS) of esophageal cancer remains relatively poor. It is becoming increasingly apparent that neoadjuvant chemoradiation followed by surgery may be beneficial in terms of increasing resectability and OS compared to surgery alone. Results from clinical trials are encouraging; however, they also demonstrated that only patients with major histopathological response (pCR) will benefit from neoadjuvant therapy. In addition, these therapies are expensive and the prognoses of patients who do not respond to trimodality treatment strategies appear to be inferior to that of patients who had surgery alone. Accordingly, the development of validated predictive molecular markers may not only be helpful in identifying EA patients who are more likely to respond, but they will also be critical in selecting more efficient treatment strategies with the means of a tailored, targeted, and effective therapy to the molecular profile of both the patient and their disease while minimizing and avoiding life-threatening toxicities.
Collapse
|
40
|
Nguyen GH, Schetter AJ, Chou DB, Bowman ED, Zhao R, Hawkes JE, Mathé EA, Kumamoto K, Zhao Y, Budhu A, Hagiwara N, Wang XW, Miyashita M, Casson AG, Harris CC. Inflammatory and microRNA gene expression as prognostic classifier of Barrett's-associated esophageal adenocarcinoma. Clin Cancer Res 2010; 16:5824-34. [PMID: 20947516 DOI: 10.1158/1078-0432.ccr-10-1110] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Esophageal cancer is one of the most aggressive and deadly forms of cancer; highlighting the need to identify biomarkers for early detection and prognostic classification. Our recent studies have identified inflammatory gene and microRNA signatures derived from tumor and nontumor tissues as prognostic biomarkers of hepatocellular, lung, and colorectal adenocarcinoma. Here, we examine the relationship between expression of these inflammatory genes and micro RNA (miRNA) expression in esophageal adenocarcinoma and patient survival. EXPERIMENTAL DESIGN We measured the expression of 23 inflammation-associated genes in tumors and adjacent normal tissues from 93 patients (58 Barrett's and 35 Sporadic adenocarcinomas) by quantitative reverse transcription-polymerase chain reaction. These data were used to build an inflammatory risk model, based on multivariate Cox regression, to predict survival in a training cohort (n = 47). We then determined whether this model could predict survival in a cohort of 46 patients. Expression data for miRNA-375 were available for these patients and was combined with inflammatory gene expression. RESULTS IFN-γ, IL-1α, IL-8, IL-21, IL-23, and proteoglycan expression in tumor and nontumor samples were each associated with poor prognosis based on Cox regression [(Z-score)>1.5] and therefore were used to generate an inflammatory risk score (IRS). Patients with a high IRS had poor prognosis compared with those with a low IRS in the training (P = 0.002) and test (P = 0.012) cohorts. This association was stronger in the group with Barrett's history. When combining with miRNA-375, the combined IRS/miR signature was an improved prognostic classifier than either one alone. CONCLUSION Transcriptional profiling of inflammation-associated genes and miRNA expression in resected esophageal Barrett's-associated adenocarcinoma tissues may have clinical utility as predictors of prognosis.
