301
|
Hu Y, Teng X, Wu L, Liu W, An J. The Clinicopathological Correlations of hTERC Amplification with Esophageal Squamous Cell Precursor Lesions. Dig Dis Sci 2019; 64:68-75. [PMID: 30311151 PMCID: PMC6318245 DOI: 10.1007/s10620-018-5318-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/03/2018] [Indexed: 12/09/2022]
Abstract
BACKGROUND Esophageal squamous cell precursor lesions remain one of the most controversial topics in pathology and clinical management. AIMS To analyze the dysregulation of human telomerase RNA component (hTERC) in esophageal squamous cell precursor lesions and the clinicopathological correlations with the characteristics of esophageal squamous cell precursor lesions. METHODS Florescence in situ hybridization was performed to detect hTERC amplification in different gradings of esophageal squamous cell precursor lesions. With retrospective follow-up data, clinicopathological correlations between hTERC and esophageal squamous cell precursor lesions were subjected to logistic regression analysis. RESULTS hTERC amplification gradually increased with upgrading of dysplasia, reaching the highest level in high-grade intraepithelial neoplasia, and there was a significant difference between the low-grade intraepithelial neoplasia group and the high-grade intraepithelial neoplasia group (P = 0.00). Logistic regression analysis showed that hTERC amplification was correlated with both dysplasia grading and ulcer characteristics of esophageal squamous cell precursor lesions (P < 0.05). CONCLUSIONS hTERC amplification with increasing grading of esophageal squamous cell precursor lesions and the presence of ulcer characteristics might provide an important molecular and pathological marker for the diagnosis and clinical prognosis of esophageal squamous cell precursor lesions, especially for those ambiguous cases with more divergence in classification.
Collapse
Affiliation(s)
- Yanping Hu
- 0000 0004 0369 153Xgrid.24696.3fDepartment of Pathology, Beijing LuHe Hospital, Capital Medical University, Beijing, 101149 China
| | - Xiaojing Teng
- 0000 0004 0369 153Xgrid.24696.3fDepartment of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050 China
| | - Linlin Wu
- 0000 0004 0369 153Xgrid.24696.3fDepartment of Pathology, Beijing LuHe Hospital, Capital Medical University, Beijing, 101149 China
| | - Wei Liu
- 0000 0004 0369 153Xgrid.24696.3fDepartment of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050 China
| | - Jianduo An
- 0000 0004 0369 153Xgrid.24696.3fDepartment of Pathology, Beijing LuHe Hospital, Capital Medical University, Beijing, 101149 China
| |
Collapse
|
302
|
Wang Y, Wang G, Ma Y, Teng J, Wang Y, Cui Y, Dong Y, Shao S, Zhan Q, Liu X. FAT1, a direct transcriptional target of E2F1, suppresses cell proliferation, migration and invasion in esophageal squamous cell carcinoma. Chin J Cancer Res 2019; 31:609-619. [PMID: 31564804 PMCID: PMC6736659 DOI: 10.21147/j.issn.1000-9604.2019.04.05] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective Growing evidence indicates that FAT atypical cadherin 1 (FAT1) has aberrant genetic alterations and exhibits potential tumor suppressive function in esophageal squamous cell carcinoma (ESCC). However, the role of FAT1 in ESCC tumorigenesis remains not well elucidated. The aim of this study was to further investigate genetic alterations and biological functions of FAT1, as well as to explore its transcriptional regulation and downstream targets in ESCC. Methods The mutations of FAT1 in ESCC were achieved by analyzing a combined study from seven published genomic data, while the copy number variants of FAT1 were obtained from an analysis of our previous data as well as of The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) databases using the cBioPortal. The transcriptional regulation of FAT1 expression was investigated by chromatin immunoprecipitation (ChIP) and the luciferase reporter assays. In-cell western, Western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to assess the indicated gene expression. In addition, colony formation and Transwell migration/invasion assays were employed to test cell proliferation, migration and invasion. Finally, RNA sequencing was used to study the transcriptomes. Results FAT1 was frequently mutated in ESCC and was deleted in multiple cancers. Furthermore, the transcription factor E2F1 occupied the promoter region of FAT1, and depletion of E2F1 led to a decrease in transcription activity and mRNA levels of FAT1. Moreover, we found that knockdown of FAT1 promoted KYSE30 and KYSE150 cell proliferation, migration and invasion; while overexpression of FAT1 inhibited KYSE30 and KYSE410 cell proliferation, migration and invasion. In addition, knockdown of FAT1 led to enrichment of the mitogen-activated protein kinase (MAPK) signaling pathway and cell adhesion process. Conclusions Our data provided evidence for the tumor suppressive function of FAT1 in ESCC cells and elucidated the transcriptional regulation of FAT1 by E2F1, which may facilitate the understanding of molecular mechanisms of the progression of ESCC.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Guangchao Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yunping Ma
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Jinglei Teng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yongping Cui
- Shenzhen Peking University-The Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yan Dong
- College of Stomatology, Dalian Medical University, Dalian 116044, China
| | - Shujuan Shao
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Qimin Zhan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xuefeng Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
303
|
Shao Y, Hua Z, Zhao L, Shen Y, Guo X, Niu C, Wei W, Liu F. Time Trends of Gastrointestinal Cancers Incidence and Mortality in Yangzhong From 1991 to 2015: An Updated Age-Period-Cohort Analysis. Front Oncol 2018; 8:638. [PMID: 30619771 PMCID: PMC6306425 DOI: 10.3389/fonc.2018.00638] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/06/2018] [Indexed: 12/29/2022] Open
Abstract
Background: Gastrointestinal (GI) cancers are the common cause of morbidity and mortality in China which seriously threaten people's health and lives. The aim of this study was to describe the temporal trend in the epidemiology of GI cancers from 1991 to 2015, with an emphasis on the effects of age, period and cohort in Yangzhong City, Jiangsu province, a high-risk area of GI cancers in China. Methods: Our study extracted cases of gastric cancer, esophageal cancer and colorectal cancer diagnosed from 1991 to 2015 from Yangzhong Cancer Registry. Age-standardized rates (ASRs) were calculated and joinpoint regression was used to compute the estimated annual percent changes. Age-period-cohort (APC) model was performed to investigate the independent effects of age, calendar period, and birth cohort. Results: Between 1991 and 2015, 18,006 new cases and 10,262 deaths were registered with GI cancers in Yangzhong. The age-standardized incidence rates (ASIRs) of gastric cancer decreased in both sexes during the study period. And the incidence rates of esophageal cancer stabilized at first then continued to decline, the turning point was in 2005 for men and 2001 for women. Changes in the mortality rates of gastric cancer and esophageal cancer showed significant declined trends around 2000-2010 in both genders. The incidence rates of colorectal cancer increased steadily during the entire study period, and the increase was more pronounced in the mortality rates of men. The results of APC analysis suggest that general decreases in incidence and mortality of esophageal cancer and gastric cancer might be caused by the downward trend of the period and cohort effects, while the increases in colorectal cancer might be caused by the uptrend of the period effects. Conclusions: The incidence and mortality rates of esophageal and gastric cancers showed a downward trend and colorectal cancer was on the rise as a whole in Yangzhong City. The different burden of gastrointestinal cancer indicating heterogeneous risk factors exist and may have contributed to these temporal variations.
