1
|
Gambella A, Fiocca R, Lugaresi M, D'Errico A, Malvi D, Spaggiari P, Tomezzoli A, Albarello L, Ristimäki A, Bottiglieri L, Bonora E, Krishnadath KK, Raulli GD, Rosati R, Romario UF, De Manzoni G, Räsänen J, Mattioli S, Grillo F, Mastracci L. Pre-Surgical Endoscopic Biopsies Are Representative of Esophageal and Esophago-Gastric Junction Adenocarcinoma Histologic Classes and Survival Risk. Cancers (Basel) 2024; 16:4045. [PMID: 39682231 DOI: 10.3390/cancers16234045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Background and Objectives: The Esophageal Adenocarcinoma Study Group Europe (EACSGE) recently proposed a granular histologic classification of esophageal-esophago-gastric junctional adenocarcinomas (EA-EGJAs) based on the study of naïve surgically resected specimens that, when combined with the pTNM stage, is an efficient indicator of prognosis, molecular events, and response to treatment. In this study, we compared histologic classes of endoscopic biopsies taken before surgical resection with those of the surgical specimen, to evaluate the potential of the EACSGE classification at the initial diagnostic workup. Methods: A total of 106 EA-EGJA cases with available endoscopic biopsies and matched surgical resection specimens were retrieved from five Italian institutions. Histologic classification was performed on all specimens to identify well-differentiated glandular adenocarcinoma (WD-GAC), poorly differentiated glandular adenocarcinoma (PD-GAC), mucinous muconodular carcinoma (MMC), infiltrative mucinous carcinoma (IMC), diffuse desmoplastic carcinoma, diffuse anaplastic carcinoma (DAC), and mixed subtypes. Related risk subgroups (low-risk versus high-risk) were also assessed. The correlations of histologic classes and risk subgroups between diagnostic biopsies and surgical resection specimens were explored with Spearman's correlation test. Sensitivity, specificity, accuracy, positive predictive value, negative predictive value, true positives, true negatives, false positives, and false negatives were also calculated. Results: A strong positive correlation between biopsies and surgical specimens occurred for both histologic classes (coefficient: 0.75, p < 0.001) and risk subgroups (coefficient: 0.65, p < 0.001). The highest sensitivities and specificities were observed for MMC, IMC, and DAC (100% and 99% for all), followed by WD-GAC (sensitivity 91%, specificity 79%) and PD-GAC (sensitivity 722%, specificity 86%). The low-risk and high-risk groups presented a sensitivity and specificity of 89% and 76% (low-risk) and 76% and 89% (high-risk). Conclusions: The EACSGE histologic classification of EA-EGJAs and associated prognostic subgroups can be reliably assessed on pre-operative diagnostic biopsies. Further studies on larger and more representative cohorts of EA-EGJAs will allow us to validate our findings and confirm if the EA-EGJA biopsy histomorphology and clinical TNM staging will be as efficient as the surgical specimen histomorphology and pTNM in predicting patient prognoses and tailoring personalized therapeutic approaches.
Collapse
Affiliation(s)
- Alessandro Gambella
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Roberto Fiocca
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
| | - Marialuisa Lugaresi
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- Division of Thoracic Surgery, Maria Cecilia Hospital, GVM Care & Research Group, Cotignola, 48022 Ravenna, Italy
| | - Antonietta D'Errico
- Pathology Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Deborah Malvi
- Pathology Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Paola Spaggiari
- Unit of Anatomic Pathology, Humanitas University, 20089 Milan, Italy
| | - Anna Tomezzoli
- Unit of Anatomic Pathology, Azienda Ospedaliera di Verona, 37122 Verona, Italy
| | - Luca Albarello
- Pathology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ari Ristimäki
- Department of Pathology, HUSLAB and HUS Diagnostic Center, University of Helsinki, 00170 Helsinki, Finland
- Helsinki University Hospital, 00170 Helsinki, Finland
| | - Luca Bottiglieri
- Unit of Anatomic Pathology, Istituto Europeo di Oncologia, 20122 Milan, Italy
| | - Elena Bonora
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy
| | - Kausilia K Krishnadath
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Department of Gastroenterology and Hepatology, University Hospital Antwerp, 2650 Antwerp, Belgium
| | | | - Riccardo Rosati
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, Vita Salute San Raffaele University, 20132 Milan, Italy
| | | | - Giovanni De Manzoni
- Department of Surgery, General and Upper G.I. Surgery Division, University of Verona, 37126 Verona, Italy
| | - Jari Räsänen
- Department of General Thoracic and Esophageal Surgery, Helsinki University Hospital, Helsinki University, 00170 Helsinki, Finland
| | - Sandro Mattioli
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- Division of Thoracic Surgery, Maria Cecilia Hospital, GVM Care & Research Group, Cotignola, 48022 Ravenna, Italy
| | - Federica Grillo
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
- IRCCS San Martino Policlinic Hospital of Genoa, 16132 Genoa, Italy
| | - Luca Mastracci
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy
- IRCCS San Martino Policlinic Hospital of Genoa, 16132 Genoa, Italy
| |
Collapse
|
2
|
Abushukair H, Abushukair A, Singh M, Saeed A. Immune Checkpoint Inhibitors in Esophageal Carcinoma: Assessment of Efficacy Predictors of Response in Clinical Trials. Surg Oncol Clin N Am 2024; 33:583-593. [PMID: 38789200 DOI: 10.1016/j.soc.2023.12.024] [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: 05/26/2024]
Abstract
Esophageal carcinoma (EC) is among the most common causes of cancer-related deaths worldwide. Even with the improvement of multidisciplinary treatment modalities, advanced unresectable EC patients had limited systemic therapeutic options for an extended period. Recently, immune checkpoint inhibitors (ICIs) have been introduced to advanced EC management in both first-line and second-line options, as well as in postoperative settings in resectable EC after preoperative chemoradiation. Herein, the authors present a comprehensive review of clinical trials on administering ICIs in EC patients while discussing reported clinical, molecular, and immune biomarkers and their predictive value for treatment efficacy.
Collapse
Affiliation(s)
- Hassan Abushukair
- Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Aya Abushukair
- Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Meghana Singh
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Polewko-Klim A, Zhu S, Wu W, Xie Y, Cai N, Zhang K, Zhu Z, Qing T, Yuan Z, Xu K, Zhang T, Lu M, Ye W, Chen X, Suo C, Rudnicki WR. Identification of Candidate Therapeutic Genes for More Precise Treatment of Esophageal Squamous Cell Carcinoma and Adenocarcinoma. Front Genet 2022; 13:844542. [PMID: 35664298 PMCID: PMC9161154 DOI: 10.3389/fgene.2022.844542] [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: 12/28/2021] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
The standard therapy administered to patients with advanced esophageal cancer remains uniform, despite its two main histological subtypes, namely esophageal squamous cell carcinoma (SCC) and esophageal adenocarcinoma (AC), are being increasingly considered to be different. The identification of potential drug target genes between SCC and AC is crucial for more effective treatment of these diseases, given the high toxicity of chemotherapy and resistance to administered medications. Herein we attempted to identify and rank differentially expressed genes (DEGs) in SCC vs. AC using ensemble feature selection methods. RNA-seq data from The Cancer Genome Atlas and the Fudan-Taizhou Institute of Health Sciences (China). Six feature filters algorithms were used to identify DEGs. We built robust predictive models for histological subtypes with the random forest (RF) classification algorithm. Pathway analysis also be performed to investigate the functional role of genes. 294 informative DEGs (87 of them are newly discovered) have been identified. The areas under receiver operator curve (AUC) were higher than 99.5% for all feature selection (FS) methods. Nine genes (i.e., ERBB3, ATP7B, ABCC3, GALNT14, CLDN18, GUCY2C, FGFR4, KCNQ5, and CACNA1B) may play a key role in the development of more directed anticancer therapy for SCC and AC patients. The first four of them are drug targets for chemotherapy and immunotherapy of esophageal cancer and involved in pharmacokinetics and pharmacodynamics pathways. Research identified novel DEGs in SCC and AC, and detected four potential drug targeted genes (ERBB3, ATP7B, ABCC3, and GALNT14) and five drug-related genes.
Collapse
Affiliation(s)
- Aneta Polewko-Klim
- Institute of Computer Science, University in Bialystok, Białystok, Poland
| | - Sibo Zhu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weicheng Wu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Yijing Xie
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Ning Cai
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Kexun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Zhen Zhu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Tao Qing
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Ziyu Yuan
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Kelin Xu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Ming Lu
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Weimin Ye
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Xingdong Chen
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Chen Suo
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China
| | - Witold R. Rudnicki
- Institute of Computer Science, University in Bialystok, Białystok, Poland
- Computational Centre, University of Bialystok, Białystok, Poland
| |
Collapse
|
4
|
Impact of Environmental and Pharmacologic Changes on the Upper Gastrointestinal Microbiome. Biomedicines 2021; 9:biomedicines9060617. [PMID: 34072493 PMCID: PMC8229529 DOI: 10.3390/biomedicines9060617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 02/08/2023] Open
Abstract
Diseases of the upper gastrointestinal tract have become more prevalent over time. Mechanisms of disease formation are still only partially understood. Recent literature has shown that the surrounding microbiome affects the propensity for disease formation in various parts of the upper gastrointestinal tract. A review was performed of any literature to our best knowledge concerning the effects of pharmacologic agents, environmental changes, and surgical intervention on the microbiome of the upper gastrointestinal tract. Searches of the literature were performed using specific keywords related to drugs, surgical procedures, and environmental factors. Many prescription and nonprescription drugs that are commonly used have varying effects on the upper gastrointestinal tract. Proton pump inhibitors may affect the relative prevalence of some organisms in the lower esophagus and have less effect in the proximal esophagus. Changes in the esophageal microbiome correlate with some esophageal diseases. Drugs that induce weight loss have also been shown to affect the microbiomes of the esophagus and stomach. Common surgical procedures are associated with shifts in the microbial community in the gastrointestinal tract. Environmental factors have been shown to affect the microbiome in the upper gastrointestinal tract, as geographic differences correlate with alterations in the microbiome of the gastrointestinal tract. Understanding the association of environmental and pharmacologic changes on the microbiome of the upper gastrointestinal tract will facilitate treatment plans to reduce morbidity from disease.
Collapse
|
5
|
Gou Y, Jin D, He S, Han S, Bai Q. RNF168 is highly expressed in esophageal squamous cell carcinoma and contributes to the malignant behaviors in association with the Wnt/β-catenin signaling pathway. Aging (Albany NY) 2021; 13:5403-5414. [PMID: 33493132 PMCID: PMC7950303 DOI: 10.18632/aging.202471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 12/09/2020] [Indexed: 12/09/2022]
Abstract
E3 ubiquitin ligase RING finger protein 168 (RNF168) is one of the key proteins in DNA damage repair. Abnormal expression of RNF168 has recently been found in some tumors. However, the role of RNF168 in the development of esophageal squamous cell carcinoma (ESCC) has not been fully elucidated. Here we report that expression of RNF168 in esophageal squamous cell carcinoma is increased with respect to normal esophageal epithelial tissue. Notably, in ESCC patients, increased RNF168 expression was associated with tumor stage and depth of invasion. Knockdown of the RNF168 gene inhibited proliferation of esophageal cancer cells, promoted cell apoptosis, and interfered with cell movement, ultimately inhibiting tumor xenograft growth. Mechanistic studies showed that RNF168 influenced the malignant behavior of esophageal cancer cells by regulating the Wnt/ β-catenin signaling pathway. In addition, RNF168 expression was positively correlated with wingless-type MMTV integration site family member 3A (WNT3A) expression, and high expression of RNF168 and WNT3A predicted a low survival rate. In conclusion, our findings highlight the important role of RNF168 in ESCC tumorigenesis and provide new biomarkers and therapeutic targets for the treatment of ESCC.