Collapse
Affiliation(s)
- Giang Huong Nguyen
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Karaman A, Binici DN, Kabalar ME, Koca T, Dursun H. Genomic instability in patients with Barrett's esophagus. ACTA ACUST UNITED AC 2010; 201:88-93. [DOI: 10.1016/j.cancergencyto.2010.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 04/08/2010] [Accepted: 05/04/2010] [Indexed: 12/12/2022]
|
42
|
Rugge M, Fassan M, Zaninotto G, Pizzi M, Giacomelli L, Battaglia G, Rizzetto C, Parente P, Ancona E. Aurora kinase A in Barrett's carcinogenesis. Hum Pathol 2010; 41:1380-6. [PMID: 20656315 DOI: 10.1016/j.humpath.2010.02.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 02/04/2010] [Accepted: 02/10/2010] [Indexed: 01/02/2023]
Abstract
In Barrett's mucosa, both aneuploidy and TP53 mutations are consistently recognized as markers of an increased risk of Barrett's adenocarcinoma. Overexpression of the mitotic kinase encoding gene (AURKA) results in chromosome instability (assessed from the micronuclei count) and ultimately in aneuploidy. Eighty-seven esophageal biopsy samples representative of all the phenotypic lesions occurring in the multistep process of Barrett's carcinogenesis (gastric metaplasia in 25, intestinal metaplasia in 25, low-grade intraepithelial neoplasia in 16, high-grade intraepithelial neoplasia in 11, and Barrett's adenocarcinoma in 10) were obtained from long segments of Barrett's mucosa. Twenty-five additional biopsy samples of native esophageal mucosa were used for control purposes. In all tissue samples, the immunohistochemical expression of both AURKA and TP53 gene products was scored; and the micronuclei index was calculated. AURKA immunostaining increased progressively and significantly along with dedifferentiation of the histologic phenotype (P < .001). Nine of 10 Barrett's adenocarcinomas showed AURKA immunostaining. AURKA expression correlated significantly with p53 expression and the micronuclei index (both Ps < .001). AURKA overexpression is significantly associated with Barrett's mucosa progressing to Barrett's adenocarcinoma and contributes to esophageal carcinogenesis via chromosome instability. The identification of AURKA as a novel molecular target of cancer progression in Barrett's mucosa provides a lead for the development of new therapeutic approaches in Barrett's mucosa patients.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medical Diagnostic Sciences & Special Therapies, Pathology Unit, University of Padova, Padua, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Odze RD, Maley CC. Neoplasia without dysplasia: lessons from Barrett esophagus and other tubal gut neoplasms. Arch Pathol Lab Med 2010; 134:896-906. [PMID: 20524867 DOI: 10.5858/134.6.896] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Classic pathology teaching emphasizes that neoplastic lesions of the gastrointestinal tract are characterized by architectural and cytologic abnormalities that distinguish it from normal tissue. Recent studies suggest that many important-and in some cases clonal-molecular abnormalities that lead to dysregulation of cell proliferation and differentiation (neoplasia) occur before morphologic expression of dysplasia. OBJECTIVE To summarize the biologic and pathologic features of preneoplastic conditions of the tubal gut that reveal evidence of neoplastic alteration, but without the traditional morphologic features of dysplasia, in order to provide guidance on how to identify these lesions. Particular attention is given to Barrett esophagus, a chronic inflammatory condition in which early molecular and morphologic events that drive carcinogenesis are best understood. DATA SOURCES Selected references and abstracts were obtained by a PubMed (US National Library of Medicine) search by using the search headings neoplasia, preneoplasia, dysplasia, adenoma, serrated polyps, and Barrett's esophagus between the years 1980 and 2009. CONCLUSIONS Many types of lesions throughout the tubal gut fulfill the most basic and classic principles of a neoplastic precursor lesion but lack conventional morphologic evidence of dysplasia and/or maintain the capacity for cell differentiation and maturation. All of these lesions, such as squamous dysplasia of the esophagus, dysplasia in Barrett esophagus, and hyperplastic/serrated polyps of the colon, represent early neoplastic precursor lesions but without conventional histologic features of dysplasia. It is important for pathologists to be aware of these lesions, both for diagnostic and prognostic purposes, but also so that future studies can be performed with regard to risk stratification of patients.