Collapse
Affiliation(s)
- Yi Shao
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Zhaolai Hua
- Department of Epidemiology, Yangzhong Cancer Research Institute, Yangzhong, China
| | - Lei Zhao
- Department of Molecular Physiology and Biophysics, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Yi Shen
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Xudong Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Chen Niu
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Wenqiang Wei
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fen Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| |
Collapse
|
304
|
Liu T, Wang Z, Zhou R, Liang W. Focally amplified lncRNA on chromosome 1 regulates apoptosis of esophageal cancer cells via DRP1 and mitochondrial dynamics. IUBMB Life 2018; 71:254-260. [PMID: 30501006 DOI: 10.1002/iub.1971] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/24/2018] [Accepted: 10/10/2018] [Indexed: 01/05/2023]
Abstract
Long noncoding RNAs (lncRNAs), a family of noncoding RNA transcripts with a length of <200 nucleotides (nts), have been associated with the pathological development of various types of carcinogenesis. Focally amplified lncRNA on chromosome 1 (FAL1) is a recently identified lncRNA. In the current study, we aimed to investigate the physiological function of FAL1 in esophageal squamous cell carcinoma (ESCC). Our findings demonstrate that FAL1 was associated with esophageal cancer cell survival by regulating mitochondrial fission. First, we found that the expression of the mitochondrial fission protein dynamin-related protein 1 (DRP1) was significantly reduced, but the expression of the mitochondrial fusion protein mitofusin 1 (Mfn1) was increased in ESCC tissues and esophageal cancer cell lines as compared with adjacent normal tissues and a normal esophagus epithelial cell line. In addition, we found that reduced expression of DRP1 in the esophageal cancer cell lines KYSE450 and EC9706 cells was associated with increased expression of FAL1. Inhibition of FAL1 promoted mitochondrial fission and mitochondrial dysfunction in KYSE450 and EC9706 cells mediated by DRP1. Silencing of DRP1 abolished FAL1-induced apoptosis through a mitochondrial-dependent pathway. Our findings suggest that FAL1/DRP1 could be a therapeutic target for the treatment of ESCC. © 2018 IUBMB Life, 71(1):254-260, 2019.
Collapse
Affiliation(s)
- Tao Liu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Zhi Wang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Riqiang Zhou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Wulin Liang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| |
Collapse
|
305
|
Sugimoto K, Ito T, Woo J, Tully E, Sato K, Orita H, Brock MV, Gabrielson E. Prognostic Impact of Phosphorylated Discoidin Domain Receptor-1 in Esophageal Cancer. J Surg Res 2018; 235:479-486. [PMID: 30691832 DOI: 10.1016/j.jss.2018.10.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/17/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is common in East Asia and also is often deadly. We sought to determine whether measuring the discoidin domain receptor-1 (DDR1)-both total and phosphorylated proteins-could improve our ability to predict recurrence in ESCC. MATERIALS AND METHODS Total DDR1 and phosphorylated DDR1 (pDDR1) were measured using semiquantitative immunohistochemistry in a cohort of 60 patients with ESCC. Association between these immunohistochemical measurements and standard clinical-pathological variables such as patient recurrence-free survival was examined using univariate and multivariate analyses. RESULTS Six patients (10.0%) had regional recurrence and eight patients (13.3%) had distant recurrence. In univariate analysis, early disease recurrence correlated with intense staining of total DDR1 (P = 0.03) as well as intense staining of pDDR1 (P < 0.001). On multivariate analysis, only regional lymph node metastasis (P = 0.04, HR = 4.20) and intensity of pDDR1 immunohistochemistry (P = 0.03, HR = 4.27) emerged as significant independent prognostic factors for recurrence. CONCLUSIONS This study suggests that immunohistochemical measurements of both the DDR1 protein and pDDR1 can provide prognostic value in ESCC, even when other clinical and pathological factors are also being considered.
Collapse
Affiliation(s)
- Kiichi Sugimoto
- Department of Surgery, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Coloproctological Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Tomoaki Ito
- Department of Surgery, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Surgery, Juntendo University Shizuoka Hospital, Izunokuni-shi, Shizuoka, Japan
| | - Juhyung Woo
- Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ellen Tully
- Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Koichi Sato
- Department of Surgery, Juntendo University Shizuoka Hospital, Izunokuni-shi, Shizuoka, Japan
| | - Hajime Orita
- Department of Surgery, Juntendo University Shizuoka Hospital, Izunokuni-shi, Shizuoka, Japan
| | - Malcolm V Brock
- Department of Surgery, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward Gabrielson
- Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
306
|
Chen L, You C, Jin X, Zhou L, Huang L, Wang Y. Cytoskeleton-associated protein 4 is a novel serodiagnostic marker for esophageal squamous-cell carcinoma. Onco Targets Ther 2018; 11:8221-8226. [PMID: 30538491 PMCID: PMC6251352 DOI: 10.2147/ott.s183790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Recent years have witnessed significant progress in the treatment of esophageal squamous-cell carcinoma (ESCC); however, the prognosis of ESCC is still unsatisfactory. Bio-markers are required to improve identification of high-risk populations and help management of ESCC. This study was to evaluate the role of serum CKAP4 in ESCC. Methods This longitudinal study recruited 207 ESCC patients and age-/sex-matched healthy controls. Circulating levels of CKAP4 were measured using ELISA kits, while the expression of CKAP4 in esophageal tissue was evaluated using Western blotting. Results Serum CKAP4 levels were higher in ESCC patients (380.2±171.3 pg/mL) than healthy controls (271.8±97.4 pg/mL; P<0.001). The area under the receiver-operating characteristic curve of serum CKAP4 levels to identify the presence of ESCC was 0.675 (95% CI 0.622–0.728; P<0.001). According to Youden’s index, the best cutoff value was 429.1 pg/mL (sensitivity 0.415 and specificity 0.995). Furthermore, after follow-up, multivariate analyses identified that pathological lymph node metastases were the poorest prognostic factor (HR 1.862, 95% CI 1.093–3.173; P=0.022), followed by serum CKAP4 (HR 1.437, 95% CI 1.025–2.014; P=0.035). When stratified by tertiles of serum CKAP4, subjects in the first tertile presented a mean survival time of 75.4 months (95% CI 68.0–81.9), which decreased significantly in the second tertile (73.8 months, 95% CI 61.4–86.3) and the third tertile (59.9 months, 95% CI 49.8–70.0, log-rank χ2=8.235; P=0.016). Conclusion These results suggested that serum CKAP4 could be a potential biomarker for clinical management of ESCC.
Collapse
Affiliation(s)
- Lei Chen
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Chuanwen You
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Xiaowei Jin
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Lei Zhou
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Liyou Huang
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Yanhua Wang
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| |
Collapse
|
307
|
Wen J, Hu Y, Liu Q, Ling Y, Zhang S, Luo K, Xie X, Fu J, Yang H. miR-424 coordinates multilayered regulation of cell cycle progression to promote esophageal squamous cell carcinoma cell proliferation. EBioMedicine 2018; 37:110-124. [PMID: 30361064 PMCID: PMC6284509 DOI: 10.1016/j.ebiom.2018.10.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Dysregulation of the cell cycle has been implicated in esophageal squamous cell carcinoma (ESCC) progression. This study aimed to evaluate the role of miR-424 in cell cycle regulation and ESCC proliferation. METHODS The role of miR-424 in cell proliferation was evaluated in vitro and in vivo. Transcriptional activation of miR-424 was determined using chromatin immunoprecipitation, and binding of miR-424 to targets was verified using miRNA ribonucleoprotein complex immunoprecipitation. FINDINGS miR-424 was upregulated and correlated with poor survival in ESCC patients. Repression or overexpression of miR-424 respectively decreased or increased ESCC cell proliferation in vitro and in vivo. miR-424 expression is transcriptionally regulated by E2F1 and increased during G1/S transition. Knockdown or overexpression of miR-424 respectively inhibited or promoted both G1/S and G2/M cell cycle transitions in ESCC cells, and these effects were mediated by two newly identified miR-424 targets, PRKCD and WEE1, respectively. Consequently, elevation of PRKCD by miR-424 knockdown led to enhanced stability of the p21Cip1 protein via increased activation of PRKCD and downstream p38 MAPK and JNK signaling to block CDK2 activation and G1/S transition, while elevated WEE1 maintained CDC2 in an inactive state to block G2/M transition. However, circLARP4 could sponge the binding of miR-424 to PRKCD, thus compromising the regulation of G1/S progression by miR-424. INTERPRETATION miR-424 coordinates a previously unknown, multilayered regulation of ESCC cell cycle progression to promote ESCC proliferation, and may be used as a novel prognostic marker and an effective therapeutic target for ESCCs. FUND: National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yi Hu
- Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Qianwen Liu
- Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yihong Ling
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Shuishen Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou 510080, China
| | - Kongjia Luo
- Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Xiuying Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Jianhua Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Hong Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Esophageal Cancer Institute, 651 Dongfeng East Road, Guangzhou 510060, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China.