Collapse
Affiliation(s)
- Yunjiu Gou
- Department of Thoracic Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Dacheng Jin
- Department of Thoracic Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Shengliang He
- Department of Thoracic Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Songchen Han
- Department of Thoracic Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Qizhou Bai
- Department of Thoracic Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| |
Collapse
|
6
|
The Expression of Tripartite Motif Protein 36 and β-Catenin Correlates with the Prognosis of Esophageal Cancer. Gastroenterol Res Pract 2020; 2020:7641761. [PMID: 32802046 PMCID: PMC7415112 DOI: 10.1155/2020/7641761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 01/03/2023] Open
Abstract
Aims Tripartite motif protein 36 (TRIM36) plays a tumor-suppressive role in prostate cancer. However, there is little information on the clinical relevance of TRIM36 expression in esophageal cancer (ESCA). Methods TRIM36 expression was analyzed by using The Cancer Genome Atlas (TCGA) ESCA dataset as well as by quantitative real-time polymerase chain reaction (PCR) and immunohistochemical (IHC) staining on samples from our hospital. Results In the current study, the analysis of TCGA ESCA dataset suggested the decreased expression of TRIM36 in ESCA tissues. Further analyses on samples from our hospital demonstrated that TRIM36 was significantly downregulated in ESCA tissues than in the noncancerous controls at both the mRNA and protein levels. Moreover, gene set enrichment analysis on TCGA ESCA dataset suggested that TRIM36 expression was inversely correlated with the β-catenin pathway. IHC staining data showed that 66.25% (53/80) and 51.25% (41/80) of ESCA cases had a low expression of TRIM36 and a high expression of β-catenin, respectively. By Fisher's exact test, we found that TRIM36 protein expression was significantly correlated with tumor size (P = 0.0104), tumor stage (P = 0.0169), lymph node metastasis (P = 0.0021), vital status (P = 0.0443), and β-catenin expression (P = 0.0329). These findings suggest the potential clinical significance of TRIM36 in ESCA. Kaplan–Meier and log-rank test demonstrated that a low expression of TRIM6 and a high expression of β-catenin were associated with poor overall survival of ESCA patients. Conclusions Our study provides evidence for the prognostic value of TRIM36 in ESCA.
Collapse
|
7
|
Zhang J, Wang JL, Zhang CY, Ma YF, Zhao R, Wang YY. The prognostic role of FZD6 in esophageal squamous cell carcinoma patients. Clin Transl Oncol 2019; 22:1172-1179. [PMID: 31748958 DOI: 10.1007/s12094-019-02243-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/03/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a kind of cancer with heterogeneous biological characteristics, which is affected by a complex network of gene interactions. Identification of molecular biomarkers paves the way for individualized therapy based on gene expression profiles, which can overcome the heterogeneity of ESCC. METHODS In this study, GSE20347, GSE23400 and GSE45670 datasets were retrieved from Gene Expression Omnibus (GEO) database, and the overlapping differentially expressed genes (DEGs) in three datasets were screened. Then the overlapping DEGs function was annotated by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway-enrichment analysis. The prognostic value of the top five KEGG pathway-related genes were further validated in The Cancer Genome Atlas (TCGA) database. After extensive statistical analysis, four genes (CDC25B, CXCL8, FZD6 and MCM4) were identified as potential prognostic markers. Among the four candidate genes, the prognostic value of FZD6 in ESCC patients has not been evaluated. Therefore, we finally used immunohistochemistry method to evaluate the effect of FZD6 on the prognosis of patients with ESCC. Additionally, we detected the expression level of FZD6 in ESCC cell line and normal esophageal epithelial cell line, and observed the cell viability of ESCC cell line after FZD6 knockdown. RESULTS The results showed that the overexpression of FZD6 predicted poor overall survival (OS) (P = 0.005) and progression-free survival (PFS) (P = 0.004) in ESCC patients. COX regression analysis showed that N stage (P = 0.026) and FZD6 expression level (P = 0.001) were independent prognostic factors of OS for ESCC patients. Furthermore, compared with normal esophageal epithelial cell line, the up-regulation of FZD6 was detected in ESCC cell line. Knockdown of FZD6 could significantly inhibit the proliferation of ESCC cells (P < 0.001). CONCLUSION CDC25B, CXCL8, FZD6 and MCM4 were screened as candidate genes for prognosis assessment of patients with ESCC. The prognostic role of FZD6 in ESCC patients was confirmed in current study.
Collapse
Affiliation(s)
- J Zhang
- Graduate School, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.,The First Peoples Hospital of Yinchuan, Yinchuan, 750001, Ningxia, China
| | - J-L Wang
- Graduate School, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - C-Y Zhang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, No.804 Shengli Str., Yinchuan, 750004, Ningxia, China.,Department of Radiation Oncology, General Hospital of Chinese Armed Police Force Border Defence Force, Shenzhen, 518000, Guangdong, China
| | - Y-F Ma
- Department of Thoracic Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - R Zhao
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, No.804 Shengli Str., Yinchuan, 750004, Ningxia, China
| | - Y-Y Wang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, No.804 Shengli Str., Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
8
|
Rafieenia F, Abbaszadegan MR, Poursheikhani A, Razavi SMS, Jebelli A, Molaei F, Aghaee‐Bakhtiari SH. In silico evidence of high frequency of miRNA‐related SNPs in Esophageal Squamous Cell Carcinoma. J Cell Physiol 2019; 235:966-978. [DOI: 10.1002/jcp.29012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/31/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Fatemeh Rafieenia
- Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad Iran
- Immunology Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Arash Poursheikhani
- Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad Iran
| | | | - Amir Jebelli
- Stem Cell and Regenerative Medicine Research Department Iranian Academic Center for Education, Culture and Research (ACECR), Mashhad Branch Mashhad Iran
| | - Fatemeh Molaei
- Medical Genetics Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Hamid Aghaee‐Bakhtiari
- Bioinformatics Research Group Mashhad University of Medical Sciences Mashhad Iran
- Department of Medical Biotechnology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
9
|
Ma W, Zhang CQ, Dang CX, Cai HY, Li HL, Miao GY, Wang JK, Zhang LJ. Upregulated long-non-coding RNA DLEU2 exon 9 expression was an independent indicator of unfavorable overall survival in patients with esophageal adenocarcinoma. Biomed Pharmacother 2019; 113:108655. [PMID: 30849637 DOI: 10.1016/j.biopha.2019.108655] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 01/17/2023] Open
Abstract
In this study, we aimed to explore the expression profiles of some known functional lncRNAs in esophageal adenocarcinoma (EAD) and to screening the potential prognostic makers, using data from The Cancer Genome Atlas (TCGA)-esophageal carcinoma (ESCA). Results showed that DLEU2 is a high potential OS related marker among 73 functional lncRNAs. DLEU2 and its intronic miR-15a and miR-16-1 expression were significantly upregulated in EAD compared with adjacent normal tissues. However, miR-15a and miR-16-1 expression were only weakly correlated with DLEU2 expression. Univariate and multivariate analysis confirmed that DLEU2 expression, but not miR-15a or miR-16-1 expression is an independent prognostic marker in terms of OS (HR:1.688, 95%CI: 1.085-2.627, p = 0.020) in EAD patients. The exon 9 of DLEU2 is very strongly co-expressed with DLEU2 (Pearson's r = 0.96) and showed better predictive value than total DLEU2 expression in predicting the OS of EAD patients. Multivariate analysis confirmed its independent prognostic value (HR:1.970, 95%CI: 1.266-3.067, p = 0.003), after adjustment of histologic grade, pathological stages and the presence of residual tumor. By checking the methylation status of DLEU2 gene, we excluded the possibility of the influence of two CpG sites near the DLEU2 exon 9 locus on its expression. In addition, although copy number alterations (CNAs) were observed DLEU2 gene, heterozygous loss (-1), low-level copy gain (+1) and high-level amplification (+2) had no significant association with DLEU2 transcription. Based on these findings, we infer that DLEU2 exon 9 expression might serve as a valuable biomarker of unfavorable OS in EAD patients.
Collapse
Affiliation(s)
- Wen Ma
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China; Department of Radiotherapy, Gansu Province Hospital, Lanzhou, 730000, China
| | - Chang-Qing Zhang
- Department of Tumor Center, Gansu Province Hospital, Lanzhou, 730000, China
| | - Cheng-Xue Dang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China.
| | - Hong-Yi Cai
- Department of Radiotherapy, Gansu Province Hospital, Lanzhou, 730000, China
| | - Hong-Ling Li
- Department of Tumor Center, Gansu Province Hospital, Lanzhou, 730000, China
| | - Guo-Ying Miao
- Department of Radiotherapy, Gansu Province Hospital, Lanzhou, 730000, China
| | - Jian-Kai Wang
- Department of Radiotherapy, Gansu Province Hospital, Lanzhou, 730000, China
| | - Li-Juan Zhang
- Department of Radiotherapy, Gansu Province Hospital, Lanzhou, 730000, China
| |
Collapse
|
10
|
Zhu X, Luo X, Feng G, Huang H, He Y, Ma W, Zhang C, Zeng M, Liu H. CENPE expression is associated with its DNA methylation status in esophageal adenocarcinoma and independently predicts unfavorable overall survival. PLoS One 2019; 14:e0207341. [PMID: 30716092 PMCID: PMC6361429 DOI: 10.1371/journal.pone.0207341] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/30/2018] [Indexed: 12/20/2022] Open
Abstract
Centrosome-associated protein E (CENPE) is a plus end-directed kinetochore motor protein, which plays a critical role in mitosis. In this in silico study, using data from the Cancer Genome Atlas-Esophageal Carcinoma (TCGA-ESCA), we analyzed the expression profile of CENPE mRNA in esophageal squamous cell carcinoma (ESCC) and adenocarcinoma (EA), its independent prognostic value and the potential mechanisms of its dysregulation in EA. Results showed that both ESCC and EA tissues had significantly elevated CENPE expression compared with their respective adjacent normal tissues. However, Kaplan-Meier survival curves showed that high CENPE was associated with unfavorable OS in EA. Univariate and multivariate analysis confirmed that CENPE expression was an independent indicator of unfavorable OS in EA patients, as a continuous variable (HR: 1.861, 95%CI: 1.235–2.806, p = 0.003) or as categorical variables (HR: 2.550, 95%CI: 1.294–5.025, p = 0.007). However, CENPE expression had no prognostic value in ESCC. Compared with the methylation status in normal samples, 3 CpG sites were hypomethylated (cg27388036, cg27443373, and cg24651824) in EA, among which two sites (cg27443373 and cg24651824) showed moderately negative correlation with CENPE expression. In addition, we also found that although heterozygous loss (-1) was frequent in EA (50/88, 56.8%), it was not necessarily associated with decreased CENPE expression compared with the copy neutral (0) cases. The methylation of the -1 group was significantly lower than that of the +1/0 group (p = 0.04). Based on these findings, we infer that CENPE upregulation in EA might serve as a valuable indicator of unfavorable OS. The methylation status of cg27443373 and cg24651824 might play a critical role in modulating CENPE expression.