Collapse
Affiliation(s)
- Robert D Odze
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
44
|
Gu J, Ajani JA, Hawk ET, Ye Y, Lee JH, Bhutani MS, Hofstetter WL, Swisher SG, Wang KK, Wu X. Genome-wide catalogue of chromosomal aberrations in barrett's esophagus and esophageal adenocarcinoma: a high-density single nucleotide polymorphism array analysis. Cancer Prev Res (Phila) 2010; 3:1176-86. [PMID: 20651033 DOI: 10.1158/1940-6207.capr-09-0265] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To better understand the molecular mechanisms behind esophageal adenocarcinoma (EAC) tumorigenesis, we used high-density single nucleotide polymorphism arrays to profile chromosomal aberrations at each of the four sequential progression stages, Barrett's metaplasia (BM), low-grade dysplasia (LGD), high-grade dysplasia (HGD), and EAC, in 101 patients. We observed a significant trend toward increasing loss of chromosomes with higher progression stage. For BM, LGD, HGD, and EAC, respectively, the average numbers of chromosome arms with loss per sample were 0.30, 3.21, 7.70, and 11.90 (P for trend = 4.82 x 10(-7)), and the mean percentages of single nucleotide polymorphisms with allele loss were 0.1%, 1.8%, 6.6%, and 17.2% (P for trend = 2.64 x 10(-6)). In LGD, loss of 3p14.2 (68.4%) and 16q23.1 (47.4%) was limited to narrow regions within the FHIT (3p14.2) and WWOX (16q23.1) genes, whereas loss of 9p21 (68.4%) occurred in larger regions. A significant increase in the loss of other chromosomal regions was seen in HGD and EAC. Loss of 17p (47.6%) was one of the most frequent events in EAC. Many recurrent small regions of chromosomal loss disrupted single genes, including FHIT, WWOX, RUNX1, KIF26B, MGC48628, PDE4D, C20orf133, GMDS, DMD, and PARK2, most of which are common fragile site regions in the human genome. However, RUNX1 at 21q22 seemed to be a potential tumor suppressor gene in EAC. Amplifications were less frequent than losses and mostly occurred in EAC. 8q24 (containing Myc) and 8p23.1 (containing CTSB) were the two most frequently amplified regions. In addition, a significant trend toward increasing amplification was associated with higher progression stage.
Collapse
Affiliation(s)
- Jian Gu
- Department of Epidemiology, The University of Texas M.D. Anderson Cancer Center, Unit 1340, 1155 Pressler Boulevard, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rossi E, Villanacci V, Bassotti G, Donato F, Festa A, Cengia G, Grisanti S, Cestari R. TOPOIIalpha and HER-2/neu overexpression/amplification in Barrett's oesophagus, dysplasia and adenocarcinoma. Histopathology 2010; 57:81-9. [PMID: 20557373 PMCID: PMC2916224 DOI: 10.1111/j.1365-2559.2010.03580.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rossi E, Villanacci V, Bassotti G, Donato F, Festa A, Cengia G, Grisanti S & Cestari R (2010) Histopathology57, 81–89 TOPOIIα and HER-2/neu overexpression/amplification in Barrett’s oesophagus, dysplasia and adenocarcinoma
Collapse
Affiliation(s)
- Elisa Rossi
- 2nd Department of Pathology, University of Brescia-Spedali Civili, Brescia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang DH, Clemons NJ, Miyashita T, Dupuy AJ, Zhang W, Szczepny A, Corcoran-Schwartz IM, Wilburn DL, Montgomery EA, Wang JS, Jenkins NA, Copeland NA, Harmon JW, Phillips WA, Watkins DN. Aberrant epithelial-mesenchymal Hedgehog signaling characterizes Barrett's metaplasia. Gastroenterology 2010; 138:1810-22. [PMID: 20138038 PMCID: PMC3422577 DOI: 10.1053/j.gastro.2010.01.048] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Revised: 01/19/2010] [Accepted: 01/27/2010] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS The molecular mechanism underlying epithelial metaplasia in Barrett's esophagus remains unknown. Recognizing that Hedgehog signaling is required for early esophageal development, we sought to determine if the Hedgehog pathway is reactivated in Barrett's esophagus, and if genes downstream of the pathway could promote columnar differentiation of esophageal epithelium. METHODS Immunohistochemistry, immunofluorescence, and quantitative real-time polymerase chain reaction were used to analyze clinical specimens, human esophageal cell lines, and mouse esophagi. Human esophageal squamous epithelial (HET-1A) and adenocarcinoma (OE33) cells were subjected to acid treatment and used in transfection experiments. Swiss Webster mice were used in a surgical model of bile reflux injury. An in vivo transplant culture system was created using esophageal epithelium from Sonic hedgehog transgenic mice. RESULTS Marked up-regulation of Hedgehog ligand expression, which can be induced by acid or bile exposure, occurs frequently in Barrett's epithelium and is associated with stromal expression of the Hedgehog target genes PTCH1 and BMP4. BMP4 signaling induces expression of SOX9, an intestinal crypt transcription factor, which is highly expressed in Barrett's epithelium. We further show that expression of Deleted in Malignant Brain Tumors 1, the human homologue of the columnar cell factor Hensin, occurs in Barrett's epithelium and is induced by SOX9. Finally, transgenic expression of Sonic hedgehog in mouse esophageal epithelium induces expression of stromal Bmp4, epithelial Sox9, and columnar cytokeratins. CONCLUSIONS Epithelial Hedgehog ligand expression may contribute to the initiation of Barrett's esophagus through induction of stromal BMP4, which triggers reprogramming of esophageal epithelium in favor of a columnar phenotype.
