501
|
Luo H, Li H, Hu Z, Wu H, Liu C, Li Y, Zhang X, Lin P, Hou Q, Ding G, Wang Y, Li S, Wei D, Qiu F, Li Y, Wu S. Noninvasive diagnosis and monitoring of mutations by deep sequencing of circulating tumor DNA in esophageal squamous cell carcinoma. Biochem Biophys Res Commun 2016; 471:596-602. [PMID: 26876573 DOI: 10.1016/j.bbrc.2016.02.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 02/04/2016] [Indexed: 12/17/2022]
Abstract
Circulating tumor DNA (ctDNA) is becoming an important biomarker in noninvasive diagnosis and monitoring of tumor dynamics. This study tested the feasibility of plasma ctDNA for the non-invasive analysis of tumor mutations in esophageal squamous cell carcinoma (ESCC) by sequencing of tumor, tumor-adjacent, and normal tissue, as well as pre-surgery and post-surgery plasma. Exome sequencing of eight patients identified between 29 and 134 somatic mutations in ESCCs, many of which were also determined in ctDNA. Comparison of pre-surgery and post-surgery plasma has shown that mutations had reduced frequency or disappeared after surgery treatment. We further evaluated the TruSight Cancer sequencing panel by using it to detect mutations in the plasma of three patients. Tumor mutations were only found in one of them. To design a sequencing panel with improved targeting, we identified significantly mutated genes by meta-analysis of 532 ESCC genomes. Our results confirmed the well-known driver genes and found several uncharacterized genes. The new panel consisted of 90 recurrent genes, which theoretically achieved 94% and 75% of sensitivity when detecting at least 1 and 2 mutant genes in ESCC patients, respectively. Our results demonstrate the feasibility of using ctDNA to detect ESCCs and monitor treatment effect. The low-cost and sensitive target panel could facilitate clinical usage of ctDNA as a noninvasive biomarker.
Collapse
Affiliation(s)
- Honglei Luo
- Department of Radiotherapy, Huai'an First People's Hospital, Huai'an, China
| | - Hong Li
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhaoyang Hu
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China
| | - Hongjin Wu
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China
| | - Chenglin Liu
- School of Life Sciences and Biotechnology, Shanghai Jiaotong University, Shanghai, China
| | - Ying Li
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China
| | - Xiaoyan Zhang
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China
| | - Ping Lin
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Hou
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China
| | - Guohui Ding
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Wang
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China
| | - Shuang Li
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China
| | - Dongkai Wei
- Basepair Biotechnology, Co.,Ltd, Suzhou, China
| | - Feng Qiu
- Basepair Biotechnology, Co.,Ltd, Suzhou, China
| | - Yixue Li
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Shixiu Wu
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China.
| |
Collapse
|
502
|
Dobashi A, Tsuyama N, Asaka R, Togashi Y, Ueda K, Sakata S, Baba S, Sakamoto K, Hatake K, Takeuchi K. Frequent BCOR aberrations in extranodal NK/T-Cell lymphoma, nasal type. Genes Chromosomes Cancer 2016; 55:460-71. [PMID: 26773734 DOI: 10.1002/gcc.22348] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/08/2016] [Accepted: 01/09/2016] [Indexed: 12/11/2022] Open
Abstract
Extranodal natural killer/T cell lymphoma (ENKTL) is a rare subtype of lymphoma. Recurrent mutations in the JAK-STAT pathway, recently reported in ENKTL cases, are interesting in terms of both pathogenesis and inhibitor therapy. However, the frequencies of these mutations are low and variable among reports, and other pathognomonic mutations in ENKTL remain to be elucidated. In the present study, targeted capture sequencing of 602 cancer-related genes from 25 frozen ENKTL samples was performed, 11 of which were matched to normal samples. Several recurrent somatic mutations involving BCOR (32%), TP53 (16%), DDX3X (12%), FAT4 (8%), NRAS (8%), MLL3 (12%), and MIR17HG (8%) were identified. The pattern of BCOR aberrations (1 nonsense and 5 frame-shift mutations, a mutation leading to a splicing error, and gene loss) suggested that loss of function of BCOR was the functionally important outcome of such changes. The literature was reviewed and the public data on BCOR aberrations was reanalyzed and it was found that the aberrations were frequently found in myeloid neoplasms, but, interestingly, were highly specific to ENKTL among lymphoid malignancies. Given the high frequency and pattern of aberration, BCOR is likely to play an important role in ENKTL pathogenesis as a tumor suppressor gene.
Collapse
Affiliation(s)
- Akito Dobashi
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Naoko Tsuyama
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Reimi Asaka
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo.,Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Yuki Togashi
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo.,Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Kyoko Ueda
- Hematology and Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Seiji Sakata
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Satoko Baba
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Kana Sakamoto
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Kiyohiko Hatake
- Hematology and Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto, Tokyo
| | - Kengo Takeuchi
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo.,Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo
| |
Collapse
|
503
|
Hu N, Kadota M, Liu H, Abnet CC, Su H, Wu H, Freedman ND, Yang HH, Wang C, Yan C, Wang L, Gere S, Hutchinson A, Song G, Wang Y, Ding T, Qiao YL, Koshiol J, Dawsey SM, Giffen C, Goldstein AM, Taylor PR, Lee MP. Genomic Landscape of Somatic Alterations in Esophageal Squamous Cell Carcinoma and Gastric Cancer. Cancer Res 2016; 76:1714-23. [PMID: 26857264 DOI: 10.1158/0008-5472.can-15-0338] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 12/08/2015] [Indexed: 01/01/2023]
Abstract
Gastric cancer and esophageal cancer are the second and sixth leading causes of cancer-related death worldwide. Multiple genomic alterations underlying gastric cancer and esophageal squamous cell carcinoma (ESCC) have been identified, but the full spectrum of genomic structural variations and mutations have yet to be uncovered. Here, we report the results of whole-genome sequencing of 30 samples comprising tumor and blood from 15 patients, four of whom presented with ESCC, seven with gastric cardia adenocarcinoma (GCA), and four with gastric noncardia adenocarcinoma. Analyses revealed that an A>C mutation was common in GCA, and in addition to the preferential nucleotide sequence of A located 5 prime to the mutation as noted in previous studies, we found enrichment of T in the 5 prime base. The A>C mutations in GCA suggested that oxidation of guanine may be a potential mechanism underlying cancer mutagenesis. Furthermore, we identified genes with mutations in gastric cancer and ESCC, including well-known cancer genes, TP53, JAK3, BRCA2, FGF2, FBXW7, MSH3, PTCH, NF1, ERBB2, and CHEK2, and potentially novel cancer-associated genes, KISS1R, AMH, MNX1, WNK2, and PRKRIR Finally, we identified recurrent chromosome alterations in at least 30% of tumors in genes, including MACROD2, FHIT, and PARK2 that were often intragenic deletions. These structural alterations were validated using the The Cancer Genome Atlas dataset. Our studies provide new insights into understanding the genomic landscape, genome instability, and mutation profile underlying gastric cancer and ESCC development. Cancer Res; 76(7); 1714-23. ©2016 AACR.
Collapse
Affiliation(s)
- Nan Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Mitsutaka Kadota
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Huaitian Liu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Hua Su
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Hailong Wu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Howard H Yang
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Chaoyu Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Chunhua Yan
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Lemin Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Sheryl Gere
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland. Cancer Genomics Research Laboratory, Leidos, Gaithersburg, Maryland
| | - Guohong Song
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Yuan Wang
- Shanxi Cancer Hospital, Taiyuan, Shanxi, PR China
| | - Ti Ding
- Shanxi Cancer Hospital, Taiyuan, Shanxi, PR China
| | - You-Lin Qiao
- Cancer Institute, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Jill Koshiol
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Sanford M Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Carol Giffen
- Information Management Services, Inc., Silver Spring, Maryland
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Philip R Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland.
| | - Maxwell P Lee
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
504
|
Whole-Genome Sequencing Reveals Diverse Models of Structural Variations in Esophageal Squamous Cell Carcinoma. Am J Hum Genet 2016; 98:256-74. [PMID: 26833333 PMCID: PMC4746371 DOI: 10.1016/j.ajhg.2015.12.013] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 12/15/2015] [Indexed: 01/02/2023] Open
Abstract
Comprehensive identification of somatic structural variations (SVs) and understanding their mutational mechanisms in cancer might contribute to understanding biological differences and help to identify new therapeutic targets. Unfortunately, characterization of complex SVs across the whole genome and the mutational mechanisms underlying esophageal squamous cell carcinoma (ESCC) is largely unclear. To define a comprehensive catalog of somatic SVs, affected target genes, and their underlying mechanisms in ESCC, we re-analyzed whole-genome sequencing (WGS) data from 31 ESCCs using Meerkat algorithm to predict somatic SVs and Patchwork to determine copy-number changes. We found deletions and translocations with NHEJ and alt-EJ signature as the dominant SV types, and 16% of deletions were complex deletions. SVs frequently led to disruption of cancer-associated genes (e.g., CDKN2A and NOTCH1) with different mutational mechanisms. Moreover, chromothripsis, kataegis, and breakage-fusion-bridge (BFB) were identified as contributing to locally mis-arranged chromosomes that occurred in 55% of ESCCs. These genomic catastrophes led to amplification of oncogene through chromothripsis-derived double-minute chromosome formation (e.g., FGFR1 and LETM2) or BFB-affected chromosomes (e.g., CCND1, EGFR, ERBB2, MMPs, and MYC), with approximately 30% of ESCCs harboring BFB-derived CCND1 amplification. Furthermore, analyses of copy-number alterations reveal high frequency of whole-genome duplication (WGD) and recurrent focal amplification of CDCA7 that might act as a potential oncogene in ESCC. Our findings reveal molecular defects such as chromothripsis and BFB in malignant transformation of ESCCs and demonstrate diverse models of SVs-derived target genes in ESCCs. These genome-wide SV profiles and their underlying mechanisms provide preventive, diagnostic, and therapeutic implications for ESCCs.