| |
Collapse
|
308
|
RIT1 suppresses esophageal squamous cell carcinoma growth and metastasis and predicts good prognosis. Cell Death Dis 2018; 9:1085. [PMID: 30348939 PMCID: PMC6197279 DOI: 10.1038/s41419-018-0979-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/23/2018] [Accepted: 06/26/2018] [Indexed: 01/05/2023]
Abstract
Ras-like without CAAX1 (RIT1) protein is a member of Ras family, which plays critical roles in signaling pathways and cellular process regulation. However, the role of RIT1 in esophageal squamous cell carcinoma (ESCC) is unclear. In this study, we found that the expression of RIT1 is downregulated in ESCC compared to corresponding non-tumor tissues. The low-level expression of RIT1 was correlated with poorer prognosis. Then we showed that RIT1 inhibited proliferation, invasion, and migration of ESCC cells, and silencing RIT1 by shRNA promoted tumorigenicity and metastasis in nude mice. We further demonstrated that RIT1 inhibited the malignant behaviors of ESCC through inhibiting the PI3K/AKT and MAPK pathway and epithelial–mesenchymal transition in ESCC cells. Our study also revealed that RIT1 increased drug sensitivity to cisplatin (CDDP), and this function could be carried out through downregulating stemness of ESCC. In conclusion, our study indicates for the first time that RIT1 displays tumor-suppressing functions in ESCC, and these functions were carried out by inhibiting MAPK and PI3K/AKT signaling pathway, inhibiting EMT, and downregulating cancer stemness of ESCC cells.
Collapse
|
309
|
Roy R, Kandimalla R, Sonohara F, Koike M, Kodera Y, Takahashi N, Yamada Y, Goel A. A comprehensive methylation signature identifies lymph node metastasis in esophageal squamous cell carcinoma. Int J Cancer 2018; 144:1160-1169. [PMID: 30006931 DOI: 10.1002/ijc.31755] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/25/2018] [Indexed: 01/01/2023]
Abstract
Treatment modalities in esophageal squamous cell carcinoma (ESCC) depend largely on lymph node metastasis (LNM) status. With suboptimal detection sensitivity of existing imaging techniques, we propose a methylation signature which identifies patients with LNM with greater accuracy. This would allow precise stratification of high-risk patients requiring more aggressive treatment from low-risk ESCC patients who can forego radical surgery. An unbiased genome-wide methylation signature for LNM detection was established from an initial in silico discovery phase. The signature was tested in independent clinical cohorts comprising of 249 ESCC patients. The prognostic potential of the methylation signature was compared to clinical variables including LNM status. A 10-probe LNM associated signature (LNAS) was developed using stringent bioinformatics analyses. The area under the curve values for LNAS risk scores were 0.81 and 0.88 in the training and validation cohorts respectively, in association with lymphatic vessel invasion and tumor stage. High LNAS risk-score was also associated with worse overall survival [HR (95% CI) 3 (1.8-4.8), p < 0.0001 training and 3.9 (1.5-10.2), p = 0.001 validation cohort]. In conclusion, our novel methylation signature is a powerful biomarker that identifies LNM status robustly and is also associated with worse prognosis in ESCC patients.
Collapse
Affiliation(s)
- Roshni Roy
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | - Raju Kandimalla
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | - Fuminori Sonohara
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiko Koike
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoki Takahashi
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuhide Yamada
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ajay Goel
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| |
Collapse
|
310
|
Involving the microRNA Targetome in Esophageal-Cancer Development and Behavior. Cancers (Basel) 2018; 10:cancers10100381. [PMID: 30322005 PMCID: PMC6210990 DOI: 10.3390/cancers10100381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Esophageal cancer (EC) is the eighth most common and sixth leading cause of cancer-related mortality in the world. Despite breakthroughs in EC diagnosis and treatment, patients with complete pathologic response after being submitted to chemoradiotherapy are still submitted to surgery, despite its high morbidity. Single-nucleotide polymorphisms (SNPs) in miRNA, miRNA-binding sites, and in its biogenesis pathway genes can alter miRNA expression patterns, thereby influencing cancer risk and prognosis. In this review, we systematized the information available regarding the impact of these miR-SNPs in EC development and prognosis. We found 34 miR-SNPs that were associated with EC risk. Despite the promising applicability of these miR-SNPs as disease biomarkers, they still lack validation in non-Asian populations. Moreover, there should be more pathway-based approaches to evaluate the cumulative effect of multiple unfavorable genotypes and, consequently, identify miR-SNPs signatures capable of predicting EC therapy response and prognosis.
Collapse
|
311
|
Xin Z, Song X, Jiang B, Gongsun X, Song L, Qin Q, Wang Q, Shi M, Liu X. Blocking FGFR4 exerts distinct anti-tumorigenic effects in esophageal squamous cell carcinoma. Thorac Cancer 2018; 9:1687-1698. [PMID: 30267473 PMCID: PMC6275831 DOI: 10.1111/1759-7714.12883] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 08/26/2018] [Accepted: 08/29/2018] [Indexed: 02/06/2023] Open
Abstract
Background The FGFR family can be activated by FGFs and plays important roles in regulating cell growth, differentiation, migration, and angiogenesis. Recent studies have suggested that FGFR4 could regulate several processes, including tumor progression. Esophageal squamous cell carcinoma (ESCC) is a malignancy with high global occurrence. However, the molecule mechanism and the potential roles of FGFR4 in ESCC remain unknown. Methods Immunohistochemistry and Western blotting were used to detect FGFR4 expression in ESCC samples and cell lines. Cell counting kit‐8, and clonogenic, transwell, flow cytometric, and tumor xenograft in nude mice assays were utilized to determine the effect of blocking FGFR4 in proliferation, invasion, migration, and apoptosis of ESCC cells. Results FGFR4 is frequently overexpressed in ESCC tissue and cell lines. in vitro assays have shown that blocking FGFR4 by a specific blocker, H3B‐6527, significantly decreases proliferation, invasion, and migration, and alters epithelial‐mesenchymal transition markers in ESCC cells. In addition, FGFR4 blockade is associated with the induction of apoptosis and affects PI3K/Akt and MAPK/ERK pathways. Moreover, FGFR4 blockade could significantly inhibit the growth of xenograft tumors in vivo. Conclusion Our findings suggest that blocking FGFR4 significantly suppresses the malignant behaviors of ESCC and indicate that FGFR4 is a potential target for the treatment of ESCC.