Collapse
Affiliation(s)
- Xueqiang Zhu
- Cancer Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xing Luo
- Cancer Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Gang Feng
- Division of Thoracic Surgery, Department of Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Huang
- Cancer Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yangke He
- Cancer Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wen Ma
- Department of Tumor Center, Gansu Provincial People's Hospital, Lanzhou, China
| | - Changqing Zhang
- Department of Tumor Center, Gansu Provincial People's Hospital, Lanzhou, China
| | - Ming Zeng
- Cancer Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hao Liu
- Cancer Center, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- * E-mail:
| |
Collapse
|
11
|
Tungekar A, Mandarthi S, Mandaviya PR, Gadekar VP, Tantry A, Kotian S, Reddy J, Prabha D, Bhat S, Sahay S, Mascarenhas R, Badkillaya RR, Nagasampige MK, Yelnadu M, Pawar H, Hebbar P, Kashyap MK. ESCC ATLAS: A population wide compendium of biomarkers for Esophageal Squamous Cell Carcinoma. Sci Rep 2018. [PMID: 30143675 DOI: 10.1038/s41598-018-30579-3,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Esophageal cancer (EC) is the eighth most aggressive malignancy and its treatment remains a challenge due to the lack of biomarkers that can facilitate early detection. EC is identified in two major histological forms namely - Adenocarcinoma (EAC) and Squamous cell carcinoma (ESCC), each showing differences in the incidence among populations that are geographically separated. Hence the detection of potential drug target and biomarkers demands a population-centric understanding of the molecular and cellular mechanisms of EC. To provide an adequate impetus to the biomarker discovery for ESCC, which is the most prevalent esophageal cancer worldwide, here we have developed ESCC ATLAS, a manually curated database that integrates genetic, epigenetic, transcriptomic, and proteomic ESCC-related genes from the published literature. It consists of 3475 genes associated to molecular signatures such as, altered transcription (2600), altered translation (560), contain copy number variation/structural variations (233), SNPs (102), altered DNA methylation (82), Histone modifications (16) and miRNA based regulation (261). We provide a user-friendly web interface ( http://www.esccatlas.org , freely accessible for academic, non-profit users) that facilitates the exploration and the analysis of genes among different populations. We anticipate it to be a valuable resource for the population specific investigation and biomarker discovery for ESCC.
Collapse
Affiliation(s)
- Asna Tungekar
- Mbiomics, Manipal, Karnataka, India.,Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | - Sumana Mandarthi
- Mbiomics, Manipal, Karnataka, India.,Department of Biochemistry, Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| | - Pooja Rajendra Mandaviya
- Mbiomics, Manipal, Karnataka, India.,Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | - Veerendra P Gadekar
- Mbiomics, Manipal, Karnataka, India.,Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India.,Institute for Theoretical Chemistry, University of Vienna, Währingerstrasse 17, 1090, Vienna, Austria
| | - Ananthajith Tantry
- Mbiomics, Manipal, Karnataka, India.,Manipal Center for Information Sciences, Manipal University, Manipal, Karnataka, India
| | - Sowmya Kotian
- Mbiomics, Manipal, Karnataka, India.,Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | - Jyotshna Reddy
- Mbiomics, Manipal, Karnataka, India.,Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | | | - Sushma Bhat
- Mbiomics, Manipal, Karnataka, India.,Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | | | - Roshan Mascarenhas
- Mbiomics, Manipal, Karnataka, India.,Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India.,Newcastle University Medicine Malaysia, Johor Bahru, 79200, Malaysia
| | - Raghavendra Rao Badkillaya
- Mbiomics, Manipal, Karnataka, India.,Department of Biotechnology, Alva's college, Moodubidre, Karnataka, India
| | - Manoj Kumar Nagasampige
- Mbiomics, Manipal, Karnataka, India.,Department of Biotechnology, Sikkim Manipal University, Gangtok, Sikkim, 737102, India
| | - Mohan Yelnadu
- Mbiomics, Manipal, Karnataka, India.,Manipal Center for Information Sciences, Manipal University, Manipal, Karnataka, India.,Infosys Technologies Ltd, Bangalore, Karnataka, India.,Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Harsh Pawar
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Prashantha Hebbar
- Mbiomics, Manipal, Karnataka, India. .,Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India.
| | - Manoj Kumar Kashyap
- Mbiomics, Manipal, Karnataka, India. .,Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh 173229, India. .,School of Life and Allied Health Sciences, Glocal University, Saharanpur, Uttar Pradesh, 247001, India. .,Institute for Theoretical Chemistry, University of Vienna, Währingerstrasse 17, 1090, Vienna, Austria.
| |
Collapse
|
12
|
Omstead AN, Kosovec JE, Matsui D, Martin SA, Smith MA, Aaron Guel D, Kolano J, Komatsu Y, Habib F, Lai C, Christopher K, Kelly RJ, Zaidi AH, Jobe BA. Serial Endoscopic Evaluation of Esophageal Disease in a Cancer Model: A Paradigm Shift for Esophageal Adenocarcinoma (EAC) Drug Discovery and Development. Cancer Invest 2018; 36:363-370. [PMID: 30142016 DOI: 10.1080/07357907.2018.1499029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A rat model of surgically induced reflux recapitulates the development and progression of human esophageal adenocarcinoma (EAC). In this study, reflux was induced in rats followed by postoperative endoscopy with biopsy, to diagnose and monitor disease progression. Overall, percentage agreement between visual endoscopy and gold standard histology was 95%, with disease-specific classification accuracies of 100% and 75% for Barrett's with dysplasia and EAC, respectively. Additionally, the percentage agreement for biopsy in tumors >4 mm was 75%. Thereby, establishing endoscopic evaluation as a reliable tool to assess disease progression and provide biopsies for downstream correlates in a de novo EAC model.
Collapse
Affiliation(s)
- Ashten N Omstead
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Juliann E Kosovec
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Daisuke Matsui
- b Department of Gastroenterological Surgery , Kanazawa University Hospital , Kanazawa , Ishikawa , Japan
| | - Samantha A Martin
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Matthew A Smith
- c Department of Pathology and Laboratory Medicine , Allegheny Health Network , Pittsburgh , PA , USA
| | - D Aaron Guel
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Jenna Kolano
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Yoshihiro Komatsu
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Fahim Habib
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Christopher Lai
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Kevi Christopher
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Ronan J Kelly
- d Department of Oncology, Sidney Kimmel Comprehensive Cancer Center , Johns Hopkins Hospital , Baltimore , MD , USA
| | - Ali H Zaidi
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| | - Blair A Jobe
- a Esophageal and Lung Institute , Allegheny Health Network , Pittsburgh , PA , USA
| |
Collapse
|
13
|
Tungekar A, Mandarthi S, Mandaviya PR, Gadekar VP, Tantry A, Kotian S, Reddy J, Prabha D, Bhat S, Sahay S, Mascarenhas R, Badkillaya RR, Nagasampige MK, Yelnadu M, Pawar H, Hebbar P, Kashyap MK. ESCC ATLAS: A population wide compendium of biomarkers for Esophageal Squamous Cell Carcinoma. Sci Rep 2018; 8:12715. [PMID: 30143675 PMCID: PMC6109081 DOI: 10.1038/s41598-018-30579-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 08/01/2018] [Indexed: 02/07/2023] Open
Abstract
Esophageal cancer (EC) is the eighth most aggressive malignancy and its treatment remains a challenge due to the lack of biomarkers that can facilitate early detection. EC is identified in two major histological forms namely - Adenocarcinoma (EAC) and Squamous cell carcinoma (ESCC), each showing differences in the incidence among populations that are geographically separated. Hence the detection of potential drug target and biomarkers demands a population-centric understanding of the molecular and cellular mechanisms of EC. To provide an adequate impetus to the biomarker discovery for ESCC, which is the most prevalent esophageal cancer worldwide, here we have developed ESCC ATLAS, a manually curated database that integrates genetic, epigenetic, transcriptomic, and proteomic ESCC-related genes from the published literature. It consists of 3475 genes associated to molecular signatures such as, altered transcription (2600), altered translation (560), contain copy number variation/structural variations (233), SNPs (102), altered DNA methylation (82), Histone modifications (16) and miRNA based regulation (261). We provide a user-friendly web interface ( http://www.esccatlas.org , freely accessible for academic, non-profit users) that facilitates the exploration and the analysis of genes among different populations. We anticipate it to be a valuable resource for the population specific investigation and biomarker discovery for ESCC.
Collapse
Affiliation(s)
- Asna Tungekar
- Mbiomics, Manipal, Karnataka, India
- Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | - Sumana Mandarthi
- Mbiomics, Manipal, Karnataka, India
- Department of Biochemistry, Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| | - Pooja Rajendra Mandaviya
- Mbiomics, Manipal, Karnataka, India
- Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | - Veerendra P Gadekar
- Mbiomics, Manipal, Karnataka, India
- Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
- Institute for Theoretical Chemistry, University of Vienna, Währingerstrasse 17, 1090, Vienna, Austria
| | - Ananthajith Tantry
- Mbiomics, Manipal, Karnataka, India
- Manipal Center for Information Sciences, Manipal University, Manipal, Karnataka, India
| | - Sowmya Kotian
- Mbiomics, Manipal, Karnataka, India
- Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | - Jyotshna Reddy
- Mbiomics, Manipal, Karnataka, India
- Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | | | - Sushma Bhat
- Mbiomics, Manipal, Karnataka, India
- Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
| | | | - Roshan Mascarenhas
- Mbiomics, Manipal, Karnataka, India
- Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India
- Newcastle University Medicine Malaysia, Johor Bahru, 79200, Malaysia
| | - Raghavendra Rao Badkillaya
- Mbiomics, Manipal, Karnataka, India
- Department of Biotechnology, Alva's college, Moodubidre, Karnataka, India
| | - Manoj Kumar Nagasampige
- Mbiomics, Manipal, Karnataka, India
- Department of Biotechnology, Sikkim Manipal University, Gangtok, Sikkim, 737102, India
| | - Mohan Yelnadu
- Mbiomics, Manipal, Karnataka, India
- Manipal Center for Information Sciences, Manipal University, Manipal, Karnataka, India
- Infosys Technologies Ltd, Bangalore, Karnataka, India
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Harsh Pawar
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Prashantha Hebbar
- Mbiomics, Manipal, Karnataka, India.
- Manipal Life Sciences Center, Manipal University, Manipal, Karnataka, India.
| | - Manoj Kumar Kashyap
- Mbiomics, Manipal, Karnataka, India.
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh 173229, India.
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, Uttar Pradesh, 247001, India.