Collapse
|
505
|
Yang CS, Chen X, Tu S. Etiology and Prevention of Esophageal Cancer. Gastrointest Tumors 2016; 3:3-16. [PMID: 27722152 DOI: 10.1159/000443155] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 12/07/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Esophageal cancer (EC) occurs commonly, especially in Asia, and is the sixth leading cause of cancer deaths worldwide. Recently, great progress has been made in research on the etiology and prevention of EC. SUMMARY The major risk factors for esophageal squamous cell carcinoma (ESCC) are tobacco smoking and alcohol drinking, which act synergistically. Dietary parameters, including dietary carcinogens and insufficiency of micronutrients, could also be important risk factors in certain areas. A common etiological factor for both EC and some other cancers are low levels of intake of fruits and vegetables. With improvements in diet and drinking water in developing countries, the incidence of ESCC decreased. However, in economically well-developed countries, the incidence of esophageal adenocarcinoma (EAC) has markedly increased in the past 40 years. The major etiological factor for EAC is gastroesophageal reflux, which is also an etiological factor for gastric cardia adenocarcinoma (GCA). In certain areas of China, the occurrence of GCA is closely related to ESCC. Susceptibility genes for EC are starting to be discovered, and this may help to identify high-risk groups that have more need for preventive measures. Mitigation of the risk factors, early detection and treatment of precancerous lesions are effective approaches for prevention. Smoking cessation, avoidance of excessive alcohol, meat and caloric consumption, increasing physical activity and frequent consumption of vegetables and fruits are prudent lifestyle modifications for the prevention of EC as well as other diseases. KEY MESSAGE The etiology of EC includes tobacco smoking, alcohol drinking, low levels of intake of fruits and vegetables as well as gastroesophageal reflux and susceptibility genes. PRACTICAL IMPLICATIONS A healthy lifestyle including smoking cessation, increasing physical activity, consumption of vegetables as well as reduction of alcohol intake and caloric consumption are major approaches to the prevention of EC.
Collapse
Affiliation(s)
- Chung S Yang
- Department of Chemical Biology, Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, N.J, USA
| | - Xiaoxin Chen
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, N.C, USA; Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, N.C., USA
| | - Shuiping Tu
- Department of Oncology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| |
Collapse
|
506
|
Nakazato H, Takeshima H, Kishino T, Kubo E, Hattori N, Nakajima T, Yamashita S, Igaki H, Tachimori Y, Kuniyoshi Y, Ushijima T. Early-Stage Induction of SWI/SNF Mutations during Esophageal Squamous Cell Carcinogenesis. PLoS One 2016; 11:e0147372. [PMID: 26812616 PMCID: PMC4728064 DOI: 10.1371/journal.pone.0147372] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/04/2016] [Indexed: 01/26/2023] Open
Abstract
The SWI/SNF chromatin remodeling complex is frequently inactivated by somatic mutations of its various components in various types of cancers, and also by aberrant DNA methylation. However, its somatic mutations and aberrant methylation in esophageal squamous cell carcinomas (ESCCs) have not been fully analyzed. In this study, we aimed to clarify in ESCC, what components of the SWI/SNF complex have somatic mutations and aberrant methylation, and when somatic mutations of the SWI/SNF complex occur. Deep sequencing of components of the SWI/SNF complex using a bench-top next generation sequencer revealed that eight of 92 ESCCs (8.7%) had 11 somatic mutations of 7 genes, ARID1A, ARID2, ATRX, PBRM1, SMARCA4, SMARCAL1, and SMARCC1. The SMARCA4 mutations were located in the Forkhead (85Ser>Leu) and SNF2 family N-terminal (882Glu>Lys) domains. The PBRM1 mutations were located in a bromodomain (80Asn>Ser) and an HMG-box domain (1,377Glu>Lys). For most mutations, their mutant allele frequency was 31–77% (mean 61%) of the fraction of cancer cells in the same samples, indicating that most of the cancer cells in individual ESCC samples had the SWI/SNF mutations on one allele, when present. In addition, a BeadChip array analysis revealed that a component of the SWI/SNF complex, ACTL6B, had aberrant methylation at its promoter CpG island in 18 of 52 ESCCs (34.6%). These results showed that genetic and epigenetic alterations of the SWI/SNF complex are present in ESCCs, and suggested that genetic alterations are induced at an early stage of esophageal squamous cell carcinogenesis.
Collapse
Affiliation(s)
- Hidetsugu Nakazato
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- Esophageal Surgery Division, National Cancer Center Hospital, Tokyo, Japan
- Department of Thoracic and Cardiovascular Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hideyuki Takeshima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Takayoshi Kishino
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Emi Kubo
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Naoko Hattori
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Takeshi Nakajima
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | - Satoshi Yamashita
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroyasu Igaki
- Esophageal Surgery Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yuji Tachimori
- Esophageal Surgery Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yukio Kuniyoshi
- Department of Thoracic and Cardiovascular Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Toshikazu Ushijima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- * E-mail:
| |
Collapse
|
507
|
Wang H, Jiang D, Song Q, Xu C, Shi Y, Li X, Huang J, Xu Y, Sujie A, Zeng H, Zhong Y, Tan L, Hou Y. Prognostic impact and potential interaction of EGFR and c-Met in the progression of esophageal squamous cell carcinoma. Tumour Biol 2016; 37:9771-9. [PMID: 26810066 DOI: 10.1007/s13277-015-4692-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 12/16/2015] [Indexed: 12/17/2022] Open
Abstract
This study is to examine EGFR and c-Met variation in precancerous lesion, early esophageal squamous cell carcinoma (ESCC), and advanced ESCC and to explore their prognostic significance. EGFR and c-Met were detected by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH). Of 158 endoscopy resection (ER) specimens, c-Met high expression and FISH positive were 44.9 and 12.6 %, respectively. EGFR high expression and FISH positive were 2.5 and 19.6 %, respectively. Of 84 surgical specimens, c-Met high expression and FISH positive were 50 and 8.3 %, respectively. EGFR high expression and FISH positive were 7.1 and 28.5 %, respectively. A significant correlation was observed between c-Met and EGFR FISH positive both in ER (P < 0.001) and surgical specimens (P = 0.029). Patients with EGFR high expression had poorer disease-free survival (DFS) and overall survival (OS) (P = 0.031 and P = 0.013) in c-Met high-expression group but not in c-Met low-expression group (P = 0.301 and P = 0.439). C-Met FISH positive did not represent a statistically significant adverse prognosis until 24 months later (P = 0.027 and 0.048). EGFR and c-Met might be involved in the tumorigenesis and development of ESCC. EGFR high expression has different prognostic significance in patients with differing c-Met expression status. C-Met FISH positive represent delayed prognostic factor.
Collapse
Affiliation(s)
- Haixing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yuan Shi
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaojing Li
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yifan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Akesu Sujie
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Haiying Zeng
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Yunshi Zhong
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
508
|
Kishino T, Niwa T, Yamashita S, Takahashi T, Nakazato H, Nakajima T, Igaki H, Tachimori Y, Suzuki Y, Ushijima T. Integrated analysis of DNA methylation and mutations in esophageal squamous cell carcinoma. Mol Carcinog 2016; 55:2077-2088. [PMID: 26756304 DOI: 10.1002/mc.22452] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 12/01/2015] [Accepted: 12/15/2015] [Indexed: 12/12/2022]
Abstract
The recent development of next-generation sequencing technology for extensive mutation analysis, and beadarray technology for genome-wide DNA methylation analysis has made it possible to obtain integrated pictures of genetic and epigenetic alterations, using the same cancer samples. In this study, we aimed to characterize such a picture in esophageal squamous cell carcinomas (ESCCs). Base substitutions of 55 cancer-related genes and copy number alterations (CNAs) of 28 cancer-related genes were analyzed by targeted sequencing. Forty-four of 57 ESCCs (77%) had 64 non-synonymous somatic mutations, and 24 ESCCs (42%) had 35 CNAs. A genome-wide DNA methylation analysis using an Infinium HumanMethylation450 BeadChip array showed that the CpG island methylator phenotype was unlikely to be present in ESCCs, a different situation from gastric and colon cancers. Regarding individual pathways affected in ESCCs, the WNT pathway was activated potentially by aberrant methylation of its negative regulators, such as SFRP1, SFRP2, SFRP4, SFRP5, SOX17, and WIF1 (33%). The p53 pathway was inactivated by TP53 mutations (70%), and potentially by aberrant methylation of its downstream genes. The cell cycle was deregulated by mutations of CDKN2A (9%), deletions of CDKN2A and RB1 (32%), and by aberrant methylation of CDKN2A and CHFR (9%). In conclusion, ESCCs had unique methylation profiles different from gastric and colon cancers. The genes involved in the WNT pathway were affected mainly by epigenetic alterations, and those involved in the p53 pathway and cell cycle regulation were affected mainly by genetic alterations. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Takayoshi Kishino
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan.,Division of Esophageal Surgery, National Cancer Center Hospital, Tokyo, Japan.,Department of Gastroenterological Surgery, Kagawa University, Kagawa, Japan
| | - Tohru Niwa
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Satoshi Yamashita
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Takamasa Takahashi
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan.,Division of Esophageal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Hidetsugu Nakazato
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan.,Division of Esophageal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Takeshi Nakajima
- Division of Endoscopy, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyasu Igaki
- Division of Esophageal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yuji Tachimori
- Division of Esophageal Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuyuki Suzuki
- Department of Gastroenterological Surgery, Kagawa University, Kagawa, Japan
| | - Toshikazu Ushijima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
509
|
Jones S, Anagnostou V, Lytle K, Parpart-Li S, Nesselbush M, Riley DR, Shukla M, Chesnick B, Kadan M, Papp E, Galens KG, Murphy D, Zhang T, Kann L, Sausen M, Angiuoli SV, Diaz LA, Velculescu VE. Personalized genomic analyses for cancer mutation discovery and interpretation. Sci Transl Med 2016; 7:283ra53. [PMID: 25877891 DOI: 10.1126/scitranslmed.aaa7161] [Citation(s) in RCA: 312] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Massively parallel sequencing approaches are beginning to be used clinically to characterize individual patient tumors and to select therapies based on the identified mutations. A major question in these analyses is the extent to which these methods identify clinically actionable alterations and whether the examination of the tumor tissue alone is sufficient or whether matched normal DNA should also be analyzed to accurately identify tumor-specific (somatic) alterations. To address these issues, we comprehensively evaluated 815 tumor-normal paired samples from patients of 15 tumor types. We identified genomic alterations using next-generation sequencing of whole exomes or 111 targeted genes that were validated with sensitivities >95% and >99%, respectively, and specificities >99.99%. These analyses revealed an average of 140 and 4.3 somatic mutations per exome and targeted analysis, respectively. More than 75% of cases had somatic alterations in genes associated with known therapies or current clinical trials. Analyses of matched normal DNA identified germline alterations in cancer-predisposing genes in 3% of patients with apparently sporadic cancers. In contrast, a tumor-only sequencing approach could not definitively identify germline changes in cancer-predisposing genes and led to additional false-positive findings comprising 31% and 65% of alterations identified in targeted and exome analyses, respectively, including in potentially actionable genes. These data suggest that matched tumor-normal sequencing analyses are essential for precise identification and interpretation of somatic and germline alterations and have important implications for the diagnostic and therapeutic management of cancer patients.