Collapse
Affiliation(s)
- David H. Wang
- Graduate Training Program in Cellular and Molecular Medicine, Johns Hopkins University, Baltimore, MD, USA,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas J. Clemons
- Surgical Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Tomoharu Miyashita
- Department of Surgery, Department of, Johns Hopkins University, Baltimore, MD, USA
| | - Adam J. Dupuy
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
| | - Wei Zhang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Anette Szczepny
- Centre for Cancer Research, Monash Institute of Medical Research, Clayton, Victoria, Australia
| | - Ian M. Corcoran-Schwartz
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel L. Wilburn
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Jean S. Wang
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | | - John W. Harmon
- Department of Surgery, Department of, Johns Hopkins University, Baltimore, MD, USA
| | - Wayne A. Phillips
- Surgical Oncology Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - D. Neil Watkins
- Graduate Training Program in Cellular and Molecular Medicine, Johns Hopkins University, Baltimore, MD, USA,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA,Centre for Cancer Research, Monash Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
47
|
Abstract
Barrett's oesophagus is the replacement of normal squamous oesophageal epithelium with an intestinalized columnar epithelium. Although some insight has been gained as to what Barrett's oesophagus is, how this columnar epithelium emerges from within a stratified squamous epithelium remains an unanswered question. We have sought to determine whether oesophageal keratinocytes can be trans-differentiated into Barrett's oesophagus cells. Using an Affymetrix microarray, we found unexpectedly that gene-expression patterns in the Barrett's oesophagus were only slightly more similar to the normal small intestine than they were to the normal oesophagus. Thus gene-expression patterns suggest significant molecular similarities remain between Barrett's oesophagus cells and normal squamous oesophageal epithelium, despite their histological resemblance with intestine. We next determined whether directed expression of intestine-specific transcription factors could induce intestinalization of keratinocytes. Retroviral-mediated Cdx2 (Caudal-type homeobox 2) expression in immortalized human oesophageal keratinocytes engineered with human telomerase reverse transcriptase (EPC2-hTERT cells) could be established transiently, but not maintained, and was associated with a reduction in cell proliferation. Co-expression of cyclin D1 rescued proliferation in the Cdx2-expressing cells, but co-expression of dominant-negative p53 did not. Cdx2 expression in the EPC2-hTERT.D1 cells did not induce intestinalization. However, when combined with treatments that induce chromatin remodelling, there was a significant induction of Barrett's oesophagus-associated genes. Studies are ongoing to determine whether other intestinal transcription factors, either alone or in combination, can provoke greater intestinalization of oesophageal keratinocytes. We conclude that, on the basis of gene-expression patterns, Barrett's oesophagus epithelial cells may represent an intermediate between oesophageal keratinocytes and intestinal epithelial cells. Moreover, our findings suggest that it may be possible to induce Barrett's oesophagus epithelial cells from oesophageal keratinocytes by altering the expression of certain critical genes.