- Institute for Theoretical Chemistry, University of Vienna, Währingerstrasse 17, 1090, Vienna, Austria.
| |
Collapse
|
14
|
Barghash A, Golob-Schwarzl N, Helms V, Haybaeck J, Kessler SM. Elevated expression of the IGF2 mRNA binding protein 2 (IGF2BP2/IMP2) is linked to short survival and metastasis in esophageal adenocarcinoma. Oncotarget 2018; 7:49743-49750. [PMID: 27391348 PMCID: PMC5226544 DOI: 10.18632/oncotarget.10439] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/26/2016] [Indexed: 12/25/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) represents the sixth leading cause of cancer-related deaths and develops in Barret's esophagus affected tissues. The IGF2 mRNA binding protein IMP2/IGF2BP2/p62 was originally identified as an autoantigen in hepatocellular carcinoma. Aim of this study was to investigate the expression and prognostic role of IMP2 in EAC. Human EAC and Barret's esophagus tissue showed overexpression of IMP2, particularly in tumors of increased size and in metastatic tissues. Molecular classification based on published gene signatures of esophageal cancer revealed a specific subtype, in which the expression of IMP2 is high. According to GO and KEGG pathway analyses, genes showing highly correlated expression with IMP2 are associated with growth, proliferation, metabolism, inflammation, and cancerous processes. Clustering of EAC samples according to published survival marker genes strongly suggests that IMP2 overexpressing samples show poor survival. Finally, IMP2 expression correlated with short survival in patients with EAC or esophageal squamous carcinoma. Our data indicate that IMP2 might be a useful prognostic marker for Barret's esophagus and EAC.
Collapse
Affiliation(s)
- Ahmad Barghash
- Center for Bioinformatics, Saarland University, Saarbruecken, Germany.,School of Computer Engineering and Information Technology, German Jordanian University, Amman, Jordan
| | | | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Saarbruecken, Germany
| | | | - Sonja M Kessler
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbruecken, Germany
| |
Collapse
|
15
|
Visser E, Franken IA, Brosens LAA, Ruurda JP, van Hillegersberg R. Prognostic gene expression profiling in esophageal cancer: a systematic review. Oncotarget 2018; 8:5566-5577. [PMID: 27852047 PMCID: PMC5354930 DOI: 10.18632/oncotarget.13328] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/13/2016] [Indexed: 12/18/2022] Open
Abstract
Background Individual variability in prognosis of esophageal cancer highlights the need for advances in personalized therapy. This systematic review aimed at elucidating the prognostic role of gene expression profiles and at identifying gene signatures to predict clinical outcome. Methods A systematic search of the Medline, Embase and the Cochrane library databases (2000-2015) was performed. Articles associating gene expression profiles in patients with esophageal adenocarcinoma or squamous cell carcinoma to survival, response to chemo(radio)therapy and/or lymph node metastasis were identified. Differentially expressed genes and gene signatures were extracted from each study and combined to construct a list of prognostic genes per outcome and histological tumor type. Results This review includes a total of 22 studies. Gene expression profiles were related to survival in 9 studies, to response to chemo(radio)therapy in 7 studies, and to lymph node metastasis in 9 studies. The studies proposed many differentially expressed genes. However, the findings were heterogeneous and only 12 (ALDH1A3, ATR, BIN1, CSPG2, DOK1, IFIT1, IFIT3, MAL, PCP4, PHB, SPP1) of the 1.112 reported genes were identified in more than 1 study. Overall, 16 studies reported a prognostic gene signature, which was externally validated in 10 studies. Conclusion This systematic review shows heterogeneous findings in associating gene expression with clinical outcome in esophageal cancer. Larger validated studies employing RNA next-generation sequencing are required to establish gene expression profiles to predict clinical outcome and to select optimal personalized therapy.
Collapse
Affiliation(s)
- Els Visser
- Department of Surgery, University Medical Center Utrecht, The Netherlands
| | - Ingrid A Franken
- Department of Surgery, University Medical Center Utrecht, The Netherlands
| | | | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, The Netherlands
| | | |
Collapse
|
16
|
Gene expression profiling and pathway network analysis of anti-tumor activity by Jaridon 6 in esophageal cancer. Eur J Pharmacol 2017; 815:478-486. [PMID: 28800883 DOI: 10.1016/j.ejphar.2017.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 08/01/2017] [Accepted: 08/07/2017] [Indexed: 02/07/2023]
Abstract
Jaridon 6, a novel ent-kaurene diterpenoid derived from Rabdosia rubescens (Hemsl.) Hara, possesses strong anti-tumor activity in esophageal cancer cells. In this study, we explored the underlying molecular events of the anti-tumor activity of Jaridon 6. Cell viability and apoptosis results obtained by flow cytometry confirmed the tumor inhibitory effect of Jaridon 6 in esophageal cancer cells. A cDNA microarray was performed and the observations were validated using quantitative reverse transcription polymerase chain reaction. The microarray data showed that 151 genes were differentially expressed between the untreated group and the Jaridon 6-treated group, among these were 57 upregulated genes, and 94 downregulated genes (P < 0.01, fold change threshold: 2). These included genes such as Wnt, peroxisome, and genes involved in chemokine signaling pathways. In addition, Western blot analysis demonstrated that Jaridon 6 regulated the expression of Wnt pathway proteins, including reduced levels of Dvl 2, survivin and cyclin D1, and increased levels of p-β-catenin, and AXIN2 in EC109 and EC9706 esophageal cancer cells. In addition, recombinant murine Wnt3a could change the regulation of Jaridon 6 on Wnt pathway proteins. Immunohistochemical analysis indicated that the anti-tumor activity of Jaridon 6 was closely related to the Wnt signaling pathway in esophageal cancer cells.
Collapse
|
17
|
He Y, Liu J, Zhao Z, Zhao H. Bioinformatics analysis of gene expression profiles of esophageal squamous cell carcinoma. Dis Esophagus 2017; 30:1-8. [PMID: 28375447 DOI: 10.1093/dote/dow018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Indexed: 12/11/2022]
Abstract
The objective of this study is to explore the potential target genes in the pathogenesis of esophageal squamous cell carcinoma (ESCC). The mRNA expression profile data of GSE17351 were downloaded from the Gene Expression Omnibus database, including five paired ESCC and normal tissue samples from five ESCC patients. The differentially expressed genes (DEGs) between ESCC and normal samples were identified using the limma package. The identified DEGs were then performed clustering analysis and functional enrichment analysis. Additionally, gene-miRNA network, gene-transcription factor network, and protein-protein interaction (PPI) network for the DEGs were constructed, and then significant modules were selected from the constructed PPI network. Furthermore, esophageal carcinoma RNAseq data including 185 esophageal carcinoma and 13 normal samples were downloaded from The Cancer Genome Atlas database to confirm our results. A total of 409 up- and 341 downregulated DEGs were identified. The DEGs were separated into two clusters and were mainly enriched in cell cycle function. CHEK1, CCNA2, COL11A1, and MME were hub nodes in the PPI network. Besides, total seven modules were selected in the PPI network. Genes in the most significant module were upregulated and were enriched in cell cycle. The Cancer Genome Atlas data validation identified 370 DEGs, all of which were differentially expressed in the GSE17351 dataset. Besides, the expression change direction was consistent with the DEGs in GSE17351. Cell cycle may play a role in ESCC development. The genes such as CHEK1, CCNA2, COL11A1, and MME may be served as target genes in ESCC treatment.
Collapse
|
18
|
Lin J, Myers AL, Wang Z, Nancarrow DJ, Ferrer-Torres D, Handlogten A, Leverenz K, Bao J, Thomas DG, Wang TD, Orringer MB, Reddy RM, Chang AC, Beer DG, Lin L. Osteopontin (OPN/SPP1) isoforms collectively enhance tumor cell invasion and dissemination in esophageal adenocarcinoma. Oncotarget 2016; 6:22239-57. [PMID: 26068949 PMCID: PMC4673160 DOI: 10.18632/oncotarget.4161] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/14/2015] [Indexed: 01/26/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) is often diagnosed at an advanced stage, thus understanding the molecular basis for EAC invasion and metastasis is critical. Here we report that SPP1/OPN was highly overexpressed in primary EACs and intracellularly localized to tumor cells. We further demonstrate that all known OPN isoforms (OPNa, b, c, 4 and 5) were frequently co-overexpressed in primary EACs. Distinct pro-invasion and dissemination phenotypes of isoform-specific OPNb and OPNc stable transfectants were observed. Expression of OPNb significantly enhanced cell migration and adhesion to laminin. In contrast, OPNc cells showed significantly decreased cell migration yet increased cell detachment. Enhanced invasion, both in vitro and in vivo, was observed for OPNb- but not OPNc-expressing cells. Inhibition of RGD integrins, one family of OPN receptors, attenuated OPNb cell migration, abrogated OPNb cell adhesion and significantly reduced OPNb cell clonogenic survival but did not affect OPNc phenotypes, indicating that OPNb but not OPNc acts through integrin-dependent signaling. Differential expression of vimentin, E-cadherin and β-catenin in OPN stable cells may account for the variation in cell adhesion and detachment between these isoforms. We conclude that while all OPN isoforms are frequently co-overexpressed in primary EACs, isoforms OPNb and OPNc enhance invasion and dissemination through collective yet distinct mechanisms.
Collapse
Affiliation(s)
- Jules Lin
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Amy L Myers
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Zhuwen Wang
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Derek J Nancarrow
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Daysha Ferrer-Torres
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Amy Handlogten
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kimmy Leverenz
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Julia Bao
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Dafydd G Thomas
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Thomas D Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mark B Orringer
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Rishindra M Reddy
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Andrew C Chang
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - David G Beer
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lin Lin
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Pirayesh Islamian J, Mohammadi M, Baradaran B, Farajollahi A, Aghamiri SMR, Asghari Jafarabadi M, Karami H, Monfaredan A, Shanehbandi D. Enhancing radiosensitivity of TE1, TE8, and TE 11 esophageal squamous carcinoma cell lines by Hdm2-siRNA targeted gene therapy in vitro. ACTA ACUST UNITED AC 2016; 6:93-8. [PMID: 27525226 PMCID: PMC4981254 DOI: 10.15171/bi.2016.13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/24/2016] [Accepted: 06/26/2016] [Indexed: 12/17/2022]
Abstract
![]()
Introduction: Human double minute2 (hdm2) level increases in most human malignancies. Therefore, inhibition of tumor growth and also induction of radiosensitivity may be provided by hdm2 inhibitors. The effects of hdm2-siRNA on hdm2 protein expression, cell apoptosis rate, and radiosensitivity of human esophageal squamous cell carcinoma (ESCC) were studied.
Methods: The hdm2 gene was silenced in TE1, TE8, and TE11 ESCC cell lines using 200nM siRNA by liposomal transfection method followed by irradiation with 0.5, 1, 2, 4, and 6 Gy γ-rays in vitro. The gene expression levels were evaluated by real time PCR and Western Blotting methods. MTT, TUNEL, and also colony forming assays were used to compare the radiosensitivity of the cell lines before and after the treatments.
Results: Hdm2-siRNA reduced the hdm2 protein as compared to the vehicle control and scrambled groups, and also increased the radiation-induced apoptosis especially in TE11 cells. The related dose reduction factors (DRFs) for the silenced TE1, TE8, and TE11 cells calculated to be 1.20, 1.30, and 2.75, respectively.
Conclusion: Increasing radiosensitivity of tumor cells may be provided by silencing the oncogenes.