Collapse
Affiliation(s)
- Siân Jones
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | - Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Karli Lytle
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | | | | | - David R Riley
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | - Manish Shukla
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | | | - Maura Kadan
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | - Eniko Papp
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Derek Murphy
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | - Theresa Zhang
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | - Lisa Kann
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | - Mark Sausen
- Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | | | - Luis A Diaz
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
510
|
Cheng C, Cui H, Zhang L, Jia Z, Song B, Wang F, Li Y, Liu J, Kong P, Shi R, Bi Y, Yang B, Wang J, Zhao Z, Zhang Y, Hu X, Yang J, He C, Zhao Z, Wang J, Xi Y, Xu E, Li G, Guo S, Chen Y, Yang X, Chen X, Liang J, Guo J, Cheng X, Wang C, Zhan Q, Cui Y. Genomic analyses reveal FAM84B and the NOTCH pathway are associated with the progression of esophageal squamous cell carcinoma. Gigascience 2016; 5:1. [PMID: 26759717 PMCID: PMC4709967 DOI: 10.1186/s13742-015-0107-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/23/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is the sixth most lethal cancer worldwide and the fourth most lethal cancer in China. Genomic characterization of tumors, particularly those of different stages, is likely to reveal additional oncogenic mechanisms. Although copy number alterations and somatic point mutations associated with the development of ESCC have been identified by array-based technologies and genome-wide studies, the genomic characterization of ESCCs from different stages of the disease has not been explored. Here, we have performed either whole-genome sequencing or whole-exome sequencing on 51 stage I and 53 stage III ESCC patients to characterize the genomic alterations that occur during the various clinical stages of ESCC, and further validated these changes in 36 atypical hyperplasia samples. RESULTS Recurrent somatic amplifications at 8q were found to be enriched in stage I tumors and the deletions of 4p-q and 5q were particularly identified in stage III tumors. In particular, the FAM84B gene was amplified and overexpressed in preclinical and ESCC tumors. Knockdown of FAM84B in ESCC cell lines significantly reduced in vitro cell growth, migration and invasion. Although the cancer-associated genes TP53, PIK3CA, CDKN2A and their pathways showed no significant difference between stage I and stage III tumors, we identified and validated a prevalence of mutations in NOTCH1 and in the NOTCH pathway that indicate that they are involved in the preclinical and early stages of ESCC. CONCLUSIONS Our results suggest that FAM84B and the NOTCH pathway are involved in the progression of ESCC and may be potential diagnostic targets for ESCC susceptibility.
Collapse
Affiliation(s)
- Caixia Cheng
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Heyang Cui
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Ling Zhang
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Zhiwu Jia
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Bin Song
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Department of Oncology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Fang Wang
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Yaoping Li
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001 China
| | - Jing Liu
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Department of General Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Pengzhou Kong
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Ruyi Shi
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Yanghui Bi
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Bin Yang
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001 China
| | - Juan Wang
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Zhenxiang Zhao
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Yanyan Zhang
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Department of General Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Xiaoling Hu
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Jie Yang
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Chanting He
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Zhiping Zhao
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Jinfen Wang
- />Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001 China
| | - Yanfeng Xi
- />Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001 China
| | - Enwei Xu
- />Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001 China
| | - Guodong Li
- />Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001 China
| | - Shiping Guo
- />Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001 China
| | - Yunqing Chen
- />Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001 China
| | - Xiaofeng Yang
- />Department of Urology Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Xing Chen
- />Department of Endoscopy, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi 030001 China
| | - Jianfang Liang
- />Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Jiansheng Guo
- />Department of General Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Xiaolong Cheng
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| | - Chuangui Wang
- />Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620 China
| | - Qimin Zhan
- />Cancer Institute and Cancer Hospital, State Key Laboratory of Molecular Oncology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 China
| | - Yongping Cui
- />Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001 China
- />Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001 China
| |
Collapse
|
511
|
Kim HS, Kim SM, Kim H, Pyo KH, Sun JM, Ahn MJ, Park K, Keam B, Kwon NJ, Yun HJ, Kim HG, Chung IJ, Lee JS, Lee KH, Kim DJ, Lee CG, Hur J, Chung H, Park JC, Shin SK, Lee SK, Kim HR, Moon YW, Lee YC, Kim JH, Paik S, Cho BC. Phase II clinical and exploratory biomarker study of dacomitinib in recurrent and/or metastatic esophageal squamous cell carcinoma. Oncotarget 2015; 6:44971-84. [PMID: 26462025 PMCID: PMC4792605 DOI: 10.18632/oncotarget.6056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/23/2015] [Indexed: 12/14/2022] Open
Abstract
The purpose of this study was to investigate the clinical activity, safety and predictive biomarkers of dacomitinib, an irreversible pan-HER inhibitor, in patients with recurrent or metastatic esophageal squamous cell carcinoma (R/M-ESCC). Patients, whose diseases were not amenable to curative treatment and had progressed on platinum-based chemotherapy, were treated with dacomitinib 45 mg/day. The primary endpoint was objective response rate by RECISTv 1.1. Predictive biomarker analyses included the characterization of somatic mutations and gene expression using the Ion Torrent AmpliSeq Cancer Hotspot Panel and Nanostring nCounter, and investigation of their relationship with clinical outcomes. Of the 48 evaluable patients, 6 (12.5%) achieved partial responses and 29 (60.4%) had stable disease. The median response duration was 7.1 months. The median progression free survival (PFS) and overall survival (OS) was 3.3 months (95% CI, 2.4-4.3 months) and 6.4 months (95% CI, 4.4-8.4 months). Adverse events were mostly grade 1-2. Gene set enrichment analysis revealed that ERBB signaling pathway is significantly enriched in patients with PFS ≥ 4 months (n = 12) than PFS < 4 months (n = 21) (p < 0.001). Upregulation of ERBB signaling pathway was significantly associated with longer PFS (5.0 vs. 2.9 months, P = 0.016) and OS (10.0 vs. 4.8 months, P = 0.022). The most frequent mutations were TP53 (61%) followed by CDKN2A (8%), MLH1 (8%), FLT3 (8%) and EGFR (8%). Dacomitinib demonstrated clinical efficacy with manageable toxicity in platinum-failed R/M-ESCC. Screening of ERBB pathway-related gene expression profiles may help identify patients who are most likely benefit from dacomitinib.
Collapse
Affiliation(s)
- Hyo Song Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Moo Kim
- Yonsei Cancer Research Institute, JE-UK Laboratory of Molecular Cancer Therapeutics, Seoul, Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Kyoung-Ho Pyo
- Yonsei Cancer Research Institute, JE-UK Laboratory of Molecular Cancer Therapeutics, Seoul, Korea
| | - Jong-Mu Sun
- Department of Hematology-Oncology, Samsung Medical Center, Seoul, Korea
| | - Myung-Ju Ahn
- Department of Hematology-Oncology, Samsung Medical Center, Seoul, Korea
| | - Keunchil Park
- Department of Hematology-Oncology, Samsung Medical Center, Seoul, Korea
| | - Bhumsuk Keam
- Department of Hematology-Oncology, Seoul National University Hospital, Seoul, Korea
| | | | - Hwan Jung Yun
- Division of Hemato-Oncology, Chungnam National University Hospital, Daejeon, Korea
| | - Hoon-Gu Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Gyeongnam Regional Cancer Center, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Ik-Joo Chung
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Jong Seok Lee
- Department of Hematology-Oncology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyung Hee Lee
- Department of Hematology-Oncology, Yeungnam University Medical Center, Daegu, South Korea
| | - Dae Joon Kim
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Chang-Geol Lee
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Hur
- Department of Radiology, Yonsei University College of Medicine, Seoul, Korea
| | - Hyunsoo Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jun Chul Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Kwan Shin
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Kil Lee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Wha Moon
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Chan Lee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Joo Hang Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Soonmyung Paik
- Division of Pathology NSABP, Pittsburgh, PA, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
512
|
Warnecke-Eberz U, Metzger R, Hölscher AH, Drebber U, Bollschweiler E. Diagnostic marker signature for esophageal cancer from transcriptome analysis. Tumour Biol 2015; 37:6349-58. [PMID: 26631031 DOI: 10.1007/s13277-015-4400-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/05/2015] [Indexed: 12/12/2022] Open
Abstract
Esophageal cancer is often diagnosed at an advanced stage. Diagnostic markers are needed for achieving a cure in esophageal cancer detecting and treating tumor cells earlier. In patients with locally advanced squamous cell carcinoma of the esophagus (ESCC), we profiled the gene expression of ESCC compared to corresponding normal biopsies for diagnostic markers by genome microarrays. Profiling of gene expression identified 4844 genes differentially expressed, 2122 upregulated and 2722 downregulated in ESCC. Twenty-three overexpressed candidates with best scores from significance analysis have been selected for further analysis by TaqMan low-density array-technique using a validation cohort of 40 patients. The verification rate was 100 % for ESCC. Twenty-two markers were additionally overexpressed in adenocarcinoma of the esophagus (EAC). The markers significantly overexpressed already in earlier tumor stages (pT1-2) of both histological subtypes (n = 19) have been clustered in a "diagnostic signature": PLA2G7, PRAME, MMP1, MMP3, MMP12, LIlRB2, TREM2, CHST2, IGFBP2, IGFBP7, KCNJ8, EMILIN2, CTHRC1, EMR2, WDR72, LPCAT1, COL4A2, CCL4, and SNX10. The marker signature will be translated to clinical practice to prove its diagnostic impact. This diagnostic signature may contribute to the earlier detection of tumor cells, with the aim to complement clinical techniques resulting in the development of better detection of concepts of esophageal cancer for earlier therapy and more favorite prognosis.