Collapse
Affiliation(s)
- Zhongwei Xin
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Xuemin Song
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Bin Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Xin Gongsun
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Liang Song
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Qiming Qin
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Qiang Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Mo Shi
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Xiangyan Liu
- Department of Thoracic Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| |
Collapse
|
312
|
Senthebane DA, Jonker T, Rowe A, Thomford NE, Munro D, Dandara C, Wonkam A, Govender D, Calder B, Soares NC, Blackburn JM, Parker MI, Dzobo K. The Role of Tumor Microenvironment in Chemoresistance: 3D Extracellular Matrices as Accomplices. Int J Mol Sci 2018; 19:E2861. [PMID: 30241395 PMCID: PMC6213202 DOI: 10.3390/ijms19102861] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The functional interplay between tumor cells and their adjacent stroma has been suggested to play crucial roles in the initiation and progression of tumors and the effectiveness of chemotherapy. The extracellular matrix (ECM), a complex network of extracellular proteins, provides both physical and chemicals cues necessary for cell proliferation, survival, and migration. Understanding how ECM composition and biomechanical properties affect cancer progression and response to chemotherapeutic drugs is vital to the development of targeted treatments. METHODS 3D cell-derived-ECMs and esophageal cancer cell lines were used as a model to investigate the effect of ECM proteins on esophageal cancer cell lines response to chemotherapeutics. Immunohistochemical and qRT-PCR evaluation of ECM proteins and integrin gene expression was done on clinical esophageal squamous cell carcinoma biopsies. Esophageal cancer cell lines (WHCO1, WHCO5, WHCO6, KYSE180, KYSE 450 and KYSE 520) were cultured on decellularised ECMs (fibroblasts-derived ECM; cancer cell-derived ECM; combinatorial-ECM) and treated with 0.1% Dimethyl sulfoxide (DMSO), 4.2 µM cisplatin, 3.5 µM 5-fluorouracil and 2.5 µM epirubicin for 24 h. Cell proliferation, cell cycle progression, colony formation, apoptosis, migration and activation of signaling pathways were used as our study endpoints. RESULTS The expression of collagens, fibronectin and laminins was significantly increased in esophageal squamous cell carcinomas (ESCC) tumor samples compared to the corresponding normal tissue. Decellularised ECMs abrogated the effect of drugs on cancer cell cycling, proliferation and reduced drug induced apoptosis by 20⁻60% that of those plated on plastic. The mitogen-activated protein kinase-extracellular signal-regulated kinase (MEK-ERK) and phosphoinositide 3-kinase-protein kinase B (PI3K/Akt) signaling pathways were upregulated in the presence of the ECMs. Furthermore, our data show that concomitant addition of chemotherapeutic drugs and the use of collagen- and fibronectin-deficient ECMs through siRNA inhibition synergistically increased cancer cell sensitivity to drugs by 30⁻50%, and reduced colony formation and cancer cell migration. CONCLUSION Our study shows that ECM proteins play a key role in the response of cancer cells to chemotherapy and suggest that targeting ECM proteins can be an effective therapeutic strategy against chemoresistant tumors.
Collapse
Affiliation(s)
- Dimakatso Alice Senthebane
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Tina Jonker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Arielle Rowe
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Daniella Munro
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Collet Dandara
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Ambroise Wonkam
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Dhirendra Govender
- Division of Anatomical Pathology, Faculty of Health Sciences, University of Cape Town, NHLS-Groote Schuur Hospital, Cape Town 7925, South Africa.
| | - Bridget Calder
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - Nelson C Soares
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - Jonathan M Blackburn
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - M Iqbal Parker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Kevin Dzobo
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| |
Collapse
|
313
|
Su M, Xiao Y, Ma J, Cao D, Zhou Y, Wang H, Liao Q, Wang W. Long non-coding RNAs in esophageal cancer: molecular mechanisms, functions, and potential applications. J Hematol Oncol 2018; 11:118. [PMID: 30223861 PMCID: PMC6142629 DOI: 10.1186/s13045-018-0663-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 09/06/2018] [Indexed: 12/20/2022] Open
Abstract
Esophageal cancer (EC) is the sixth leading cause of cancer-related death worldwide. The lack of early diagnostic biomarkers and effective prognostic indicators for metastasis and recurrence has resulted in the poor prognosis of EC. In addition, the underlying molecular mechanisms of EC development have yet to be elucidated. Accumulating evidence has demonstrated that lncRNAs play a vital role in the pathological progression of EC. LncRNAs may regulate gene expression through the recruitment of histone-modifying complexes to the chromatin and through interactions with RNAs or proteins. Recent evidence has demonstrated that the dysregulation of lncRNAs plays important roles in the proliferation, metastasis, invasion, angiogenesis, apoptosis, chemoradiotherapy resistance, and stemness of EC, which suggests potential clinical implications. In this review, we highlight the emerging roles and regulatory mechanisms of lncRNAs in the context of EC and discuss their potential clinical applications as diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Min Su
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China. .,Department of the Central Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China.
| | - Yuhang Xiao
- Department of Pharmacy, Xiangya Hospital of Xiangya School of Medicine, Central South University, Changsha, 410001, Hunan, People's Republic of China
| | - Junliang Ma
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Deliang Cao
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Yong Zhou
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Hui Wang
- Department of Thoracic Radiotherapy, Key laboratory of Translational Radiation Oncology, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Qianjin Liao
- Department of the Central Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China.
| | - Wenxiang Wang
- Department of the 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
314
|
Kijima T, Nakagawa H, Shimonosono M, Chandramouleeswaran PM, Hara T, Sahu V, Kasagi Y, Kikuchi O, Tanaka K, Giroux V, Muir AB, Whelan KA, Ohashi S, Naganuma S, Klein-Szanto AJ, Shinden Y, Sasaki K, Omoto I, Kita Y, Muto M, Bass AJ, Diehl JA, Ginsberg GG, Doki Y, Mori M, Uchikado Y, Arigami T, Avadhani NG, Basu D, Rustgi AK, Natsugoe S. Three-Dimensional Organoids Reveal Therapy Resistance of Esophageal and Oropharyngeal Squamous Cell Carcinoma Cells. Cell Mol Gastroenterol Hepatol 2018; 7:73-91. [PMID: 30510992 PMCID: PMC6260338 DOI: 10.1016/j.jcmgh.2018.09.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/06/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Oropharyngeal and esophageal squamous cell carcinomas, especially the latter, are a lethal disease, featuring intratumoral cancer cell heterogeneity and therapy resistance. To facilitate cancer therapy in personalized medicine, three-dimensional (3D) organoids may be useful for functional characterization of cancer cells ex vivo. We investigated the feasibility and the utility of patient-derived 3D organoids of esophageal and oropharyngeal squamous cell carcinomas. METHODS We generated 3D organoids from paired biopsies representing tumors and adjacent normal mucosa from therapy-naïve patients and cell lines. We evaluated growth and structures of 3D organoids treated with 5-fluorouracil ex vivo. RESULTS Tumor-derived 3D organoids were grown successfully from 15 out of 21 patients (71.4%) and passaged with recapitulation of the histopathology of the original tumors. Successful formation of tumor-derived 3D organoids was associated significantly with poor response to presurgical neoadjuvant chemotherapy or chemoradiation therapy in informative patients (P = 0.0357, progressive and stable diseases, n = 10 vs. partial response, n = 6). The 3D organoid formation capability and 5-fluorouracil resistance were accounted for by cancer cells with high CD44 expression and autophagy, respectively. Such cancer cells were found to be enriched in patient-derived 3D organoids surviving 5-fluorouracil treatment. CONCLUSIONS The single cell-based 3D organoid system may serve as a highly efficient platform to explore cancer therapeutics and therapy resistance mechanisms in conjunction with morphological and functional assays with implications for translation in personalized medicine.