Collapse
Affiliation(s)
- Jalil Pirayesh Islamian
- Department of Medical Physics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Mohammadi
- Department of Medical Radiation Science, School of Paramedicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Farajollahi
- Department of Medical Physics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Mahmoud Reza Aghamiri
- Department of Radiation Medicine, Faculty of Nuclear Engineering, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hadi Karami
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Amir Monfaredan
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Couch G, Redman JE, Wernisch L, Newton R, Malhotra S, Dawsey SM, Lao-Sirieix P, Fitzgerald RC. The Discovery and Validation of Biomarkers for the Diagnosis of Esophageal Squamous Dysplasia and Squamous Cell Carcinoma. Cancer Prev Res (Phila) 2016; 9:558-66. [PMID: 27072986 DOI: 10.1158/1940-6207.capr-15-0379] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/11/2016] [Indexed: 02/07/2023]
Abstract
The 5-year survival rate of esophageal cancer is less than 10% in developing countries, where more than 90% of these cancers are esophageal squamous cell carcinomas (ESCC). Endoscopic screening is undertaken in high incidence areas. Biomarker analysis could reduce the subjectivity associated with histologic assessment of dysplasia and thus improve diagnostic accuracy. The aims of this study were therefore to identify biomarkers for esophageal squamous dysplasia and carcinoma. A publicly available dataset was used to identify genes with differential expression in ESCC compared with normal esophagus. Each gene was ranked by a support vector machine separation score. Expression profiles were examined, before validation by qPCR and IHC. We found that 800 genes were overexpressed in ESCC compared with normal esophagus (P < 10(-5)). Of the top 50 genes, 33 were expressed in ESCC epithelium and not in normal esophagus epithelium or stroma using the Protein Atlas website. These were taken to qPCR validation, and 20 genes were significantly overexpressed in ESCC compared with normal esophagus (P < 0.05). TNFAIP3 and CHN1 showed differential expression with IHC. TNFAIP3 expression increased gradually through normal esophagus, mild, moderate and severe dysplasia, and SCC (P < 0.0001). CHN1 staining was rarely present in the top third of normal esophagus epithelium and extended progressively towards the surface in mild, moderate, and severe dysplasia, and SCC (P < 0.0001). Two novel promising biomarkers for ESCC were identified, TNFAIP3 and CHN1. CHN1 and TNFAIP3 may improve diagnostic accuracy of screening methods for ESCC. Cancer Prev Res; 9(7); 558-66. ©2016 AACR.
Collapse
Affiliation(s)
- George Couch
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - James E Redman
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Lorenz Wernisch
- MRC Biostatistics Unit, Robinson Way, Cambridge, United Kingdom
| | - Richard Newton
- MRC Biostatistics Unit, Robinson Way, Cambridge, United Kingdom
| | - Shalini Malhotra
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Sanford M Dawsey
- Division of Cancer Epidemiology & Genetics, NCI, Bethesda, Maryland
| | - Pierre Lao-Sirieix
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
21
|
González-Plaza JJ, Hulak N, García-Fuentes E, Garrido-Sánchez L, Zhumadilov Z, Akilzhanova A. Oesophageal squamous cell carcinoma (ESCC): Advances through omics technologies, towards ESCC salivaomics. Drug Discov Ther 2016; 9:247-57. [PMID: 26370523 DOI: 10.5582/ddt.2015.01042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Oesophageal Squamous Cell Carcinoma (ESCC) is one of the two main subtypes of oesophageal cancer, affecting mainly populations in Asia. Though there have been great efforts to develop methods for a better prognosis, there is still a limitation in the staging of this affection. As a result, ESCC is detected at advances stages, when the interventions on the patient do not have such a positive outcome, leading in many cases to recurrence and to a very low 5-year survival rate, causing high mortality. A way to decrease the number of deaths is the use of biomarkers that can trace the advance of the disease at early stages, when surgical or chemotherapeutic methodologies would have a greater effect on the evolution of the subject. The new high throughput omics technologies offer an unprecedented chance to screen for thousands of molecules at the same time, from which a new set of biomarkers could be developed. One of the most convenient types of samples is saliva, an accessible body fluid that has the advantage of being non-invasive for the patient, being easy to store or to process. This review will focus on the current status of the new omics technologies regarding salivaomics in ESCC, or when not evaluated yet, the achievements in related diseases.
Collapse
Affiliation(s)
- Juan José González-Plaza
- Laboratory of Genomic and Personalized Medicine, Center for Life Sciences, PI "National Laboratory Astana", AOE "NazarbayevUniversity"
| | | | | | | | | | | |
Collapse
|
22
|
Insulin-like growth factor binding protein-3 is a new predictor of radiosensitivity on esophageal squamous cell carcinoma. Sci Rep 2015; 5:17336. [PMID: 26670461 PMCID: PMC4680797 DOI: 10.1038/srep17336] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/29/2015] [Indexed: 01/04/2023] Open
Abstract
Insulin-like growth factor binding protein-3 (IGFBP-3) plays an essential role in radiosensitivity of esophageal squamous cell carcinoma (ESCC). However, the underlying mechanism is not completely understood. Here, we observed that IGFBP-3 had favorable impact on the tumorigenicity of ESCC cells in nude mice by using an in vivo imaging system (IVIS) to monitor tumor growth treated with ionizing radiation (IR). Downregulation of IGFBP-3 expression enhanced tumor growth, inhibited anti-proliferative and apoptotic activity and result in IR resistance in vivo. Cell cycle antibody array suggested that silencing IGFBP-3 promoted transition from G0/G1 to S phase, perhaps though influencing Smad3 dephosphorylation and retinoblastoma protein (Rb) phosphorylation. Downregulation of P21 and P27, and upregulation of p-P27 (phospho-Thr187), cyclin-dependent kinase 2 (CDK2), and cyclin E1 might contribute to the G0/G1 to S phase transition promoted by IGFBP-3. Our results suggest that Smad3-P27/P21-cyclin E1/CDK2-phosphorylated retinoblastoma protein pathways might be involved in this IGFBP-3 mediated radiosensitivity transition in ESCC.
Collapse
|
23
|
Varghese S, Newton R, Ross-Innes CS, Lao-Sirieix P, Krishnadath KK, O'Donovan M, Novelli M, Wernisch L, Bergman J, Fitzgerald RC. Analysis of dysplasia in patients with Barrett's esophagus based on expression pattern of 90 genes. Gastroenterology 2015; 149:1511-1518.e5. [PMID: 26248086 DOI: 10.1053/j.gastro.2015.07.053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 07/03/2015] [Accepted: 07/19/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Diagnoses of dysplasia, based on histologic analyses, dictate management decisions for patients with Barrett's esophagus (BE). However, there is much intra- and inter-observer variation in identification of dysplasia-particularly low-grade dysplasia. We aimed to identify a biomarker that could be used to assign patients with low-grade dysplasia to a low- or high-risk group. METHODS We performed a stringent histologic assessment of 150 frozen esophageal tissues samples collected from 4 centers in the United Kingdom (from 2000 through 2006). The following samples with homogeneous diagnoses were selected for gene expression profiling: 28 from patients with nondysplastic BE, 10 with low-grade dysplasia, 13 with high-grade dysplasia (HGD), and 8 from patients with esophageal adenocarcinoma. A leave-one-out cross-validation analysis was used identify a gene expression signature associated with HGD vs nondysplastic BE. Functional pathways associated with gene signature sets were identified using the MetaCore analysis. Gene expression signature sets were validated using gene expression data on BE and esophageal adenocarcinoma accessed through National Center for Biotechnology Information Gene Expression Omnibus, as well as a separate set of samples (n = 169) collected from patients who underwent endoscopy in the United Kingdom or the Netherlands and analyzed histologically. RESULTS We identified an expression pattern of 90 genes that could separate nondysplastic BE tissues from those with HGD (P < .0001). Genes in a pathway regulated by retinoic acid-regulated nuclear protein made the largest contribution to this gene set (P < .0001); the transcription factor MYC regulated at least 30% of genes within the signature (P < .0001). In the National Center for Biotechnology Information Gene Expression Omnibus validation set, the signature separated nondysplastic BE samples from esophageal adenocarcinoma samples (P = .0012). In the UK and Netherlands validation cohort, the signature identified dysplastic tissues with an area under the curve value of 0.87 (95% confidence interval: 0.82-0.93). Of samples with low-grade dysplasia (LGD), 64% were considered high risk according to the 90-gene signature; these patients had a higher rate of disease progression than those with a signature categorized as low risk (P = .047). CONCLUSIONS We identified an expression pattern of 90 genes in esophageal tissues of patients with BE that was associated with low- or high-risk for disease progression. This pattern might be used in combination with histologic analysis of biopsy samples to stratify patients for treatment. It would be most beneficial for analysis of patients without definitive evidence of HGD but for whom early endoscopic intervention is warranted.
Collapse
Affiliation(s)
- Sibu Varghese
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | | | - Caryn S Ross-Innes
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | - Pierre Lao-Sirieix
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | | | - Maria O'Donovan
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | - Marco Novelli
- GI Services, University College Hospital, NHS Foundation Trust, London, United Kingdom
| | | | | | - Rebecca C Fitzgerald
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom.
| |
Collapse
|
24
|
Golgi phosphoprotein 2 (GOLPH2) is a novel bile acid-responsive modulator of oesophageal cell migration and invasion. Br J Cancer 2015; 113:1332-42. [PMID: 26461057 PMCID: PMC4815786 DOI: 10.1038/bjc.2015.350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 09/07/2015] [Accepted: 09/09/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The aetiology of Barrett's oesophagus (BO) and oesophageal cancer is poorly understood. We previously demonstrated that Golgi structure and function is altered in oesophageal cancer cells. A Golgi-associated protein, GOLPH2, was previously established as a tissue biomarker for BO. Cellular functions for GOLPH2 are currently unknown, therefore in this study we sought to investigate functional roles for this Golgi-associated protein in oesophageal disease. METHODS Expression, intracellular localisation and secretion of GOLPH2 were identified by immunofluorescence, immunohistochemistry and western blot. GOLPH2 expression constructs and siRNA were used to identify cellular functions for GOLPH2. RESULTS We demonstrate that the structure of the Golgi is fragmented and the intracellular localisation of GOLPH2 is altered in BO and oesophageal adenocarcinoma tissue. GOLPH2 is secreted by oesophageal cancer cells and GOLPH2 expression, cleavage and secretion facilitate cell migration and invasion. Furthermore, exposure of cells to DCA, a bile acid component of gastric refluxate and known tumour promoter for oesophageal cancer, causes disassembly of the Golgi structure into ministacks, resulting in cleavage and secretion of GOLPH2. CONCLUSIONS This study demonstrates that GOLPH2 may be a useful tissue biomarker for oesophageal disease. We provide a novel mechanistic insight into the aetiology of oesophageal cancer and reveal novel functions for GOLPH2 in regulating tumour cell migration and invasion, important functions for the metastatic process in oesophageal cancer.