Collapse
Affiliation(s)
- Ute Warnecke-Eberz
- Laboratory for Molecular Oncology, General, Visceral and Cancer Surgery, University Hospital of Cologne (CIO), Kerpener Straße 62, 50937, Cologne, Germany.
| | - Ralf Metzger
- Caritasklinikum Saarbrücken, Rheinstraße 2, 66113, Saarbrücken, Germany
| | - Arnulf H Hölscher
- General, Visceral and Cancer Surgery, University Hospital of Cologne (CIO), Kerpener Straße 62, 50937, Cologne, Germany
| | - Uta Drebber
- Institute for Pathology, University Hospital of Cologne, Center for Integrated Oncology (CIO), Kerpener Straße 62, 50937, Cologne, Germany
| | - Elfriede Bollschweiler
- General, Visceral and Cancer Surgery, University Hospital of Cologne (CIO), Kerpener Straße 62, 50937, Cologne, Germany
| |
Collapse
|
513
|
Ohashi S, Miyamoto S, Kikuchi O, Goto T, Amanuma Y, Muto M. Recent Advances From Basic and Clinical Studies of Esophageal Squamous Cell Carcinoma. Gastroenterology 2015; 149:1700-15. [PMID: 26376349 DOI: 10.1053/j.gastro.2015.08.054] [Citation(s) in RCA: 393] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/17/2015] [Accepted: 08/17/2015] [Indexed: 02/08/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive squamous cell carcinomas and is highly prevalent in Asia. Alcohol and its metabolite, acetaldehyde, are considered definite carcinogens for the esophagus. Polymorphisms in the aldehyde dehydrogenase 2 gene, which encodes an enzyme that eliminates acetaldehyde, have been associated with esophageal carcinogenesis. Studies of the mutagenic and carcinogenic effects of acetaldehyde support this observation. Several recent large-scale comprehensive analyses of the genomic alterations in ESCC have shown a high frequency of mutations in genes such as TP53 and others that regulate the cell cycle or cell differentiation. Moreover, whole genome and whole exome sequencing studies have frequently detected somatic mutations, such as G:C→A:T transitions or G:C→C:G transversions, in ESCC tissues. Genomic instability, caused by abnormalities in the Fanconi anemia DNA repair pathway, is also considered a pathogenic mechanism of ESCC. Advances in diagnostic techniques such as magnifying endoscopy with narrow band imaging or positron emission tomography have increased the accuracy of diagnosis of ESCC. Updated guidelines from the National Comprehensive Cancer Network standardize the practice for the diagnosis and treatment of esophageal cancer. Patients with ESCC are treated endoscopically or with surgery, chemotherapy, or radiotherapy, based on tumor stage. Minimally invasive treatments help improve the quality of life of patients who undergo such treatments. We review recent developments in the diagnosis and treatment of ESCC and advances gained from basic and clinical research.
Collapse
Affiliation(s)
- Shinya Ohashi
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Clinical Oncology, Kyoto University Hospital Cancer Center, Kyoto, Japan
| | - Shin'ichi Miyamoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Osamu Kikuchi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoyuki Goto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yusuke Amanuma
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Clinical Oncology, Kyoto University Hospital Cancer Center, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Clinical Oncology, Kyoto University Hospital Cancer Center, Kyoto, Japan.
| |
Collapse
|
514
|
Cao W, Wu W, Yan M, Tian F, Ma C, Zhang Q, Li X, Han P, Liu Z, Gu J, Biddle FG. Multiple region whole-exome sequencing reveals dramatically evolving intratumor genomic heterogeneity in esophageal squamous cell carcinoma. Oncogenesis 2015; 4:e175. [PMID: 26619400 PMCID: PMC4670960 DOI: 10.1038/oncsis.2015.34] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/02/2015] [Accepted: 10/08/2015] [Indexed: 12/13/2022] Open
Abstract
Cancer is a disease of genome instability and genomic alterations; now, genomic heterogeneity is rapidly emerging as a defining feature of cancer, both within and between tumors. Motivation for our pilot study of tumor heterogeneity in esophageal squamous cell carcinoma (ESCC) is that it is not well studied, but the highest incidences of esophageal cancers are found in China and ESCC is the most common type. We profiled the mutations and changes in copy number that were identified by whole-exome sequencing and array-based comparative genomic hybridization in multiple regions within an ESCC from two patients. The average mutational heterogeneity rate was 90% in all regions of the individual tumors in each patient; most somatic point mutations were nonsynonymous substitutions, small Indels occurred in untranslated regions of genes, and copy number alterations varied among multiple regions of a tumor. Independent Sanger sequencing technology confirmed selected gene mutations with more than 88% concordance. Phylogenetic analysis of the somatic mutation frequency demonstrated that multiple, genomically heterogeneous divergent clones evolve and co-exist within a primary ESCC and metastatic subclones result from the dispersal and adaptation of an initially non-metastatic parental clone. Therefore, a single-region sampling will not reflect the evolving architecture of a genomically heterogeneous landscape of mutations in ESCC tumors and the divergent complexity of this genomic heterogeneity among patients will complicate any promise of a simple genetic or epigenetic diagnostic signature in ESCC. We conclude that any potential for informative biomarker discovery in ESCC and targeted personalized therapies will require a deeper understanding of the functional biology of the ontogeny and phylogeny of the tumor heterogeneity.
Collapse
Affiliation(s)
- W Cao
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - W Wu
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China.,Department of Pathology and Laboratory Medicine, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary Alberta, Canada
| | - M Yan
- Medical School, Zhengzhou University, Zhengzhou, China
| | - F Tian
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - C Ma
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Q Zhang
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - X Li
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - P Han
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Z Liu
- Linzhou Cancer Hospital, Linzhou, China
| | - J Gu
- Science and Education Department, Health Bureau of Zhengzhou, Zhengzhou, China
| | - F G Biddle
- Departments of Medical Genetics and Biological Sciences, University of Calgary, Calgary, Canada
| |
Collapse
|
515
|
Yang H, Schramek D, Adam RC, Keyes BE, Wang P, Zheng D, Fuchs E. ETS family transcriptional regulators drive chromatin dynamics and malignancy in squamous cell carcinomas. eLife 2015; 4:e10870. [PMID: 26590320 PMCID: PMC4739765 DOI: 10.7554/elife.10870] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/20/2015] [Indexed: 01/08/2023] Open
Abstract
Tumor-initiating stem cells (SCs) exhibit distinct patterns of transcription factors and gene expression compared to healthy counterparts. Here, we show that dramatic shifts in large open-chromatin domain (super-enhancer) landscapes underlie these differences and reflect tumor microenvironment. By in vivo super-enhancer and transcriptional profiling, we uncover a dynamic cancer-specific epigenetic network selectively enriched for binding motifs of a transcription factor cohort expressed in squamous cell carcinoma SCs (SCC-SCs). Many of their genes, including Ets2 and Elk3, are themselves regulated by SCC-SC super-enhancers suggesting a cooperative feed-forward loop. Malignant progression requires these genes, whose knockdown severely impairs tumor growth and prohibits progression from benign papillomas to SCCs. ETS2-deficiency disrupts the SCC-SC super-enhancer landscape and downstream cancer genes while ETS2-overactivation in epidermal-SCs induces hyperproliferation and SCC super-enhancer-associated genes Fos, Junb and Klf5. Together, our findings unearth an essential regulatory network required for the SCC-SC chromatin landscape and unveil its importance in malignant progression. DOI:http://dx.doi.org/10.7554/eLife.10870.001 Many cancers contain a mixture of different types of cells. Of these, cells known as cancer stem cells can form new tumours and drive the growth and spread of the cancer around the body. A central question is how cancer stem cells differ from healthy adult stem cells. Recent evidence suggests that, in addition to having genetic mutations, cancer stem cells live in a very different environment to other cells within the tumour. This 'microenvironment'also has a major impact on how these cells behave compared to normal stem cells. Together, the genetic and environmental differences profoundly change the way genes are expressed in the cancer cells. In 2013, a group of researchers identified regions of DNA called super-enhancers. These regions are long stretches of DNA that proteins called transcription factors can interact with to coordinate the expression of nearby genes to alter the production of certain proteins. Super-enhancers contain several transcription factor-binding sites that are close to each other with the different sites being associated with transcription factors that are only active in specific types of cells. Furthermore, super-enhancers are often self-regulatory, meaning that the binding of transcription factors to a super-enhancer can lead to an increase in the expression of the genes that encode the same transcription factors. Yang, Schramek et al. have now identified the super-enhancers in a skin cancer called squamous cell carcinoma and showed that they differ dramatically from the super-enhancers of normal skin stem cells. Their experiments show that the active super-enhancers in cancer stem cells are associated with a very different set of genes that are highly and often specifically expressed in cancer stem cells. In the cancer stem cells, a transcription factor called ETS2 binds to the super-enhancers and reprograms the expression of genes to promote the development of cancer. Yang, Schramek et al. also show that over-active ETS2 is a major driver of squamous cell carcinoma. Furthermore, ETS2 also increases the expression of genes that cause inflammation and promote the growth of cancers. Yang, Schramek et al.’s findings reveal a new regulatory network that governs the expression of genes involved in cancer. Furthermore, the experiments show that high levels of ETS2 are linked with poor outcomes for patients with head and neck squamous cell carcinoma, which is one of the most life-threatening cancers world-wide. In the future, these findings might lead to the development of new therapies to treat these cancers. DOI:http://dx.doi.org/10.7554/eLife.10870.002
Collapse
Affiliation(s)
- Hanseul Yang
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, United States
| | - Daniel Schramek
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, United States
| | - Rene C Adam
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, United States
| | - Brice E Keyes
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, United States
| | - Ping Wang
- Department of Neurology, Albert Einstein College of Medicine, New York, United States
| | - Deyou Zheng
- Department of Neurology, Albert Einstein College of Medicine, New York, United States.,Departments of Genetics and Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, United States
| |
Collapse
|
516
|
Cho HY, Marzec J, Kleeberger SR. Functional polymorphisms in Nrf2: implications for human disease. Free Radic Biol Med 2015; 88:362-372. [PMID: 26117318 PMCID: PMC6779133 DOI: 10.1016/j.freeradbiomed.2015.06.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 12/18/2022]
Abstract
Nuclear factor (erythroid derived)-2 like 2 (NFE2L2), also known as nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2), is a ubiquitous transcription factor essential for protecting cells and tissues from oxidative stress-induced injury. Positional cloning and studies with Nrf2 knockout mice have identified important roles for this transcription factor in disease phenotypes for many organ systems. Studies have also characterized the means through which human Nrf2 is regulated and the mechanisms of interaction with antioxidant response elements (ARE) in promoters of effector genes. Moreover, single nucleotide polymorphisms (SNPs) in Nrf2 have been identified and evaluated for effects on gene expression and function, and translational investigations have sought to determine whether loss of function SNPs associate with disease progression. In this review, we present 1) an overview of the human Nrf2 gene and protein domain, 2) identification of genetic mutations in Nrf2 and associations of the mutations with multiple diseases, and 3) the role of somatic mutations in Nrf2 in diseases, primarily various cancers.