Collapse
Key Words
- 3D Organoids
- 3D, 3-dimensional
- 5-Fluorouracil
- 5FU, 5-fluorouracil
- AV, autophagy vesicle
- Autophagy
- CD44
- CD44H, high expression of CD44
- CQ, chloroquine
- DMEM, Dulbecco’s modified Eagle medium
- EMT, epithelial-mesenchymal transition
- ESCC, esophageal squamous cell carcinoma
- FBS, fetal bovine serum
- H&E, hematoxylin and eosin
- IC50, half maximal inhibitory concentration
- IHC, immunohistochemistry
- LC3, light chain 3
- OPSCC, oropharyngeal squamous cell carcinoma
- PI, propidium iodide
- SCCs, squamous cell carcinomas
- TE11R, 5-fluorouracil–resistant derivative of TE11
Collapse
Affiliation(s)
- Takashi Kijima
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan; Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hiroshi Nakagawa
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania.
| | - Masataka Shimonosono
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan; Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Prasanna M Chandramouleeswaran
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Takeo Hara
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Varun Sahu
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Yuta Kasagi
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Osamu Kikuchi
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Koji Tanaka
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania; Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Veronique Giroux
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Amanda B Muir
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kelly A Whelan
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania; Fels Institute for Cancer Research & Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Shinya Ohashi
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seiji Naganuma
- Department of Pathology, Kochi University School of Medicine, Nankoku, Japan
| | - Andres J Klein-Szanto
- Histopathology Facility and Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yoshiaki Shinden
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ken Sasaki
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Itaru Omoto
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoshiaki Kita
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Adam J Bass
- Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - J Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Gregory G Ginsberg
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasuto Uchikado
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takaaki Arigami
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Narayan G Avadhani
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Devraj Basu
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Anil K Rustgi
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania.
| | - Shoji Natsugoe
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.
| |
Collapse
|
315
|
Wu IC, Syu HY, Jen CP, Lu MY, Chen YT, Wu MT, Kuo CT, Tsai YY, Wang HC. Early identification of esophageal squamous neoplasm by hyperspectral endoscopic imaging. Sci Rep 2018; 8:13797. [PMID: 30218087 PMCID: PMC6138669 DOI: 10.1038/s41598-018-32139-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/03/2018] [Indexed: 12/13/2022] Open
Abstract
Esophageal squamous neoplasm presents a spectrum of different diatheses. A precise assessment for individualized treatment depends on the accuracy of the initial diagnosis. Detection relies on comprehensive and accurate white-light, iodine staining, and narrow-band imaging endoscopy. These methods have limitations in addition to its invasive nature and the potential risks related to the method. These limitations include difficulties in precise tumor delineation to enable complete resection, inflammation and malignancy differentiation, and stage determination. The resolution of these problems depends on the surgeon's ability and experience with available technology for visualization and resection. We proposed a method for identifying early esophageal cancerous lesion by endoscopy and hyperspectral endoscopic imaging. Experimental result shows the characteristic spectrum of a normal esophagus, precancerous lesion, canceration, and intraepithelial papillary capillary loop can be identified through principal component score chart. The narrow-band imaging (NBI) image shows remarkable spectral characteristic distribution, and the sensitivity and specificity of the proposed method are higher than those of other methods by ~0.8 and ~0.88, respectively. The proposed method enables the accurate visualization of target organs, it may be useful to capsule endoscope and telemedicine, which requires highly precise images for diagnosis.
Collapse
Affiliation(s)
- I-Chen Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Hao-Yi Syu
- Graduate Institute of Opto-Mechatronics, National Chung Cheng University, 168 University Rd., Min-Hsiung, Chia-Yi, 62102, Taiwan
| | - Chun-Ping Jen
- Department of Mechanical Engineering, National Chung Cheng University, 168 University Rd., Min-Hsiung, Chia-Yi, 62102, Taiwan
| | - Ming-Yen Lu
- Department of Materials Science and Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Road, Hsinchu, 30013, Taiwan
| | - Yi-Ting Chen
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Ming-Tsang Wu
- Department of Public Health, Graduate Institute of Clinical Medicine, Research Center for Environmental Medicine, Kaohsiung Medical University, No. 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Chie-Tong Kuo
- Department of Physics, National Sun Yat-sen University, 70 Lienhai Rd., Kaohsiung, 80424, Taiwan
| | - Yu-Yuan Tsai
- Department of Gastroenterology, Kaohsiung Armed Forces General Hospital, 2, Zhongzheng 1st.Rd., Lingya District, Kaohsiung City, 80284, Taiwan.
| | - Hsiang-Chen Wang
- Graduate Institute of Opto-Mechatronics, National Chung Cheng University, 168 University Rd., Min-Hsiung, Chia-Yi, 62102, Taiwan.
| |
Collapse
|
316
|
Abstract
Esophageal cancer affects more than 4,50,000 persons worldwide, and its incidence has increased in recent years. It is the eighth most common cancer across the globe. The main histologic types are esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EA), and their associated risk factors are well known. Achalasia, an idiopathic esophageal disorder that conditions aperistalsis and the absence of lower esophageal sphincter relaxation, stands out among them. The prevalence of ESCC in subjects with esophageal achalasia is 26 in every 1,000 cases, whereas the prevalence of EA is 4 in every 1,000. Patients with achalasia have a 50 times higher risk of presenting with ESCC than the general population, and the disease manifests 20-25 years after achalasia symptom onset. Multiple mechanisms are related to the development of ESCC in achalasia, and they include bacterial overgrowth, food stasis, genetic alterations, and chronic inflammation. Regarding the risk of EA in achalasia patients, most cases are associated with Barrett's esophagus, due to uncontrolled chronic acid reflux. Given that achalasia is a well-established factor for ESCC/EA, clinicians must be aware of said associations to enable the development of programs for the prevention and opportune detection of these cancers in patients with achalasia.
Collapse
Affiliation(s)
- Maura Torres-Aguilera
- Department of Pediatric Gastroenteritis, Hospital CMN "La Raza", Mexico City, Mexico
| | - José María Remes Troche
- Digestive Physiology and Motility Laboratory, Medical Biological Research Institute, Universidad Veracruzana, Veracruz, Mexico,
| |
Collapse
|
317
|
Hamai Y, Hihara J, Emi M, Furukawa T, Murakami Y, Nishibuchi I, Ibuki Y, Yamakita I, Kurokawa T, Nagata Y, Okada M. Evaluation of Prognostic Factors for Esophageal Squamous Cell Carcinoma Treated with Neoadjuvant Chemoradiotherapy Followed by Surgery. World J Surg 2018; 42:1496-1505. [PMID: 29030675 DOI: 10.1007/s00268-017-4283-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Intensive trimodality therapy is needed for locally advanced esophageal squamous cell carcinoma (ESCC). However, some patients develop recurrence and die of cancer even after trimodality therapy. METHODS We evaluated prognostic factors based on data from 125 patients with ESCC who underwent neoadjuvant chemoradiotherapy (NCRT) comprising concurrent chemotherapy and 40 Gy of radiation, followed by curative-intent esophagectomy. RESULTS Thirty-four (27.2%) patients achieved a pathological complete response (pCR) after NCRT. The 5-year recurrence-free (RFS) and overall survival (OS) rates of all patients were 49.2 and 52.9%, respectively, and were significantly better for patients with pCR than without pCR (p = 0.01 and 0.02, respectively). Univariate and multivariate analyses selected performance status [PS 0 vs. 1: hazard ratio (HR) 2.05; 95% confidence interval (CI) 1.30-4.84; p = 0.01] and ypN (0 vs. 1: HR 2.33; 95% CI 1.12-4.84; p = 0.02; 0 vs. 2/3: HR 3.73; 95% CI 1.68-8.28; p = 0.001) as independent covariates for RFS. Furthermore, PS (0 vs. 1; HR 2.94; 95% CI 1.51-5.72; p = 0.002) and ypN (0 vs. 1; HR 2.26; 95% CI 1.09-4.69; p = 0.03; 0 vs. 2/3: HR 3.90; 95% CI 1.79-8.48; p = 0.001) were also independent covariates for OS. CONCLUSIONS Performance status 1 and ypN+ were significantly associated with a poor prognosis after trimodality therapy for ESCC.