Collapse
|
25
|
Hao Y, Wu W, Shi F, Dalmolin RJS, Yan M, Tian F, Chen X, Chen G, Cao W. Prediction of long noncoding RNA functions with co-expression network in esophageal squamous cell carcinoma. BMC Cancer 2015; 15:168. [PMID: 25885227 PMCID: PMC4377028 DOI: 10.1186/s12885-015-1179-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/09/2015] [Indexed: 02/01/2023] Open
Abstract
Background Long non-coding RNAs (lncRNAs) are pervasively transcribed in the genome. They have important regulatory functions in chromatin remodeling and gene expression. Dysregulated lncRNAs have been studied in cancers, but their role in esophageal squamous cell carcinoma (ESCC) remains largely unknown. We have conducted lncRNA expression screening and a genome-wide analysis of lncRNA and coding gene expression on primary tumor and adjacent normal tissue from four ESCC patients, tend to understand the functionality of lncRNAs in carcinogenesis of esopheagus in combination with experimental and bioinformatics approach. Methods LncRNA array was used for coding and non-coding RNA expression. R program and Bioconductor packages (limma and RedeR) were used for differential expression and co-expression network analysis, followed by independent confirmation and functional studies of inferred onco-lncRNA ESCCAL-1 using quantitative real time polymerase chain reaction, small interfering RNA-mediated knockdown, apoptosis and invasion assays in vitro. Results The global coding and lncRNA gene expression pattern is able to distinguish ESCC from adjacent normal tissue. The co-expression network from differentially expressed coding and lncRNA genes in ESCC was constructed, and the lncRNA function may be inferred from the co-expression network. LncRNA ESCCAL-1 is such an example as a predicted novel onco-lncRNA, and it is overexpressed in 65% of an independent ESCC patient cohort (n = 26). More over, knockdown of ESCCAL-1 expression increases esophageal cancer cell apoptosis and reduces the invasion in vitro. Conclusion Our study uncovered the landscape of ESCC-associated lncRNAs. The systematic analysis of coding and lncRNAs co-expression network increases our understanding of lncRNAs in biological network. ESCCAL-1 is a novel putative onco-lncRNA in esophageal cancer development. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1179-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yibin Hao
- Zhengzhou Central Hospital, Affiliated to Zhengzhou University, China, 195 Tongbai Road, Zhengzhou, 450007, PR, China.
| | - Wei Wu
- Zhengzhou Central Hospital, Affiliated to Zhengzhou University, China, 195 Tongbai Road, Zhengzhou, 450007, PR, China. .,Department of Pathology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.
| | - Fachun Shi
- Science and Education Department, Health Bureau of ZhengZhou, Zhengzhou, China.
| | - Rodrigo J S Dalmolin
- Department of Biochemistry, Bioscience Center and Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.
| | - Ming Yan
- Medical School, Zhengzhou University, Zhengzhou, Henan, China.
| | - Fu Tian
- Zhengzhou Central Hospital, Affiliated to Zhengzhou University, China, 195 Tongbai Road, Zhengzhou, 450007, PR, China.
| | - Xiaobing Chen
- Zhengzhou Central Hospital, Affiliated to Zhengzhou University, China, 195 Tongbai Road, Zhengzhou, 450007, PR, China.
| | - Guoyong Chen
- Zhengzhou Central Hospital, Affiliated to Zhengzhou University, China, 195 Tongbai Road, Zhengzhou, 450007, PR, China.
| | - Wei Cao
- Zhengzhou Central Hospital, Affiliated to Zhengzhou University, China, 195 Tongbai Road, Zhengzhou, 450007, PR, China.
| |
Collapse
|
26
|
Ong CAJ, Shannon NB, Ross-Innes CS, O'Donovan M, Rueda OM, Hu DE, Kettunen MI, Walker CE, Noorani A, Hardwick RH, Caldas C, Brindle K, Fitzgerald RC. Amplification of TRIM44: pairing a prognostic target with potential therapeutic strategy. J Natl Cancer Inst 2014; 106:dju050. [PMID: 24777112 PMCID: PMC4112924 DOI: 10.1093/jnci/dju050] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 01/24/2014] [Accepted: 02/03/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Many prognostic biomarkers have been proposed recently. However, there is a lack of therapeutic strategies exploiting novel prognostic biomarkers. We aimed to propose therapeutic options in patients with overexpression of TRIM44, a recently identified prognostic gene. METHODS Genomic and transcriptomic data of epithelial cancers (n = 1932), breast cancers (BCs; n = 1980) and esophago-gastric cancers (EGCs; n = 163) were used to identify genomic aberrations driving TRIM44 overexpression. The driver gene status of TRIM44 was determined using a small interfering RNA (siRNA) screen of the 11p13 amplicon. Integrative analysis was applied across multiple datasets to identify pathway activation and potential therapeutic strategies. Validation of the in silico findings were performed using in vitro assays, xenografts, and patient samples (n = 160). RESULTS TRIM44 overexpression results from genomic amplification in 16.1% of epithelial cancers, including 8.1% of EGCs and 6.1% of BCs. This was confirmed using fluorescent in situ hybridization. The siRNA screen confirmed TRIM44 to be a driver of the amplicon. In silico analysis revealed an association between TRIM44 and mTOR signalling, supported by a decrease in mTOR signalling after siRNA knockdown of TRIM44 in cell lines and colocalization of TRIM44 and p-mTOR in patient samples. In vitro inhibition studies using an mTOR inhibitor (everolimus) decreased cell viability in two TRIM44-amplified cells lines by 88% and 70% compared with 35% in the control cell line. These findings were recapitulated in xenograft models. CONCLUSIONS Genomic amplification drives TRIM44 overexpression in EGCs and BCs. Targeting the mTOR pathway provides a potential therapeutic option for TRIM44-amplified tumors.
Collapse
Affiliation(s)
- Chin-Ann Johnny Ong
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Nicholas B Shannon
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Caryn S Ross-Innes
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Maria O'Donovan
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Oscar M Rueda
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - De-En Hu
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Mikko I Kettunen
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Christina Elaine Walker
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Ayesha Noorani
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Richard H Hardwick
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Carlos Caldas
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Kevin Brindle
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH)
| | - Rebecca C Fitzgerald
- Affiliations of authors: MRC Cancer Unit, Cambridge, UK (C-AJO, CSR-I, MO, CEW, AN, RCF); Cancer Research UK Cambridge Institute, Cambridge, UK (NBS, OMR, D-eH, MIK, CC, KB); UK Cambridge Esophagogastric Centre, Addenbrooke's Hospital, Cambridge, UK (RHH).
| |
Collapse
|
27
|
Hyland PL, Hu N, Rotunno M, Su H, Wang C, Wang L, Pfeiffer RM, Gherman B, Giffen C, Dykes C, Dawsey SM, Abnet CC, Johnson KM, Acosta RD, Young PE, Cash BD, Taylor PR. Global changes in gene expression of Barrett's esophagus compared to normal squamous esophagus and gastric cardia tissues. PLoS One 2014; 9:e93219. [PMID: 24714516 PMCID: PMC3979678 DOI: 10.1371/journal.pone.0093219] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 03/03/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Barrett's esophagus (BE) is a metaplastic precursor lesion of esophageal adenocarcinoma (EA), the most rapidly increasing cancer in western societies. While the prevalence of BE is increasing, the vast majority of EA occurs in patients with undiagnosed BE. Thus, we sought to identify genes that are altered in BE compared to the normal mucosa of the esophagus, and which may be potential biomarkers for the development or diagnosis of BE. DESIGN We performed gene expression analysis using HG-U133A Affymetrix chips on fresh frozen tissue samples of Barrett's metaplasia and matched normal mucosa from squamous esophagus (NE) and gastric cardia (NC) in 40 BE patients. RESULTS Using a cut off of 2-fold and P<1.12E-06 (0.05 with Bonferroni correction), we identified 1324 differentially-expressed genes comparing BE vs NE and 649 differentially-expressed genes comparing BE vs NC. Except for individual genes such as the SOXs and PROM1 that were dysregulated only in BE vs NE, we found a subset of genes (n = 205) whose expression was significantly altered in both BE vs NE and BE vs NC. These genes were overrepresented in different pathways, including TGF-β and Notch. CONCLUSION Our findings provide additional data on the global transcriptome in BE tissues compared to matched NE and NC tissues which should promote further understanding of the functions and regulatory mechanisms of genes involved in BE development, as well as insight into novel genes that may be useful as potential biomarkers for the diagnosis of BE in the future.
Collapse
Affiliation(s)
- Paula L. Hyland
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nan Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Melissa Rotunno
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hua Su
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Chaoyu Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lemin Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ruth M. Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Carol Giffen
- Information Management Services, Inc, Silver Spring, Maryland, United States of America
| | - Cathy Dykes
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States of America
| | - Sanford M. Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christian C. Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kathryn M. Johnson
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States of America
| | - Ruben D. Acosta
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States of America
| | - Patrick E. Young
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States of America
| | - Brooks D. Cash
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States of America
| | - Philip R. Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
28
|
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
|
29
|
Frankel A, Armour N, Nancarrow D, Krause L, Hayward N, Lampe G, Smithers BM, Barbour A. Genome-wide analysis of esophageal adenocarcinoma yields specific copy number aberrations that correlate with prognosis. Genes Chromosomes Cancer 2014; 53:324-38. [PMID: 24446147 DOI: 10.1002/gcc.22143] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 12/18/2013] [Indexed: 12/20/2022] Open
Abstract
The incidence of esophageal adenocarcinoma (EAC) has been increasing rapidly for the past 3 decades in Western (Caucasian) populations. Curative treatment is based around esophagectomy, which has a major impact on quality of life. For those suitable for treatment with curative intent, 5-year survival is ∼30%. More accurate prognostic tools are therefore needed, and copy number aberrations (CNAs) may offer the ability to act as prospective biomarkers in this regard. We performed a genome-wide examination of CNAs in 54 samples of EAC using single-nucleotide polymorphism (SNP) arrays. Our aims were to describe frequent regions of CNA, to define driver CNAs, and to identify CNAs that correlated with survival. Regions of frequent amplification included oncogenes such as EGFR, MYC, KLF12, and ERBB2, while frequently deleted regions included tumor suppressor genes such as CDKN2A/B, PTPRD, FHIT, and SMAD4. The genomic identification of significant targets in cancer (GISTIC) algorithm identified 24 regions of gain and 28 regions of loss that were likely to contain driver changes. We discovered 61 genes in five regions that, when stratified by CNA type (gain or loss), correlated with a statistically significant difference in survival. Pathway analysis of the genes residing in both the GISTIC and prognostic regions showed they were significantly enriched for cancer-related networks. Finally, we discovered that copy-neutral loss of heterozygosity is a frequent mechanism of CNA in genes currently targetable by chemotherapy, potentially leading to under-reporting of cases suitable for such treatment.
Collapse
Affiliation(s)
- Adam Frankel
- Surgical Oncology Group, School of Medicine, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, 4102 Brisbane, QLD 4006, Australia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Shackelford RE, Abdelbaqi MQ, Almhanna K, Meredith K, Coppola D. Molecular Pathology and Diagnostics in Esophago-gastric Cancer. MOLECULAR PATHOLOGY AND DIAGNOSTICS OF CANCER 2014:177-210. [DOI: 10.1007/978-94-007-7192-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
31
|
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
|
32
|
Integrated analysis of long noncoding RNA and coding RNA expression in esophageal squamous cell carcinoma. Int J Genomics 2013; 2013:480534. [PMID: 24222893 PMCID: PMC3814080 DOI: 10.1155/2013/480534] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/26/2013] [Indexed: 01/14/2023] Open
Abstract
Tumorigenesis is a complex dynamic biological process that includes multiple steps of genetic and epigenetic alterations, aberrant expression of noncoding RNA, and changes in the expression profiles of coding genes. We call the collection of those perturbations in genome space the “cancer initiatome.” Long noncoding RNAs (lncRNAs) are pervasively transcribed in the genome and they have key regulatory functions in chromatin remodeling and gene expression. Spatiotemporal variation in the expression of lncRNAs has been observed in development and disease states, including cancer. A few dysregulated lncRNAs have been studied in cancers, but the role of lncRNAs in the cancer initiatome remains largely unknown, especially in esophageal squamous cell carcinoma (ESCC). We conducted a genome-wide screen of the expression of lncRNAs and coding RNAs from ESCC and matched adjacent nonneoplastic normal tissues. We identified differentially expressed lncRNAs and coding RNAs in ESCC relative to their matched normal tissue counterparts and validated the result using polymerase chain reaction analysis. Furthermore, we identified differentially expressed lncRNAs that are co-located and co-expressed with differentially expressed coding RNAs in ESCC and the results point to a potential interaction between lncRNAs and neighboring coding genes that affect ether lipid metabolism, and the interaction may contribute to the development of ESCC. These data provide compelling evidence for a potential novel genomic biomarker of esophageal squamous cell cancer.