Collapse
Affiliation(s)
- Hye-Youn Cho
- Inflammation, Immunity, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Jacqui Marzec
- Inflammation, Immunity, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Steven R Kleeberger
- Inflammation, Immunity, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
517
|
Profiling of somatic mutations in acute myeloid leukemia with FLT3-ITD at diagnosis and relapse. Blood 2015; 126:2491-501. [PMID: 26438511 DOI: 10.1182/blood-2015-05-646240] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/22/2015] [Indexed: 01/15/2023] Open
Abstract
Acute myeloid leukemia (AML) with an FLT3 internal tandem duplication (FLT3-ITD) mutation is an aggressive hematologic malignancy with a grave prognosis. To identify the mutational spectrum associated with relapse, whole-exome sequencing was performed on 13 matched diagnosis, relapse, and remission trios followed by targeted sequencing of 299 genes in 67 FLT3-ITD patients. The FLT3-ITD genome has an average of 13 mutations per sample, similar to other AML subtypes, which is a low mutation rate compared with that in solid tumors. Recurrent mutations occur in genes related to DNA methylation, chromatin, histone methylation, myeloid transcription factors, signaling, adhesion, cohesin complex, and the spliceosome. Their pattern of mutual exclusivity and cooperation among mutated genes suggests that these genes have a strong biological relationship. In addition, we identified mutations in previously unappreciated genes such as MLL3, NSD1, FAT1, FAT4, and IDH3B. Mutations in 9 genes were observed in the relapse-specific phase. DNMT3A mutations are the most stable mutations, and this DNMT3A-transformed clone can be present even in morphologic complete remissions. Of note, all AML matched trio samples shared at least 1 genomic alteration at diagnosis and relapse, suggesting common ancestral clones. Two types of clonal evolution occur at relapse: either the founder clone recurs or a subclone of the founder clone escapes from induction chemotherapy and expands at relapse by acquiring new mutations. Relapse-specific mutations displayed an increase in transversions. Functional assays demonstrated that both MLL3 and FAT1 exert tumor-suppressor activity in the FLT3-ITD subtype. An inhibitor of XPO1 synergized with standard AML induction chemotherapy to inhibit FLT3-ITD growth. This study clearly shows that FLT3-ITD AML requires additional driver genetic alterations in addition to FLT3-ITD alone.
Collapse
|
518
|
Colombo M, Galletti S, Garavelli S, Platonova N, Paoli A, Basile A, Taiana E, Neri A, Chiaramonte R. Notch signaling deregulation in multiple myeloma: A rational molecular target. Oncotarget 2015; 6:26826-40. [PMID: 26308486 PMCID: PMC4694956 DOI: 10.18632/oncotarget.5025] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/03/2015] [Indexed: 12/16/2022] Open
Abstract
Despite recent therapeutic advances, multiple myeloma (MM) is still an incurable neoplasia due to intrinsic or acquired resistance to therapy. Myeloma cell localization in the bone marrow milieu allows direct interactions between tumor cells and non-tumor bone marrow cells which promote neoplastic cell growth, survival, bone disease, acquisition of drug resistance and consequent relapse. Twenty percent of MM patients are at high-risk of treatment failure as defined by tumor markers or presentation as plasma cell leukemia. Cumulative evidences indicate a key role of Notch signaling in multiple myeloma onset and progression. Unlike other Notch-related malignancies, where the majority of patients carry gain-of-function mutations in Notch pathway members, in MM cell Notch signaling is aberrantly activated due to an increased expression of Notch receptors and ligands; notably, this also results in the activation of Notch signaling in surrounding stromal cells which contributes to myeloma cell proliferation, survival and migration, as well as to bone disease and intrinsic and acquired pharmacological resistance. Here we review the last findings on the mechanisms and the effects of Notch signaling dysregulation in MM and provide a rationale for a therapeutic strategy aiming at inhibiting Notch signaling, along with a complete overview on the currently available Notch-directed approaches.
Collapse
Affiliation(s)
- Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy
| | - Serena Galletti
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano; Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milano, Italy
| | - Silvia Garavelli
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy
| | - Natalia Platonova
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano; Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milano, Italy
| | - Alessandro Paoli
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy
| | - Andrea Basile
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy
| | - Elisa Taiana
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano; Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milano, Italy
| | - Antonino Neri
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano; Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milano, Italy
| | - Raffaella Chiaramonte
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milano, Italy
| |
Collapse
|
519
|
Wang Q, Bai J, Abliz A, Liu Y, Gong K, Li J, Shi W, Pan Y, Liu F, Lai S, Yang H, Lu C, Zhang L, Chen W, Xu R, Cai H, Ke Y, Zeng C. An Old Story Retold: Loss of G1 Control Defines A Distinct Genomic Subtype of Esophageal Squamous Cell Carcinoma. GENOMICS PROTEOMICS & BIOINFORMATICS 2015; 13:258-70. [PMID: 26386145 PMCID: PMC4610972 DOI: 10.1016/j.gpb.2015.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/03/2015] [Accepted: 06/19/2015] [Indexed: 01/12/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) has a high mortality rate. To determine the molecular basis of ESCC development, this study sought to identify characteristic genome-wide alterations in ESCC, including exonic mutations and structural alterations. The clinical implications of these genetic alterations were also analyzed. Exome sequencing and verification were performed for nine pairs of ESCC and the matched blood samples, followed by validation with additional samples using Sanger sequencing. Whole-genome SNP arrays were employed to detect copy number alteration (CNA) and loss of heterozygosity (LOH) in 55 cases, including the nine ESCC samples subjected to exome sequencing. A total of 108 non-synonymous somatic mutations (NSSMs) in 102 genes were verified in nine patients. The chromatin modification process was found to be enriched in our gene ontology (GO) analysis. Tumor genomes with TP53 mutations were significantly more unstable than those without TP53 mutations. In terms of the landscape of genomic alterations, deletion of 9p21.3 covering CDKN2A/2B (30.9%), amplification of 11q13.3 covering CCND1 (30.9%), and TP53 point mutation (50.9%) occurred in two-thirds of the cases. These results suggest that the deregulation of the G1 phase during the cell cycle is a key event in ESCC. Furthermore, six minimal common regions were found to be significantly altered in ESCC samples and three of them, 9p21.3, 7p11.2, and 3p12.1, were associated with lymph node metastasis. With the high correlation of TP53 mutation and genomic instability in ESCC, the amplification of CCND1, the deletion of CDKN2A/2B, and the somatic mutation of TP53 appear to play pivotal roles via G1 deregulation and therefore helps to classify this cancer into different genomic subtypes. These findings provide clinical significance that could be useful in future molecular diagnoses and therapeutic targeting.