Collapse
Affiliation(s)
- Yoichi Hamai
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan.
| | - Jun Hihara
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Manabu Emi
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Takaoki Furukawa
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Yuji Murakami
- Department of Radiation Oncology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Ikuno Nishibuchi
- Department of Radiation Oncology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuta Ibuki
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Ichiko Yamakita
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Tomoaki Kurokawa
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
318
|
Kajiwara C, Fumoto K, Kimura H, Nojima S, Asano K, Odagiri K, Yamasaki M, Hikita H, Takehara T, Doki Y, Morii E, Kikuchi A. p63-Dependent Dickkopf3 Expression Promotes Esophageal Cancer Cell Proliferation via CKAP4. Cancer Res 2018; 78:6107-6120. [PMID: 30181180 DOI: 10.1158/0008-5472.can-18-1749] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/20/2018] [Accepted: 08/28/2018] [Indexed: 11/16/2022]
Abstract
Dickkopf3 (DKK3) is a secretory protein that belongs to the DKK family, but exhibits structural divergence from other family members, and its corresponding receptors remain to be identified. Although DKK3 has been shown to have oncogenic functions in certain cancer types, the underlying mechanism by which DKK3 promotes tumorigenesis remains to be clarified. We show here that DKK3 stimulates esophageal cancer cell proliferation via cytoskeleton-associated protein 4 (CKAP4), which acts as a receptor for DKK3. DKK3 was expressed in approximately 50% of tumor lesions of esophageal squamous cell carcinoma (ESCC) cases; simultaneous expression of DKK3 and CKAP4 was associated with poor prognosis. Anti-CKAP4 antibody inhibited both binding of DKK3 to CKAP4 and xenograft tumor formation induced by ESCC cells. p63, a p53-related transcriptional factor frequently amplified in ESCC, bound to the upstream region of the DKK3 gene. Knockdown of p63 decreased DKK3 expression in ESCC cells, and reexpression of DKK3 partially rescued cell proliferation in p63-depleted ESCC cells. Expression of ΔNp63α and DKK3 increased the size of tumor-like esophageal organoids, and anti-CKAP4 antibody inhibited growth of esophageal organoids. Taken together, these results suggest that the DKK3-CKAP4 axis might serve as a novel molecular target for ESCC.Significance: In esophageal cancer, findings identify DKK3 as a poor prognostic indicator and demonstrate CKAP4 inhibition as an effective therapeutic strategy. Cancer Res; 78(21); 6107-20. ©2018 AACR.
Collapse
Affiliation(s)
- Chihiro Kajiwara
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Katsumi Fumoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hirokazu Kimura
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Keita Asano
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kazuki Odagiri
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan.
| |
Collapse
|
319
|
Zhao X, Ren Y, Hu Y, Cui N, Wang X, Cui Y. Neoadjuvant chemotherapy versus neoadjuvant chemoradiotherapy for cancer of the esophagus or the gastroesophageal junction: A meta-analysis based on clinical trials. PLoS One 2018; 13:e0202185. [PMID: 30138325 PMCID: PMC6107145 DOI: 10.1371/journal.pone.0202185] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 07/30/2018] [Indexed: 11/25/2022] Open
Abstract
Background The benefit of neoadjuvant chemotherapy and neoadjuvant chemoradiotherapy for treating cancer of the esophagus or the gastroesophageal junction remains controversial. In the present study, we conducted a comprehensive meta-analysis to examine the efficacy of these two management strategies. Methods The MEDLINE (PubMed), SinoMed, Embase, and Cochrane Library databases were searched for eligible studies. We searched for the most relevant studies published until the end of September 2017. Data were extracted independently and were analyzed using RevMan statistical software version 5.3 (Cochrane Collaboration, http://tech.cochrane.org/revman/download). Weighted mean differences, risk ratios (RRs), and 95% confidence intervals (CIs) were calculated. Cochrane Collaboration’s risk of bias tool was used to assess the risk of bias. In this comprehensive meta-analysis, we examined the efficiency of neoadjuvant chemotherapy and neoadjuvant chemoradiotherapy for the treatment of cancer of the esophagus or the gastroesophageal junction as reported in qualified clinical trials. Results Six qualified articles that included a total of 866 patients were identified. The meta-analysis showed that for 3-year and 5-year survival rates in primary outcomes, the results favored neoadjuvant chemoradiotherapy strategies compared with neoadjuvant chemotherapy (RR = 0.78, 95% CI = 0.62–0.98, P = 0.03; RR = 0.69, 95% CI = 0.50–0.96, P = 0.03, respectively). In terms of secondary outcomes, neoadjuvant chemoradiotherapy significantly increased the rate of R0 resection and pathological complete response as well (RR = 0.87, 95% CI = 0.81–0.92, P < 0.0001; RR = 0.16, 95% CI = 0.09–0.28, P < 0.00001, respectively). However, there were no significant differences in postoperative mortality between the two groups (RR = 1.85, 95% CI = 0.93–3.65, P = 0.08). For the results of postoperative complications, revealed that there was a statistically significant difference between the two groups in the incidence of postoperative complications such as pulmonary, anastomotic leak and cardiovascular complications. The subgroup analysis of patients with esophageal adenocarcinoma or squamous cell carcinoma showed that both esophageal adenocarcinoma and squamous cell carcinoma patients achieved a high rate of R0 resection (RR = 0.85, 95% CI = 0.77–0.93, P = 0.0006; RR = 0.88, 95% CI = 0.81–0.96, P = 0.005, respectively) and pathological complete response benefit of neoadjuvant chemoradiotherapy (RR = 0.23, 95% CI = 0.09–0.57, P = 0.001; RR = 0.18, 95% CI = 0.03–0.96, P = 0.05, respectively). Conclusion Our findings suggested that compared with neoadjuvant chemotherapy, neoadjuvant chemoradiotherapy should be recommended with a significant long-term survival benefit in patients with cancer of the esophagus or the gastroesophageal junction. In view of the clinical heterogeneity, whether these conclusions are broadly applicable should be further determined.