Collapse
|
33
|
Shah AK, Saunders NA, Barbour AP, Hill MM. Early diagnostic biomarkers for esophageal adenocarcinoma--the current state of play. Cancer Epidemiol Biomarkers Prev 2013; 22:1185-209. [PMID: 23576690 DOI: 10.1158/1055-9965.epi-12-1415] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is one of the two most common types of esophageal cancer with alarming increase in incidence and very poor prognosis. Aiming to detect EAC early, currently high-risk patients are monitored using an endoscopic-biopsy approach. However, this approach is prone to sampling error and interobserver variability. Diagnostic tissue biomarkers related to genomic and cell-cycle abnormalities have shown promising results, although with current technology these tests are difficult to implement in the screening of high-risk patients for early neoplastic changes. Differential miRNA profiles and aberrant protein glycosylation in tissue samples have been reported to improve performance of existing tissue-based diagnostic biomarkers. In contrast to tissue biomarkers, circulating biomarkers are more amenable to population-screening strategies, due to the ease and low cost of testing. Studies have already shown altered circulating glycans and DNA methylation in BE/EAC, whereas disease-associated changes in circulating miRNA remain to be determined. Future research should focus on identification and validation of these circulating biomarkers in large-scale trials to develop in vitro diagnostic tools to screen population at risk for EAC development.
Collapse
Affiliation(s)
- Alok Kishorkumar Shah
- The University of Queensland Diamantina Institute; and School of Medicine, The University of Queensland, Woolloongabba, Queensland, Australia
| | | | | | | |
Collapse
|
34
|
Pennathur A, Xi L, Litle VR, Gooding WE, Krasinskas A, Landreneau RJ, Godfrey TE, Luketich JD. Gene expression profiles in esophageal adenocarcinoma predict survival after resection. J Thorac Cardiovasc Surg 2013; 145:505-12; discussion 512-3. [PMID: 23321130 DOI: 10.1016/j.jtcvs.2012.10.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 09/28/2012] [Accepted: 10/22/2012] [Indexed: 01/22/2023]
Abstract
OBJECTIVE The incidence of esophageal adenocarcinoma is rapidly increasing in the western population. Despite aggressive treatment, survival after esophagectomy is suboptimal. The main objective of the present study was to evaluate the gene expression profiles in esophageal adenocarcinoma and determine their association with survival after resection. METHODS We conducted a prospective National Institutes of Health/National Cancer Institute funded study to evaluate the prognostic significance of gene expression in patients with esophageal adenocarcinoma undergoing esophagectomy. Gene expression in tumor tissue was analyzed using high-throughput oligonucleotide arrays. The association of gene expression and overall survival was analyzed using the tail-strength statistic and Cox regression analysis. Gene signatures were constructed with semisupervised methods using principal components. A cross-validated risk score was devised by conducting 10-fold cross-validation, 100 times. RESULTS We evaluated the gene expression in 64 patients with esophageal adenocarcinoma who underwent esophagectomy. The median overall survival was 27 months (95% confidence interval 22 to not reached). After filtering, 10,214 probe sets were used for survival analysis. The tail-strength statistic for these probe sets (0.318) indicated a significant association with overall survival. Patients were classified into high- and low-risk groups, according to the gene signature. High-risk patients had a predicted median survival of 19 months, but the median was not reached for the low-risk group (P < .05). On multivariate analysis, the gene signature was independently associated with survival (hazard ratio, 2.22; P = .04). CONCLUSIONS Global gene expression levels were significantly associated with overall survival after esophagectomy. Furthermore, individual genes could be successfully combined into a strongly predictive, internally cross-validated gene signature. If validated further, these results could help direct additional clinical trials of neoadjuvant and adjuvant therapies for esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Arjun Pennathur
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Stiekema J, Boot H, Aleman B, Wessels L, van Sandick J. Prognostication and prediction using gene expression profiling in oesophageal cancer. Eur J Surg Oncol 2013; 39:17-23. [DOI: 10.1016/j.ejso.2012.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/02/2012] [Indexed: 12/27/2022] Open
|
36
|
Wang KK, Tian JM, Gorospe E, Penfield J, Prasad G, Goddard T, Wongkeesong M, Buttar NS, Lutzke L, Krishnadath S. Medical and endoscopic management of high-grade dysplasia in Barrett's esophagus. Dis Esophagus 2012; 25:349-55. [PMID: 22409514 PMCID: PMC4134126 DOI: 10.1111/j.1442-2050.2012.01342.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The management of high-grade dysplasia in Barrett's esophagus has clearly changed over recent years. The risk of cancer development is still substantial, with about one in three patients developing cancer, but a number of patients do not develop cancer. The nature of high-grade dysplasia has also been genetically elucidated with more evidence of chromosomal instability being present at this stage than previously thought. Therapy of the condition has evolved more toward endoscopic therapy, given the good results of radio-frequency ablation and photodynamic therapy in eliminating dysplasia and decreasing cancer development in randomized controlled trial. The best candidates for treatment include compliant patients that have relatively short segments of Barrett's esophagus, an anatomically straight segment, lack of nodularity, and an intact p16. However, even with excellent long-term results similar to surgical resection, the risk of recurrence is present in over 14% of patients, which indicates that there will be a need to continue surveillance endoscopy in these patients.
Collapse
Affiliation(s)
- K K Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55934, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chen H, Fang Y, Tevebaugh W, Orlando RC, Shaheen NJ, Chen X. Molecular mechanisms of Barrett's esophagus. Dig Dis Sci 2011; 56:3405-20. [PMID: 21984436 PMCID: PMC3750118 DOI: 10.1007/s10620-011-1885-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 08/16/2011] [Indexed: 12/11/2022]
Abstract
Barrett's esophagus (BE) is defined as the metaplastic conversion of esophageal squamous epithelium to intestinalized columnar epithelium. As a premalignant lesion of esophageal adenocarcinoma (EAC), BE develops as a result of chronic gastroesophageal reflux disease (GERD). Many studies have been conducted to understand the molecular mechanisms of this disease. This review summarizes recent results involving squamous and intestinal transcription factors, signaling pathways, stromal factors, microRNAs, and other factors in the development of BE. A conceptual framework is proposed to guide future studies. We expect elucidation of the molecular mechanisms of BE to help in the development of improved management of GERD, BE, and EAC.
Collapse
Affiliation(s)
- Hao Chen
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA
| | - Yu Fang
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA
| | - Whitney Tevebaugh
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA
| | - Roy C. Orlando
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7080, USA
| | - Nicholas J. Shaheen
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7080, USA
| | - Xiaoxin Chen
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA,Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7080, USA,Corresponding authors: Xiaoxin Luke Chen, MD, PhD, Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA. Tel: 919-530-6425; Fax: 919-530-7780;
| |
Collapse
|
38
|
Rao S, Welsh L, Cunningham D, te-Poele RH, Benson M, Norman A, Saffery C, Giddings I, Workman P, Clarke PA. Correlation of overall survival with gene expression profiles in a prospective study of resectable esophageal cancer. Clin Colorectal Cancer 2011; 10:48-56. [PMID: 21609936 DOI: 10.3816/ccc.2011.n.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Preoperative chemotherapy has demonstrated a survival benefit for patients with potentially resectable esophageal cancer; however, currently it is not possible to predict the benefit of this treatment for an individual patient. This prospective study was designed to correlate gene expression profiles with clinical outcome in this setting. PATIENTS AND METHODS Eligible patients were deemed to have resectable disease after staging by computed tomography, endoscopic ultrasound, and laparoscopy as indicated and following discussion at the multidisciplinary team meeting. All patients received neoadjuvant platinum and fluoropyrimidine-based chemotherapy; and clinical data were entered prospectively onto a study-specific database. Total RNA was isolated from pretreatment tumor biopsies obtained at baseline endoscopy and analyzed using a cDNA array consisting of 22,000 cDNA clones. RESULTS Of the patients with adequate follow-up accrued between 2002 and 2005, 35 satisfied the quality control measures for the microarray profiling. Median follow-up was 938 days. Supervised hierarchical clustering of normalized data revealed 165 significantly differentially expressed genes based on overall survival (OS; P < .01) with 2 distinct clusters: a poor outcome group: N = 17 (1 year OS 46.2%) and a good outcome group: N = 18 (1 year OS 100%). Genes identified included those previously associated with esophageal cancer and, interestingly, a group of genes encoding proteins involved in the regulation of the TOLL receptor-signaling pathway. CONCLUSION This initial study has highlighted groups of tumors with distinct gene expression profiles based on survival and warrants further validation in a larger cohort. This approach may further our understanding of individual tumor biology and thus facilitate the development of tailored treatment.
Collapse
Affiliation(s)
- Sheela Rao
- Royal Marsden Hospital, London and Surrey, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Nancarrow DJ, Clouston AD, Smithers BM, Gotley DC, Drew PA, Watson DI, Tyagi S, Hayward NK, Whiteman DC. Whole genome expression array profiling highlights differences in mucosal defense genes in Barrett's esophagus and esophageal adenocarcinoma. PLoS One 2011; 6:e22513. [PMID: 21829465 PMCID: PMC3145652 DOI: 10.1371/journal.pone.0022513] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2011] [Accepted: 06/26/2011] [Indexed: 12/15/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) has become a major concern in Western countries due to rapid rises in incidence coupled with very poor survival rates. One of the key risk factors for the development of this cancer is the presence of Barrett's esophagus (BE), which is believed to form in response to repeated gastro-esophageal reflux. In this study we performed comparative, genome-wide expression profiling (using Illumina whole-genome Beadarrays) on total RNA extracted from esophageal biopsy tissues from individuals with EAC, BE (in the absence of EAC) and those with normal squamous epithelium. We combined these data with publically accessible raw data from three similar studies to investigate key gene and ontology differences between these three tissue states. The results support the deduction that BE is a tissue with enhanced glycoprotein synthesis machinery (DPP4, ATP2A3, AGR2) designed to provide strong mucosal defenses aimed at resisting gastro-esophageal reflux. EAC exhibits the enhanced extracellular matrix remodeling (collagens, IGFBP7, PLAU) effects expected in an aggressive form of cancer, as well as evidence of reduced expression of genes associated with mucosal (MUC6, CA2, TFF1) and xenobiotic (AKR1C2, AKR1B10) defenses. When our results are compared to previous whole-genome expression profiling studies keratin, mucin, annexin and trefoil factor gene groups are the most frequently represented differentially expressed gene families. Eleven genes identified here are also represented in at least 3 other profiling studies. We used these genes to discriminate between squamous epithelium, BE and EAC within the two largest cohorts using a support vector machine leave one out cross validation (LOOCV) analysis. While this method was satisfactory for discriminating squamous epithelium and BE, it demonstrates the need for more detailed investigations into profiling changes between BE and EAC.