Collapse
Affiliation(s)
- Qiyan Wang
- MOE Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Genetics, Peking University, Cancer Hospital & Institute, Beijing 100142, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jian Bai
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Amir Abliz
- MOE Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Genetics, Peking University, Cancer Hospital & Institute, Beijing 100142, China
| | - Ying Liu
- MOE Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Genetics, Peking University, Cancer Hospital & Institute, Beijing 100142, China
| | - Kenan Gong
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingjing Li
- MOE Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Genetics, Peking University, Cancer Hospital & Institute, Beijing 100142, China
| | - Wenjie Shi
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yaqi Pan
- MOE Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Genetics, Peking University, Cancer Hospital & Institute, Beijing 100142, China
| | - Fangfang Liu
- MOE Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Genetics, Peking University, Cancer Hospital & Institute, Beijing 100142, China
| | - Shujuan Lai
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Haijun Yang
- Anyang Cancer Hospital, Anyang 455000, China
| | | | - Lixin Zhang
- Anyang Cancer Hospital, Anyang 455000, China
| | - Wei Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ruiping Xu
- Anyang Cancer Hospital, Anyang 455000, China
| | - Hong Cai
- MOE Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Genetics, Peking University, Cancer Hospital & Institute, Beijing 100142, China.
| | - Yang Ke
- MOE Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Genetics, Peking University, Cancer Hospital & Institute, Beijing 100142, China.
| | - Changqing Zeng
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
520
|
Wang K, Johnson A, Ali SM, Klempner SJ, Bekaii-Saab T, Vacirca JL, Khaira D, Yelensky R, Chmielecki J, Elvin JA, Lipson D, Miller VA, Stephens PJ, Ross JS. Comprehensive Genomic Profiling of Advanced Esophageal Squamous Cell Carcinomas and Esophageal Adenocarcinomas Reveals Similarities and Differences. Oncologist 2015; 20:1132-9. [PMID: 26336083 DOI: 10.1634/theoncologist.2015-0156] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/25/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinomas (ESCCs) and esophageal adenocarcinomas (EACs) account for >95% of esophageal malignancies and represent a major global health burden. ESCC is the dominant histology globally but represents a minority of U.S. cases, with EAC accounting for the majority of U.S. CASES The patient outcomes for advanced ESCC and EAC are poor, and new therapeutic options are needed. Using a sensitive sequencing assay, we compared the genomic profiles of ESCC and EAC with attention to identification of therapeutically relevant genomic alterations. METHODS Next-generation sequencing-based comprehensive genomic profiling was performed on hybridization-captured, adaptor ligation-based libraries to a median coverage depth of >650× for all coding exons of 315 cancer-related genes plus selected introns from 28 genes frequently rearranged in cancer. Results from a single sample were evaluated for all classes of genomic alterations (GAs) including point mutations, short insertions and deletions, gene amplifications, homozygous deletions, and fusions/rearrangements. Clinically relevant genomic alterations (CRGAs) were defined as alterations linked to approved drugs and those under evaluation in mechanism-driven clinical trials. RESULTS There were no significant differences by sex for either tumor type, and the median age for all patients was 63 years. All ESCCs and EACs were at an advanced stage at the time of sequencing. All 71 ESCCs and 231 EACs featured GAs on profiling, with 522 GAs in ESCC (7.4 per sample) and 1,303 GAs in EAC (5.6 per sample). The frequency of clinically relevant GAs in ESCC was 94% (2.6 per sample) and 93% in EAC (2.7 per sample). CRGAs occurring more frequently in EAC included KRAS (23% EAC vs. 6% ESCC) and ERBB2 (23% EAC vs. 3% ESCC). ESCC samples were enriched for CRGA in PIK3CA (24% ESCC vs. 10% EAC), PTEN (11% ESCC vs. 4% EAC), and NOTCH1 (17% ESCC vs. 3% EAC). Other GAs that differed significantly between histologic tumor types included SMAD4 (14% EAC vs. 1% ESCC), RB1 (14% ESCC vs. 2% EAC), SOX2 (18% ESCC vs. 1% EAC), and NFE2L2 (24% ESCC vs. 1% EAC). CONCLUSION ESCC and EAC share similarly high frequencies of overall and clinically relevant genomic alterations; however, the profiles of genomic alterations in the two diseases differ widely, with KRAS and ERBB2 far more frequently altered in EAC compared with ESCC and with mammalian target of rapamycin (MTOR) pathway genes (PIK3CA and PTEN) and NOTCH1 more frequently altered in ESCC compared with EAC. Comprehensive genomic profiling highlights the promise of identifying clinically relevant genomic alterations in both ESCC and EAC and suggests new avenues for molecularly directed therapies in esophageal cancer.
Collapse
Affiliation(s)
- Kai Wang
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Adrienne Johnson
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Siraj M Ali
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Samuel J Klempner
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Tanios Bekaii-Saab
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Jeffrey L Vacirca
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Depinder Khaira
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Roman Yelensky
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Juliann Chmielecki
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Julia A Elvin
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Doron Lipson
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Vincent A Miller
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Philip J Stephens
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA; Division of Hematology-Oncology, University of California Irvine, Orange, California, USA; The Ohio State University-James Cancer Hospital, Columbus, Ohio, USA; National Translational Research Group, New York, New York, USA; Albany Medical College, Albany, New York, USA
| |
Collapse
|
521
|
Tétreault MP. Esophageal Cancer: Insights From Mouse Models. CANCER GROWTH AND METASTASIS 2015; 8:37-46. [PMID: 26380556 PMCID: PMC4558891 DOI: 10.4137/cgm.s21218] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/15/2015] [Accepted: 07/17/2015] [Indexed: 12/30/2022]
Abstract
Esophageal cancer is the eighth leading cause of cancer and the sixth most common cause of cancer-related death worldwide. Despite recent advances in the development of surgical techniques in combination with the use of radiotherapy and chemotherapy, the prognosis for esophageal cancer remains poor. The cellular and molecular mechanisms that drive the pathogenesis of esophageal cancer are still poorly understood. Hence, understanding these mechanisms is crucial to improving outcomes for patients with esophageal cancer. Mouse models constitute valuable tools for modeling human cancers and for the preclinical testing of therapeutic strategies in a manner not possible in human subjects. Mice are excellent models for studying human cancers because they are similar to humans at the physiological and molecular levels and because they have a shorter gestation time and life cycle. Moreover, a wide range of well-developed technologies for introducing genetic modifications into mice are currently available. In this review, we describe how different mouse models are used to study esophageal cancer.
Collapse
Affiliation(s)
- Marie-Pier Tétreault
- Department of Medicine, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
522
|
Rosekrans SL, Baan B, Muncan V, van den Brink GR. Esophageal development and epithelial homeostasis. Am J Physiol Gastrointest Liver Physiol 2015; 309:G216-28. [PMID: 26138464 DOI: 10.1152/ajpgi.00088.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/25/2015] [Indexed: 01/31/2023]
Abstract
The esophagus is a relatively simple organ that evolved to transport food and liquids through the thoracic cavity. It is the only part of the gastrointestinal tract that lacks any metabolic, digestive, or absorptive function. The mucosa of the adult esophagus is covered by a multilayered squamous epithelium with a remarkable similarity to the epithelium of the skin despite the fact that these tissues originate from two different germ layers. Here we review the developmental pathways involved in the establishment of the esophagus and the way these pathways regulate gut-airway separation. We summarize current knowledge of the mechanisms that maintain homeostasis in esophageal epithelial renewal in the adult and the molecular mechanism of the development of Barrett's metaplasia, the precursor lesion to esophageal adenocarcinoma. Finally, we examine the ongoing debate on the hierarchy of esophageal epithelial precursor cells and on the presence or absence of a specific esophageal stem cell population. Together the recent insights into esophageal development and homeostasis suggest that the pathways that establish the esophagus during development also play a role in the maintenance of the adult epithelium. We are beginning to understand how reflux of gastric content and the resulting chronic inflammation can transform the squamous esophageal epithelium to columnar intestinal type metaplasia in Barrett's esophagus.
Collapse
Affiliation(s)
- Sanne L Rosekrans
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, the Netherlands
| | - Bart Baan
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, the Netherlands
| | - Vanesa Muncan
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, the Netherlands
| | - Gijs R van den Brink
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
523
|
Mori T, Sumii M, Fujishima F, Ueno K, Emi M, Nagasaki M, Ishioka C, Chiba N. Somatic alteration and depleted nuclear expression of BAP1 in human esophageal squamous cell carcinoma. Cancer Sci 2015; 106:1118-29. [PMID: 26081045 PMCID: PMC4582980 DOI: 10.1111/cas.12722] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 05/27/2015] [Accepted: 06/11/2015] [Indexed: 02/02/2023] Open
Abstract
BRCA1-associated protein 1 (BAP1) is a deubiquitinating enzyme that is involved in the regulation of cell growth. Recently, many somatic and germline mutations of BAP1 have been reported in a broad spectrum of tumors. In this study, we identified a novel somatic non-synonymous BAP1 mutation, a phenylalanine-to-isoleucine substitution at codon 170 (F170I), in 1 of 49 patients with esophageal squamous cell carcinoma (ESCC). Multiplex ligation-dependent probe amplification (MLPA) of BAP1 gene in this ESCC tumor disclosed monoallelic deletion (LOH), suggesting BAP1 alterations on both alleles in this tumor. The deubiquitinase activity and the auto-deubiquitinase activity of F170I-mutant BAP1 were markedly suppressed compared with wild-type BAP1. In addition, wild-type BAP1 mostly localizes to the nucleus, whereas the F170I mutant preferentially localized in the cytoplasm. Microarray analysis revealed that expression of the F170I mutant drastically altered gene expression profiles compared with expressed wild-type BAP1. Gene-ontology analyses indicated that the F170I mutation altered the expression of genes involved in oncogenic pathways. We found that one candidate, TCEAL7, previously reported as a putative tumor suppressor gene, was significantly induced by wild-type BAP1 as compared to F170I mutant BAP1. Furthermore, we found that the level of BAP1 expression in the nucleus was reduced in 44% of ESCC examined by immunohistochemistry (IHC). Because the nuclear localization of BAP1 is important for its tumor suppressor function, BAP1 may be functionally inactivated in a substantial portion of ESCC. Taken together, BAP1 is likely to function as a tumor suppressor in at least a part of ESCC.