Collapse
Affiliation(s)
- Xin Zhao
- Tianjin Medical University, Tianjin, China
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University, Tianjin, China
| | - Yiming Ren
- Department of Bone and Joint, Tianjin Union Medicine Center, Tianjin, PR China
| | - Yong Hu
- Tianjin Medical University, Tianjin, China
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University, Tianjin, China
| | - Naiqiang Cui
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University, Tianjin, China
| | - Ximo Wang
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University, Tianjin, China
| | - Yunfeng Cui
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University, Tianjin, China
- * E-mail:
| |
Collapse
|
320
|
Yamamoto K, Takano K, Kondo A, Kurose M, Obata K, Himi T. Clinical and Prognostic Analysis of Hypopharyngeal Squamous Cell Carcinoma with Synchronous and Metachronous Multiple Malignancies. ACTA ACUST UNITED AC 2018; 32:165-170. [PMID: 29275315 DOI: 10.21873/invivo.11220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM To analyze the clinical features and prevalence of synchronous and metachronous second primary malignancies (SPMs) in patients with hypopharyngeal squamous cell carcinoma (HSCC), their associated risk factors, and cause-specific mortality. PATIENTS AND METHODS We retrospectively reviewed 136 patients treated with curative intent at our hospital. Statistical analyses were performed to determine factors predictive of SPM and cause-specific mortality. RESULTS Sixty-three of 136 patients (46.3%) developed SPM; of these, 41 (30.1%) and 42 (30.9%) had synchronous and metachronous SPMs, respectively, with patient overlap. The most common site of synchronous and metachronous SPMs was the oesophagus (65.8% and 24.4%, respectively); the corresponding overall survival rates were 34.1% and 66.5%, respectively. Furthermore, heavy drinking was significantly correlated with synchronous SPM (p<0.001). CONCLUSION Oesophageal cancer surveillance is recommended for patients with HSCC, especially heavy drinkers. Our findings may help identify and properly manage HSCC patients at high risk of SPMs.
Collapse
Affiliation(s)
- Keisuke Yamamoto
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenichi Takano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kondo
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Kurose
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazufumi Obata
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuo Himi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
321
|
Ma S, Lu CC, Yang LY, Wang JJ, Wang BS, Cai HQ, Hao JJ, Xu X, Cai Y, Zhang Y, Wang MR. ANXA2 promotes esophageal cancer progression by activating MYC-HIF1A-VEGF axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:183. [PMID: 30081903 PMCID: PMC6091180 DOI: 10.1186/s13046-018-0851-y] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 07/20/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND ANXA2 (Annexin A2) is a pleiotropic calcium-dependent phospholipid binding protein that is abnormally expressed in various cancers. We previously found that ANXA2 is upregulated in esophageal squamous cell carcinoma (ESCC). This study was designed to investigate the functional significance of ANXA2 dysregulation and underlying mechanism in ESCC. METHODS Proliferation, migration, invasion and metastasis assay were performed to examine the functional roles of ANXA2 in ESCC cells in vitro and in vivo. Real-time RT-PCR, immunoblotting, ChIP, reporter assay, confocal-immunofluorescence staining, co-immunoprecipitation and ubiquitination assay were used to explore the molecular mechanism underlying the actions of deregulated ANXA2 in ESCC cells. RESULTS Overexpression of ANXA2 promoted ESCC cells migration and invasion in vitro and metastasis in vivo through activation of the MYC-HIF1A-VEGF cascade. Notably, ANXA2 phosphorylation at Tyr23 by SRC led to its translocation into the nucleus and enhanced the metastatic potential of ESCC cells. Phosphorylated ANXA2 (Tyr23) interacted with MYC and inhibited ubiquitin-dependent proteasomal degradation of MYC protein. Accumulated MYC directly potentiated HIF1A transcription and then activated VEGF expression. Correlation between these molecules were also found in ESCC tissues. Moreover, dasatinib in combination with bevacizumab or ANXA2-siRNA produced potent inhibitory effects on the growth of ESCC xenograft tumors in vivo. CONCLUSIONS This study provides evidence that highly expressed p-ANXA2 (Tyr23) contributes to ESCC progression by promoting migration, invasion and metastasis, and suggests that targeting the SRC-ANXA2-MYC-HIF1A-MYC axis may be an efficient strategy for ESCC treatment.
Collapse
Affiliation(s)
- Sai Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Chen-Chen Lu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.,Basic Medical College, Bengbu Medical College, Bengbu, 233003, China
| | - Li-Yan Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Juan-Juan Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Bo-Shi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China
| | - Hong-Qing Cai
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jia-Jie Hao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xin Xu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yan Cai
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Ming-Rong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
322
|
Liu TT, Yang KX, Yu J, Cao YY, Ren JS, Hao JJ, Pan BQ, Ma S, Yang LY, Cai Y, Wang MR, Zhang Y. Co-targeting PLK1 and mTOR induces synergistic inhibitory effects against esophageal squamous cell carcinoma. J Mol Med (Berl) 2018; 96:807-817. [PMID: 29959473 DOI: 10.1007/s00109-018-1663-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 06/13/2018] [Accepted: 06/19/2018] [Indexed: 12/19/2022]
Abstract
Both polo-like kinase 1 (PLK1) and mammalian/mechanistic target of rapamycin (mTOR) are attractive therapeutic targets for cancer therapy. However, the efficacy of the combined inhibition of both pathways for treating esophageal squamous cell carcinoma (ESCC), an aggressive malignancy with poor prognosis, remains unknown. In this study, we found that suppression of PLK1 by specific siRNA or inhibitor attenuated mTOR activity in ESCC cells. Phosphorylated S6, a downstream effector of mTOR signaling, was significantly correlated with overexpression of PLK1 in a subset of ESCC. These data suggest that PLK1 activates mTOR signaling in vitro and in vivo. More importantly, the mTOR inhibitor rapamycin synergized with PLK1 inhibitor BI 2536 to inhibit ESCC cell proliferation in culture and in mice. Notably, combined treatment with BI 2536 and rapamycin produced more potent inhibitory effects on the activation of S6 and AKT than either alone. Further analysis reveals that PLK1 modulates both mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) cascades. Therefore, dual inhibition of PLK1 and mTOR yields stronger antitumor effects, at least partially due to synergistic abrogated the activation of S6, eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), and AKT by cooperatively blocking mTORC1 and mTORC2 cascades. These results provide evidence that the mTOR inhibitor rapamycin synergistically enhances the antitumor effect of PLK1 inhibitor BI 2536 in ESCC cells. Simultaneous targeting of PLK1 and mTOR may thus be a novel and promising therapeutic strategy for ESCC. KEY MESSAGES PLK1 potentiates both mTORC1 and mTORC2 activities in ESCC cells. PLK1 expression positively correlated with mTOR activity in a subset of ESCC. Co-targeting of PLK1 and mTOR produced stronger antitumor effects partially due to synergistic inhibition of AKT, 4E-BP1 and S6 through cooperatively blocking mTORC2 and mTORC1 cascades. Combination targeting of PLK1 and mTOR may be a novel and promising therapeutic strategy for ESCC treatment.