Collapse
|
48
|
Reid BJ, Li X, Galipeau PC, Vaughan TL. Barrett's oesophagus and oesophageal adenocarcinoma: time for a new synthesis. Nat Rev Cancer 2010; 10:87-101. [PMID: 20094044 PMCID: PMC2879265 DOI: 10.1038/nrc2773] [Citation(s) in RCA: 312] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The public health importance of Barrett's oesophagus lies in its association with oesophageal adenocarcinoma. The incidence of oesophageal adenocarcinoma has risen at an alarming rate over the past four decades in many regions of the Western world, and there are indications that the incidence of this disease is on the rise in Asian populations in which it has been rare. Much has been learned of host and environmental risk factors that affect the incidence of oesophageal adenocarcinoma, and data indicate that patients with Barrett's oesophagus rarely develop oesophageal adenocarcinoma. Given that 95% of oesophageal adenocarcinomas arise in individuals without a prior diagnosis of Barrett's oesophagus, what strategies can be used to reduce late diagnosis of oesophageal adenocarcinoma?
Collapse
Affiliation(s)
- Brian J Reid
- Divisions of Public Health Sciences and Human Biology, Fred Hutchinson Cancer Research Center, University of Washington, 98109 Seattle, USA.
| | | | | | | |
Collapse
|
49
|
Boonstra JJ, van Marion R, Beer DG, Lin L, Chaves P, Ribeiro C, Pereira AD, Roque L, Darnton SJ, Altorki NK, Schrump DS, Klimstra DS, Tang LH, Eshleman JR, Alvarez H, Shimada Y, van Dekken H, Tilanus HW, Dinjens WNM. Verification and unmasking of widely used human esophageal adenocarcinoma cell lines. J Natl Cancer Inst 2010; 102:271-4. [PMID: 20075370 DOI: 10.1093/jnci/djp499] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
For decades, hundreds of different human tumor type-specific cell lines have been used in experimental cancer research as models for their respective tumors. The veracity of experimental results for a specific tumor type relies on the correct derivation of the cell line. In a worldwide effort, we verified the authenticity of all available esophageal adenocarcinoma (EAC) cell lines. We proved that the frequently used cell lines SEG-1 and BIC-1 and the SK-GT-5 cell line are in fact cell lines from other tumor types. Experimental results based on these contaminated cell lines have led to ongoing clinical trials recruiting EAC patients, to more than 100 scientific publications, and to at least three National Institutes of Health cancer research grants and 11 US patents, which emphasizes the importance of our findings. Widespread use of contaminated cell lines threatens the development of treatment strategies for EAC.
Collapse
Affiliation(s)
- Jurjen J Boonstra
- Department of Pathology, Josephine Nefkens Institute, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Carcinoma of the oesophagus including carcinoma of gastro-oesophageal junction are rapidly increasing in incidence. During recent years there have been changes in the knowledge surrounding biology of the disease progression. Identification of dysplasia in mucosal biopsies is the most reliable pathologic indicator of an increased risk of development of squamous cell carcinoma and passes through the sequence of chronic esophagitis, low-grade and high-grade dysplasia and invasive carcinoma. Although Barrett's esophagus is a precursor to esophageal adenocarcinoma and have a well described sequence of carcinogenesis: the Barrett's metaplasia-dysplasia-adenocarcinoma sequence, not all patients with this disorder require intensive surveillance. The natural history of dysplasia is poorly understood, particularly in low-risk regions, and prospective follow-up studies are needed. Adjunctive methods to improve reproducibility, such as immunostaining for alpha-methylacyl-coenzyme A racemase (AMACR), show promise, but require confirmation in larger studies. In addition, several controversial methods such as detection of p16, p53, and DNA content abnormalities may help identify patients at particularly high risk for progression to cancer, but these techniques are not yet widely available for routine clinical application. More studies are needed to define other early nonmorphologic biomarkers for risk of squamous cell carcinoma. Recent evidence regarding the importance of several histopathologically derived prognostic factors, such as circumferential resection margin status and lymph node metastases are evaluated, including lymph node micrometastases and the sentinel node concept. With the rising use of multimodal treatments for oesophageal cancer it is important that the response of the tumour to this therapy can be carefully documented by histopathology.
Collapse
|