Collapse
Affiliation(s)
- Takahiro Mori
- Tohoku Community Cancer Services Program, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Makiko Sumii
- Tohoku Community Cancer Services Program, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Kazuko Ueno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Mitsuru Emi
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Chikashi Ishioka
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Natsuko Chiba
- Department of Cancer Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
524
|
Dahle-Smith A, Petty RD. Biomarkers and novel agents in esophago-gastric cancer: are we making progress? Expert Rev Anticancer Ther 2015; 15:1103-19. [DOI: 10.1586/14737140.2015.1071669] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
525
|
Kang X, Chen K, Li Y, Li J, D'Amico TA, Chen X. Personalized targeted therapy for esophageal squamous cell carcinoma. World J Gastroenterol 2015; 21:7648-58. [PMID: 26167067 PMCID: PMC4491954 DOI: 10.3748/wjg.v21.i25.7648] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/19/2015] [Accepted: 04/28/2015] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma continues to heavily burden clinicians worldwide. Researchers have discovered the genomic landscape of esophageal squamous cell carcinoma, which holds promise for an era of personalized oncology care. One of the most pressing problems facing this issue is to improve the understanding of the newly available genomic data, and identify the driver-gene mutations, pathways, and networks. The emergence of a legion of novel targeted agents has generated much hope and hype regarding more potent treatment regimens, but the accuracy of drug selection is still arguable. Other problems, such as cancer heterogeneity, drug resistance, exceptional responders, and side effects, have to be surmounted. Evolving topics in personalized oncology, such as interpretation of genomics data, issues in targeted therapy, research approaches for targeted therapy, and future perspectives, will be discussed in this editorial.
Collapse
|
526
|
|
527
|
Matsumoto T, Shimizu T, Takai A, Marusawa H. Exploring the Mechanisms of Gastrointestinal Cancer Development Using Deep Sequencing Analysis. Cancers (Basel) 2015; 7:1037-51. [PMID: 26083936 PMCID: PMC4491698 DOI: 10.3390/cancers7020823] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/08/2015] [Indexed: 12/17/2022] Open
Abstract
Next-generation sequencing (NGS) technologies have revolutionized cancer genomics due to their high throughput sequencing capacity. Reports of the gene mutation profiles of various cancers by many researchers, including international cancer genome research consortia, have increased over recent years. In addition to detecting somatic mutations in tumor cells, NGS technologies enable us to approach the subject of carcinogenic mechanisms from new perspectives. Deep sequencing, a method of optimizing the high throughput capacity of NGS technologies, allows for the detection of genetic aberrations in small subsets of premalignant and/or tumor cells in noncancerous chronically inflamed tissues. Genome-wide NGS data also make it possible to clarify the mutational signatures of each cancer tissue by identifying the precise pattern of nucleotide alterations in the cancer genome, providing new information regarding the mechanisms of tumorigenesis. In this review, we highlight these new methods taking advantage of NGS technologies, and discuss our current understanding of carcinogenic mechanisms elucidated from such approaches.
Collapse
Affiliation(s)
- Tomonori Matsumoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Takahiro Shimizu
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Atsushi Takai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Hiroyuki Marusawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
528
|
Lee JJ, Sholl LM, Lindeman NI, Granter SR, Laga AC, Shivdasani P, Chin G, Luke JJ, Ott PA, Hodi FS, Mihm MC, Lin JY, Werchniak AE, Haynes HA, Bailey N, Liu R, Murphy GF, Lian CG. Targeted next-generation sequencing reveals high frequency of mutations in epigenetic regulators across treatment-naïve patient melanomas. Clin Epigenetics 2015; 7:59. [PMID: 26221190 PMCID: PMC4517542 DOI: 10.1186/s13148-015-0091-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/27/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Recent developments in genomic sequencing have advanced our understanding of the mutations underlying human malignancy. Melanoma is a prototype of an aggressive, genetically heterogeneous cancer notorious for its biologic plasticity and predilection towards developing resistance to targeted therapies. Evidence is rapidly accumulating that dysregulated epigenetic mechanisms (DNA methylation/demethylation, histone modification, non-coding RNAs) may play a central role in the pathogenesis of melanoma. Therefore, we sought to characterize the frequency and nature of mutations in epigenetic regulators in clinical, treatment-naïve, patient melanoma specimens obtained from one academic institution. RESULTS Targeted next-generation sequencing for 275 known and investigative cancer genes (of which 41 genes, or 14.9 %, encoded an epigenetic regulator) of 38 treatment-naïve patient melanoma samples revealed that 22.3 % (165 of 740) of all non-silent mutations affected an epigenetic regulator. The most frequently mutated genes were BRAF, MECOM, NRAS, TP53, MLL2, and CDKN2A. Of the 40 most commonly mutated genes, 12 (30.0 %) encoded epigenetic regulators, including genes encoding enzymes involved in histone modification (MECOM, MLL2, SETD2), chromatin remodeling (ARID1B, ARID2), and DNA methylation and demethylation (TET2, IDH1). Among the 38 patient melanoma samples, 35 (92.1 %) harbored at least one mutation in an epigenetic regulator. The genes with the highest number of total UVB-signature mutations encoded epigenetic regulators, including MLL2 (100 %, 16 of 16) and MECOM (82.6 %, 19 of 23). Moreover, on average, epigenetic genes harbored a significantly greater number of UVB-signature mutations per gene than non-epigenetic genes (3.7 versus 2.4, respectively; p = 0.01). Bioinformatics analysis of The Cancer Genome Atlas (TCGA) melanoma mutation dataset also revealed a frequency of mutations in the 41 epigenetic genes comparable to that found within our cohort of patient melanoma samples. CONCLUSIONS Our study identified a high prevalence of somatic mutations in genes encoding epigenetic regulators, including those involved in DNA demethylation, histone modification, chromatin remodeling, and microRNA processing. Moreover, UVB-signature mutations were found more commonly among epigenetic genes than in non-epigenetic genes. Taken together, these findings further implicate epigenetic mechanisms, particularly those involving the chromatin-remodeling enzyme MECOM/EVI1 and histone-modifying enzyme MLL2, in the pathobiology of melanoma.
Collapse
Affiliation(s)
- Jonathan J. Lee
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Lynette M. Sholl
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Neal I. Lindeman
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Scott R. Granter
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Alvaro C. Laga
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Priyanka Shivdasani
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Gary Chin
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Jason J. Luke
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Patrick A. Ott
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - F. Stephen Hodi
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Martin C. Mihm
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Jennifer Y. Lin
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Andrew E. Werchniak
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Harley A. Haynes
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Nancy Bailey
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Robert Liu
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - George F. Murphy
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Christine G. Lian
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| |
Collapse
|
529
|
Cancer drug development in China: recent advances and future challenges. Drug Discov Today 2015; 20:766-71. [DOI: 10.1016/j.drudis.2014.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 10/29/2014] [Accepted: 11/13/2014] [Indexed: 12/17/2022]
|
530
|
Zhang L, Zhou Y, Cheng C, Cui H, Cheng L, Kong P, Wang J, Li Y, Chen W, Song B, Wang F, Jia Z, Li L, Li Y, Yang B, Liu J, Shi R, Bi Y, Zhang Y, Wang J, Zhao Z, Hu X, Yang J, Li H, Gao Z, Chen G, Huang X, Yang X, Wan S, Chen C, Li B, Tan Y, Chen L, He M, Xie S, Li X, Zhuang X, Wang M, Xia Z, Luo L, Ma J, Dong B, Zhao J, Song Y, Ou Y, Li E, Xu L, Wang J, Xi Y, Li G, Xu E, Liang J, Yang X, Guo J, Chen X, Zhang Y, Li Q, Liu L, Li Y, Zhang X, Yang H, Lin D, Cheng X, Guo Y, Wang J, Zhan Q, Cui Y. Genomic analyses reveal mutational signatures and frequently altered genes in esophageal squamous cell carcinoma. Am J Hum Genet 2015; 96:597-611. [PMID: 25839328 PMCID: PMC4385186 DOI: 10.1016/j.ajhg.2015.02.017] [Citation(s) in RCA: 270] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/26/2015] [Indexed: 02/07/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers worldwide and the fourth most lethal cancer in China. However, although genomic studies have identified some mutations associated with ESCC, we know little of the mutational processes responsible. To identify genome-wide mutational signatures, we performed either whole-genome sequencing (WGS) or whole-exome sequencing (WES) on 104 ESCC individuals and combined our data with those of 88 previously reported samples. An APOBEC-mediated mutational signature in 47% of 192 tumors suggests that APOBEC-catalyzed deamination provides a source of DNA damage in ESCC. Moreover, PIK3CA hotspot mutations (c.1624G>A [p.Glu542Lys] and c.1633G>A [p.Glu545Lys]) were enriched in APOBEC-signature tumors, and no smoking-associated signature was observed in ESCC. In the samples analyzed by WGS, we identified focal (<100 kb) amplifications of CBX4 and CBX8. In our combined cohort, we identified frequent inactivating mutations in AJUBA, ZNF750, and PTCH1 and the chromatin-remodeling genes CREBBP and BAP1, in addition to known mutations. Functional analyses suggest roles for several genes (CBX4, CBX8, AJUBA, and ZNF750) in ESCC. Notably, high activity of hedgehog signaling and the PI3K pathway in approximately 60% of 104 ESCC tumors indicates that therapies targeting these pathways might be particularly promising strategies for ESCC. Collectively, our data provide comprehensive insights into the mutational signatures of ESCC and identify markers for early diagnosis and potential therapeutic targets.