Collapse
Affiliation(s)
- Derek J Nancarrow
- Oncogenomics, Queensland Institute of Medical Research, Herston, Queensland, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Outcomes after trimodality therapy for esophageal cancer: the impact of histology on failure patterns. Am J Clin Oncol 2011; 34:259-64. [PMID: 20686405 DOI: 10.1097/coc.0b013e3181e841ce] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES This retrospective analysis of patients undergoing neoadjuvant chemoradiation followed by surgical resection was performed to determine if histology or pathologic response affected local-regional control (LRC), survival outcomes or patterns of failure. METHODS We performed a review of 164 patients who underwent neoadjuvant chemoradiation followed by surgical resection from 1992 to 2006 for esophageal cancer. Information on patient characteristics, pathologic response, failure patterns, and survival was collected. Survival was estimated by the Kaplan-Meier method, and Cox multivariable Regression model was used to analyze trends. RESULTS The median follow-up was 18 months and 27 months in surviving patients. The 3-year overall survival (OS) and LRC was 46% and 79%. The overall response for the entire cohort included a pathologic complete response (pCR) rate of 41.4%, 21.3% with microscopic residual disease (mRD) and 36.3% with gross residual disease (gRD). The 3-year OS of patients who achieved a pCR versus mRD versus gRD was 58%, 53%, and 29%. OS was significantly improved in patients with a pCR and mRD compared with gRD (P = 0.001). On multivariate analysis both pCR and mRD correlated with an improved OS. Squamous cell cancers (SCC) had a higher rate of pCR than adenocarcinomas (AC), 54% versus 34.8% (P = 0.01). The 3 year LRC for patients with SCC and AC was 100% and 71% (P = 0.03). Among SCC with recurrence, there were no local failures and all failed distantly (P = 0.001). CONCLUSIONS Patients with microscopic residual disease following trimodality therapy had similar outcomes to patients achieving a pCR. Patients with SCC were more likely to achieve a pCR, and had a higher propensity to fail distantly when compared with patients with AC. This data should be considered in the design of future clinical trials.
Collapse
|
41
|
Holmes K, Egan B, Swan N, O'Morain C. Genetic Mechanisms and Aberrant Gene Expression during the Development of Gastric Intestinal Metaplasia and Adenocarcinoma. Curr Genomics 2011; 8:379-97. [PMID: 19412438 PMCID: PMC2671722 DOI: 10.2174/138920207783406460] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 09/21/2007] [Accepted: 09/28/2007] [Indexed: 02/07/2023] Open
Abstract
Gastric adenocarcinoma occurs via a sequence of molecular events known as the Correa’s Cascade which often progresses over many years. Gastritis, typically caused by infection with the bacterium H. pylori, is the first step of the cascade that results in gastric cancer; however, not all cases of gastritis progress along this carcinogenic route. Despite recent antibiotic intervention of H. pylori infections, gastric adenocarcinoma remains the second most common cause of cancer deaths worldwide. Intestinal metaplasia is the next step along the carcinogenic sequence after gastritis and is considered to be a precursor lesion for gastric cancer; however, not all patients with intestinal metaplasia develop adenocarcinoma and little is known about the molecular and genetic events that trigger the progression of intestinal metaplasia into adenocarcinoma. This review aims to highlight the progress to date in the genetic events involved in intestinal-type gastric adenocarcinoma and its precursor lesion, intestinal metaplasia. The use of technologies such as whole genome microarray analysis, immunohistochemical analysis and DNA methylation analysis has allowed an insight into some of the events which occur in intestinal metaplasia and may be involved in carcinogenesis. There is still much that is yet to be discovered surrounding the development of this lesion and how, in many cases, it develops into a state of malignancy.
Collapse
Affiliation(s)
- K Holmes
- Department of Clinical Medicine, Trinity College Dublin, The Adelaide and Meath Hospital, Tallaght, Dublin 24, Ireland
| | | | | | | |
Collapse
|
42
|
Abstract
The incidence of esophageal adenocarcinoma is increasing in Western countries with a tendency to exceed that of squamous-cell carcinoma. Prognosis is unfavorable with 5-year survival less than 15%, irrespective of treatment and the stage. At the time of diagnosis, more than two thirds of patients have a non-operable cancer because of extension or associated co-morbidities. Most studies have included different tumoral locations (esophagus and stomach) and different histological types (adenocarcinoma and squamous-cell carcinoma), making it difficult to interpret results. Surgery is currently the standard treatment for small tumors. Surgery should be preceded by neo-adjuvant treatment for patients with locally advanced resectable tumors, either preoperative chemotherapy or preoperative chemoradiation therapy. The therapeutic choice should be decided during multidisciplinary meetings according to patient and tumor characteristics and the expertise of the center. For patients with contraindications to surgery, exclusive chemoradiation therapy is recommended. Herein we reviewed and synthesized the different therapeutic strategies for esophageal adenocarcinoma.
Collapse
|
43
|
Rumiato E, Pasello G, Montagna M, Scaini MC, De Salvo GL, Parenti A, Cagol M, Ruol A, Ancona E, Amadori A, Saggioro D. DNA copy number profile discriminates between esophageal adenocarcinoma and squamous cell carcinoma and represents an independent prognostic parameter in esophageal adenocarcinoma. Cancer Lett 2011; 310:84-93. [PMID: 21757289 DOI: 10.1016/j.canlet.2011.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 06/01/2011] [Accepted: 06/12/2011] [Indexed: 11/17/2022]
Abstract
We report multiplex ligation-dependent probe amplification analysis (MLPA) of DNA copy number alterations in 59 esophageal cancer samples, stratified by histotype. Results showed that squamous cell carcinoma (SCC) samples present clustered abnormalities with several genes altered at high frequency. Instead, esophageal adenocarcinoma (ADC) samples are characterized by a more widespread genomic instability, and in these patients total DNA copy number alterations resulted to be an independent prognostic factor. The detection of characteristic molecular changes represents a step towards a better understanding of the molecular basis of esophageal tumorigenesis, and might offer the potential for the discovery of tumor-specific biomarkers.
Collapse
Affiliation(s)
- Enrica Rumiato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV--IRCCS, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tougeron D, Richer JP, Silvain C. Traitement des adénocarcinomes de l’œsophage. JOURNAL DE CHIRURGIE VISCÉRALE 2011; 148:184-195. [DOI: 10.1016/j.jchirv.2011.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
45
|
Abstract
Barrett's esophagus is an acquired metaplastic abnormality in which the normal stratified squamous epithelium lining of the esophagus is replaced by an intestinal-like columnar epithelium. While in itself a benign and asymptomatic disorder, the clinical importance of this relatively common condition relates to its role as a precursor lesion to esophageal adenocarcinoma, the incidence of which has dramatically increased in Western populations in recent years. Although known to arise as a consequence of chronic gastroesophageal reflux, the cellular and molecular mechanisms underlying development Barrett's esophagus and its progression to cancer remain unclear.
Collapse
Affiliation(s)
- Wayne A Phillips
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| | | | | | | | | |
Collapse
|
46
|
Ong CAJ, Lao-Sirieix P, Fitzgerald RC. Biomarkers in Barrett’s esophagus and esophageal adenocarcinoma: Predictors of progression and prognosis. World J Gastroenterol 2010; 16:5669-81. [PMID: 21128316 PMCID: PMC2997982 DOI: 10.3748/wjg.v16.i45.5669] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Barrett’s esophagus is a well-known premalignant lesion of the lower esophagus that is characterized by intestinal metaplasia of the squamous epithelium. It is clinically important due to the increased risk (0.5% per annum) of progression to esophageal adenocarcinoma (EA), which has a poor outcome unless diagnosed early. The current clinical management of Barrett’s esophagus is hampered by the lack of accurate predictors of progression. In addition, when patients develop EA, the current staging modalities are limited in stratifying patients into different prognostic groups in order to guide the optimal therapy for an individual patient. Biomarkers have the potential to improve radically the clinical management of patients with Barrett’s esophagus and EA but have not yet entered mainstream clinical practice. This is in contrast to other cancers like breast and prostate for which biomarkers are utilized routinely to inform clinical decisions. This review aims to highlight the most promising predictive and prognostic biomarkers in Barrett’s esophagus and EA and to discuss what is required to move the field forward towards clinical application.
Collapse
|
47
|
Affiliation(s)
- Matthew J Schuchert
- Heart, Lung, and Esophageal Surgery Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
48
|
Power D, Reynolds J. Localized adenocarcinoma of the esophagogastric junction – Is there a standard of care? Cancer Treat Rev 2010; 36:400-9. [DOI: 10.1016/j.ctrv.2010.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 01/06/2010] [Accepted: 01/08/2010] [Indexed: 12/23/2022]
|
49
|
Souza RF. Biomarkers in Barrett's Esophagus. TECHNIQUES IN GASTROINTESTINAL ENDOSCOPY 2010; 12:116-1212. [PMID: 20657812 DOI: 10.1016/j.tgie.2010.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Biomarkers are substances that can be used to indicate normal or diseased states. In clinical settings, the term biomarker generally refers to a molecular marker produced by tissues whose detection heralds a diseased state. For patients with Barrett's esophagus, there are at least three clinical settings in which the use of molecular biomarkers has been proposed including 1) stratifying the risk of neoplastic progression, 2) serving as an adjunct to aid in the diagnosis of dysplasia, and 3) predicting response to ablative therapies. Although the routine clinical use of biomarkers in any of these clinical settings is not yet recommended, it seems reasonable to assume that biomarker validation studies will be carried out in the coming years and that movement into the clinics will be inevitable. This article reviews the current progress in using biomarkers in each of the clinical settings described above with a focus on the molecular biomarkers which have advanced the farthest toward use in routine clinical practice.
Collapse
Affiliation(s)
- Rhonda F Souza
- Departments of Medicine, VA North Texas Health Care System and the University of Texas Southwestern Medical School, and the Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas
| |
Collapse
|
50
|
Zinovyeva MV, Monastyrskaya GS, Kopantzev EP, Vinogradova TV, Kostina MB, Sass AV, Filyukova OB, Uspenskaya NY, Sukhikh GT, Sverdlov ED. Identification of some human genes oppositely regulated during esophageal squamous cell carcinoma formation and human embryonic esophagus development. Dis Esophagus 2010; 23:260-70. [PMID: 19732125 DOI: 10.1111/j.1442-2050.2009.01008.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Here we directly compared gene expression profiles in human esophageal squamous cell carcinomas and in human fetal esophagus development. We used the suppression subtractive hybridization technique to subtract cDNAs prepared from tumor and normal human esophageal samples. cDNA sequencing and reverse transcription polymerase chain reaction (RT-PCR) analysis of RNAs from human tumor and the normal esophagus revealed 10 differentially transcribed genes: CSTA, CRNN, CEACAM1, MAL, EMP1, ECRG2, and SPRR downregulated, and PLAUR, SFRP4, and secreted protein that is acidic and rich in cysteine upregulated in tumor tissue as compared with surrounding normal tissue. In turn, genes up- and downregulated in tumor tissue were down- and upregulated, respectively, during development from the fetal to adult esophagus. Thus, we demonstrated that, as reported for other tumors, gene transcriptional activation and/or suppression events in esophageal tumor progression were opposite to those observed during development from the fetal to adult esophagus. This tumor 'embryonization' supports the idea that stem or progenitor cells are implicated in esophageal cancer emergence.
Collapse
Affiliation(s)
- M V Zinovyeva
- Laboratory of Structure and Functions of Human Genes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|