Collapse
Affiliation(s)
- Ling Zhang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yong Zhou
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Caixia Cheng
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of Pathology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Heyang Cui
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China; BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Le Cheng
- BGI-Yunnan, Kunming, Yunnan 650000, China
| | - Pengzhou Kong
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | | | - Yin Li
- Department of Thoracic Surgery, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | | | - Bin Song
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Fang Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhiwu Jia
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Lin Li
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Yaoping Li
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001, China
| | - Bin Yang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001, China
| | - Jing Liu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of General Surgery, First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ruyi Shi
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yanghui Bi
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yanyan Zhang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of General Surgery, First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Juan Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhenxiang Zhao
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaoling Hu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jie Yang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Hongyi Li
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhibo Gao
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Gang Chen
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | | | - Xukui Yang
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | | | - Chao Chen
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Bin Li
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Yongkai Tan
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | | | - Minghui He
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Sha Xie
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | | | | | | | - Zhi Xia
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Longhai Luo
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Jie Ma
- Department of Molecular Pathology, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Bing Dong
- Department of Molecular Pathology, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Jiuzhou Zhao
- Department of Molecular Pathology, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yunwei Ou
- State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Enming Li
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Liyan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Jinfen Wang
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001, China
| | - Yanfeng Xi
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001, China
| | - Guodong Li
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001, China
| | - Enwei Xu
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi 030001, China
| | - Jianfang Liang
- Department of Pathology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaofeng Yang
- Department of Urology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jiansheng Guo
- Department of General Surgery, First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xing Chen
- Department of Endoscopy, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030001, China
| | - Yanbo Zhang
- Department of Statistics, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Qingshan Li
- School of Pharmaceutical Sciences, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Lixin Liu
- Experimental Center of Science and Research, First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yingrui Li
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | | | | | - Dongxin Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaolong Cheng
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yongjun Guo
- Department of Molecular Pathology, Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Jun Wang
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Yongping Cui
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
531
|
A versatile orthotopic nude mouse model for study of esophageal squamous cell carcinoma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:910715. [PMID: 25834829 PMCID: PMC4365327 DOI: 10.1155/2015/910715] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/20/2015] [Indexed: 12/29/2022]
Abstract
Increasing evidence indicates tumor-stromal interactions play a crucial role in cancer. An in vivo esophageal squamous cell carcinoma (ESCC) orthotopic animal model was developed with bioluminescence imaging established with a real-time monitoring platform for functional and signaling investigation of tumor-stromal interactions. The model was produced by injection of luciferase-labelled ESCC cells into the intraesophageal wall of nude mice. Histological examination indicates this orthotopic model is highly reproducible with 100% tumorigenesis among the four ESCC cell lines tested. This new model recapitulates many clinical and pathological properties of human ESCC, including esophageal luminal stricture by squamous cell carcinoma with nodular tumor growth, adventitia invasion, lymphovascular invasion, and perineural infiltration. It was tested using an AKT shRNA knockdown of ESCC cell lines and the in vivo tumor suppressive effects of AKT knockdown were observed. In conclusion, this ESCC orthotopic mouse model allows investigation of gene functions of cancer cells in a more natural tumor microenvironment and has advantages over previous established models. It provides a versatile platform with potential application for metastasis and therapeutic regimen testing.
Collapse
|
532
|
The cancer COMPASS: navigating the functions of MLL complexes in cancer. Cancer Genet 2015; 208:178-91. [PMID: 25794446 DOI: 10.1016/j.cancergen.2015.01.005] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 12/13/2022]
Abstract
The mixed-lineage leukemia family of histone methyltransferases (MLL1-4, or KMT2A-D) were previously linked to cancer through the founding member, MLL1/KMT2A, which is often involved in translocation-associated gene fusion events in childhood leukemias. However, in recent years, a multitude of tumor exome sequencing studies have revealed that orthologues MLL3/KMT2C and MLL2/KMT2D are mutated in a significant percentage of a large variety of malignancies, particularly solid tumors. These unexpected findings necessitate a deeper inspection into the activities and functional differences between the MLL/KMT2 family members. This review provides an overview of this protein family and its relation to cancers, focusing on the recent links between MLL3/KMT2C and MLL2/4/KMT2D and their potential roles as tumor suppressors in an assortment of cell types.
Collapse
|
533
|
Maraver A, Fernandez-Marcos PJ, Cash TP, Mendez-Pertuz M, Dueñas M, Maietta P, Martinelli P, Muñoz-Martin M, Martínez-Fernández M, Cañamero M, Roncador G, Martinez-Torrecuadrada JL, Grivas D, de la Pompa JL, Valencia A, Paramio JM, Real FX, Serrano M. NOTCH pathway inactivation promotes bladder cancer progression. J Clin Invest 2015; 125:824-30. [PMID: 25574842 DOI: 10.1172/jci78185] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/04/2014] [Indexed: 12/23/2022] Open
Abstract
NOTCH signaling suppresses tumor growth and proliferation in several types of stratified epithelia. Here, we show that missense mutations in NOTCH1 and NOTCH2 found in human bladder cancers result in loss of function. In murine models, genetic ablation of the NOTCH pathway accelerated bladder tumorigenesis and promoted the formation of squamous cell carcinomas, with areas of mesenchymal features. Using bladder cancer cells, we determined that the NOTCH pathway stabilizes the epithelial phenotype through its effector HES1 and, consequently, loss of NOTCH activity favors the process of epithelial-mesenchymal transition. Evaluation of human bladder cancer samples revealed that tumors with low levels of HES1 present mesenchymal features and are more aggressive. Together, our results indicate that NOTCH serves as a tumor suppressor in the bladder and that loss of this pathway promotes mesenchymal and invasive features.
Collapse
|
534
|
Abstract
Cell competition where 'loser' cells are eliminated by neighbors with higher fitness is a widespread phenomenon in development. However, a growing body of evidence argues cells with somatic mutations compete with their wild type counterparts in the earliest stages of cancer development. Recent studies have begun to shed light on the molecular and cellular mechanisms that alter the competitiveness of cells carrying somatic mutations in adult tissues. Cells with a 'winner' phenotype create clones which may expand into extensive fields of mutant cells within normal appearing epithelium, favoring the accumulation of further genetic alterations and the evolution of cancer. Here we focus on how mutations which disrupt the Notch signaling pathway confer a 'super competitor' status on cells in squamous epithelia and consider the broader implications for cancer evolution.
Collapse
Affiliation(s)
- Maria P Alcolea
- MRC Cancer Unit; University of Cambridge; Hutchison/MRC Research Center; Cambridge Biomedical Campus; Cambridge, UK
| | - Philip H Jones
- MRC Cancer Unit; University of Cambridge; Hutchison/MRC Research Center; Cambridge Biomedical Campus; Cambridge, UK
| |
Collapse
|
535
|
|
536
|
Zhu C, Li L, Zhao B. The regulation and function of YAP transcription co-activator. Acta Biochim Biophys Sin (Shanghai) 2015; 47:16-28. [PMID: 25487920 DOI: 10.1093/abbs/gmu110] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Hippo pathway was initially identified in Drosophila by genetic mosaic screens for tumor suppressor genes. Researches indicated that the Hippo pathway is a key regulator of organ size and is conserved during evolution. Furthermore, studies of mouse models and clinical samples demonstrated the importance of Hippo pathway dysregulation in human cancer development. In addition, the Hippo pathway contributes to progenitor cell and stem cell self-renewal and is thus involved in tissue regeneration. In the Hippo pathway, MST1/2 kinases together with the adaptor protein SAV phosphorylate LATS1/2 kinases. Interaction with an adaptor protein MOB is also important for LATS1/2 activation. Activated LATS1/2 in turn phosphorylate and inhibit Yes-associated protein (YAP). YAP is a key downstream effector of the Hippo pathway, and is a transcriptional co-activator that mainly interacts with TEAD family transcription factors to promote gene expression. Alteration of gene expression by YAP leads to cell proliferation, apoptosis evasion, and also stem cell amplification. In this review, we mainly focus on YAP, discussing its regulation and mechanisms of action in the context of organ size control, tissue regeneration and tumorigenesis.
Collapse
Affiliation(s)
- Chu Zhu
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou 310058, China
| | - Li Li
- Institute of Aging Research, Hangzhou Normal University, Hangzhou 311121, China
| | - Bin Zhao
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
537
|
Vyazunova I, Maklakova VI, Berman S, De I, Steffen MD, Hong W, Lincoln H, Morrissy AS, Taylor MD, Akagi K, Brennan CW, Rodriguez FJ, Collier LS. Sleeping Beauty mouse models identify candidate genes involved in gliomagenesis. PLoS One 2014; 9:e113489. [PMID: 25423036 PMCID: PMC4244117 DOI: 10.1371/journal.pone.0113489] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 10/27/2014] [Indexed: 01/01/2023] Open
Abstract
Genomic studies of human high-grade gliomas have discovered known and candidate tumor drivers. Studies in both cell culture and mouse models have complemented these approaches and have identified additional genes and processes important for gliomagenesis. Previously, we found that mobilization of Sleeping Beauty transposons in mice ubiquitously throughout the body from the Rosa26 locus led to gliomagenesis with low penetrance. Here we report the characterization of mice in which transposons are mobilized in the Glial Fibrillary Acidic Protein (GFAP) compartment. Glioma formation in these mice did not occur on an otherwise wild-type genetic background, but rare gliomas were observed when mobilization occurred in a p19Arf heterozygous background. Through cloning insertions from additional gliomas generated by transposon mobilization in the Rosa26 compartment, several candidate glioma genes were identified. Comparisons to genetic, epigenetic and mRNA expression data from human gliomas implicates several of these genes as tumor suppressor genes and oncogenes in human glioblastoma.
Collapse
Affiliation(s)
- Irina Vyazunova
- School of Pharmacy and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI, United States of America
| | - Vilena I. Maklakova
- School of Pharmacy and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI, United States of America
| | - Samuel Berman
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ishani De
- School of Pharmacy and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI, United States of America
| | - Megan D. Steffen
- School of Pharmacy and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI, United States of America
| | - Won Hong
- School of Pharmacy and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI, United States of America
| | - Hayley Lincoln
- School of Pharmacy and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI, United States of America
| | - A. Sorana Morrissy
- Division of Neurosurgery, Arthur & Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Michael D. Taylor
- Division of Neurosurgery, Arthur & Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Keiko Akagi
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Cameron W. Brennan
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Fausto J. Rodriguez
- Department of Pathology, Division of Neuropathology, Johns Hopkins University, Baltimore, MD, United States of America
| | - Lara S. Collier
- School of Pharmacy and University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, Madison, WI, United States of America
| |
Collapse
|