Collapse
Affiliation(s)
- Ting-Ting Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Kai-Xia Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jing Yu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Ying-Ya Cao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jian-Song Ren
- Program Office for Cancer Screening in Urban China, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jia-Jie Hao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Bei-Qing Pan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Sai Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Li-Yan Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yan Cai
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Ming-Rong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Yu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
323
|
Lu Z, Zhang G, Zhang Y, Hua P, Fang M, Wu M, Liu T. Isoalantolactone induces apoptosis through reactive oxygen species-dependent upregulation of death receptor 5 in human esophageal cancer cells. Toxicol Appl Pharmacol 2018; 352:46-58. [DOI: 10.1016/j.taap.2018.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/15/2018] [Accepted: 05/21/2018] [Indexed: 01/20/2023]
|
324
|
Shi P, Sun J, He B, Song H, Li Z, Kong W, Wang J, Wang J, Xue H. Profiles of differentially expressed circRNAs in esophageal and breast cancer. Cancer Manag Res 2018; 10:2207-2221. [PMID: 30087579 PMCID: PMC6061203 DOI: 10.2147/cmar.s167863] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Introduction Circular RNAs (circRNAs) function as efficient microRNA sponges with gene-regulatory potential and are promising cancer biomarkers. In this study, we used the Arraystar Human circRNA Array to construct a genome-wide circRNA profile of esophageal squamous cell cancer (ESCC) and breast cancer (BC). Patients and methods Expression levels between cancer lesions and adjacent normal-appearing tissues were compared. We observed 469 upregulated circRNAs and 275 downregulated circRNAs in ESCC. Hsa_circRNA_103670 was upregulated 20.3-fold, while hsa_circRNA_030162 was downregulated 12.1-fold. For BC, 715 circRNAs were upregulated, and 440 circRNAs were downregulated. Hsa_circRNA_005230 was upregulated 12.2-fold, while hsa_circRNA_406225 was downregulated 12.4-fold. Results When we set the criteria as fold change in expression ≥2 between cancer and adjacent normal-appearing tissue with a P-value <0.01, there were 22 common circRNAs (11 upregulated and 11 downregulated) in relation to both ESCC and BC. Gene ontology and the Kyoto encyclopedia of genes and genomes analyses showed that these circRNAs were involved in the tumorigenesis of human cancers. Conclusion Our study revealed that circRNAs are promising candidates as valuable biomarkers for ESCC and BC, although relevant research is still in its infancy and the functional role of specific circRNAs in tumorigenesis is just starting to be elucidated.
Collapse
Affiliation(s)
- Peiyi Shi
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China,
| | - Jian Sun
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, Yancheng, People's Republic of China
| | - Biyu He
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China,
| | - Huan Song
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China,
| | - Zhongqi Li
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China,
| | - Weimin Kong
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, Yancheng, People's Republic of China
| | - Jianping Wang
- Department of Thoracic Surgery, People's Hospital of Yangzhong, Yangzhong, People's Republic of China,
| | - Jianming Wang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China,
| | - Hengchuan Xue
- Department of Thoracic Surgery, People's Hospital of Yangzhong, Yangzhong, People's Republic of China,
| |
Collapse
|
325
|
Koizumi S, Motoyama S, Iijima K. Is the incidence of esophageal adenocarcinoma increasing in Japan? Trends from the data of a hospital-based registration system in Akita Prefecture, Japan. J Gastroenterol 2018; 53:827-833. [PMID: 29134330 DOI: 10.1007/s00535-017-1412-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND At present, squamous cell carcinoma accounts for most esophageal cancers in Japan; however, there is concern that the incidence of esophageal adenocarcinoma in Japan is increasing in comparison to Western countries. Akita Prefecture, with a population of 1 million, has the highest rate of esophageal cancer-associated mortality in Japan; the vast majority of these cases are squamous cell carcinoma. The aim of the present study was to investigate the latest trends in the incidence of esophageal adenocarcinoma in Akita Prefecture using a registration system. METHODS We examined the data from a collaborative hospital-based registration system for cancer patients. Eleven of the main hospitals in Akita Prefecture participate in this system, which covers 90% of the cancer cases in the region and which provides near population-based data. Using this registration system, we examined the sequential trends in the percentage of adenocarcinoma cases in relation to total esophageal cancer cases in Akita Prefecture from 2007 to 2014. RESULTS In 2007-2014, a total of 2527 esophageal cancer patients were registered in the system. Among these, only 43 (1.8%) had adenocarcinoma. However, the number of adenocarcinoma patients gradually increased from 1 case in 2007 to 14 cases in 2014. Consequently, the percentage of adenocarcinoma cases showed a significant increase from 0.66% in the first 4 years of the study period to 2.5% in the latter 4 years of the study period (P < 0.01). CONCLUSIONS The latest hospital-based registration data for 2007-2014 suggest that the incidence of esophageal adenocarcinoma may have begun to increase in Akita Prefecture; this may reflect recent trends in other parts of Japan.
Collapse
Affiliation(s)
- Shigeto Koizumi
- Department of Gastroenterology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Japan
| | - Satoru Motoyama
- Department of Esophageal Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Katsunori Iijima
- Department of Gastroenterology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Japan.
| |
Collapse
|
326
|
Fankhauser CD, Mostafid H. Genetic polymorphisms may explain association between alcohol consumption and bladder cancer risk in East Asian men. Transl Androl Urol 2018; 7:S252-S254. [PMID: 29928627 PMCID: PMC5989115 DOI: 10.21037/tau.2018.04.17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
| | - Hugh Mostafid
- Department of Urology, Royal Surrey County Hospital, Egerton Road, Guildford, Surrey, UK
| |
Collapse
|
327
|
Huang B, Zhong N, Xia L, Yu G, Cao H. Sparse Representation-Based Patient-Specific Diagnosis and Treatment for Esophageal Squamous Cell Carcinoma. Bull Math Biol 2018; 80:2124-2136. [PMID: 29869044 DOI: 10.1007/s11538-018-0449-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 05/25/2018] [Indexed: 11/28/2022]
Abstract
Precision medicine and personalized treatment have attracted attention in recent years. However, most genetic medicines mainly target one genetic site, while complex diseases like esophageal squamous cell carcinoma (ESCC) usually present heterogeneity that involves variations of many genetic markers. Here, we seek an approach to leverage genetic data and ESCC knowledge data to forward personalized diagnosis and treatment for ESCC. First, 851 ESCC-related gene markers and their druggability were studied through a comprehensive literature analysis. Then, a sparse representation-based variable selection (SRVS) was employed for patient-specific genetic marker selection using gene expression datasets. Results showed that the SRVS method could identify a unique gene vector for each patient group, leading to significantly higher classification accuracies compared to randomly selected genes (100, 97.17, 100, 100%; permutation p values: 0.0032, 0.0008, 0.0004, and 0.0008). The SRVS also outperformed an ANOVA-based gene selection method in terms of the classification ratio. The patient-specific gene markers are targets of ESCC effective drugs, providing specific guidance for medicine selection. Our results suggest the effectiveness of integrating previous database utilizing SRVS in assisting personalized medicine selection and treatment for ESCC.
Collapse
Affiliation(s)
- Bin Huang
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Southeast University Medical College, No. 163 Shoushan Rd, Jiangyin, 214400, Jiangsu, China
| | - Ning Zhong
- Department of Cardiothoracic Surgery, The First People's Hospital of Kunshan, Kunshan, 215300, Jiangsu, China
| | - Lili Xia
- Department of Ultrasound, The People's Hospital of Tongling, Tongling, 215300, Anhui, China
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Southeast University Medical College, No. 163 Shoushan Rd, Jiangyin, 214400, Jiangsu, China.
| | - Hongbao Cao
- Department of Genomics Research, R&D Solutions, Elsevier Inc., Rockville, MD, 20852, USA. .,Unit on Statistical Genomics, National Institute of Health (NIH), Bethesda, MD, 20892, USA.
| |
Collapse
|
328
|
Surface Modification of Esophageal Stent Materials by a Drug-Eluting Layer for Better Anti-Restenosis Function. COATINGS 2018. [DOI: 10.3390/coatings8060215] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
329
|
Zheng H, Tang H, Wang H, Fang Y, Shen Y, Feng M, Xu S, Fan H, Ge D, Wang Q, Tan L. Nomogram to predict lymph node metastasis in patients with early oesophageal squamous cell carcinoma. Br J Surg 2018; 105:1464-1470. [PMID: 29863776 DOI: 10.1002/bjs.10882] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/11/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
Abstract
Background
Lymph node status is crucial in determining the prognosis for early oesophageal squamous cell carcinoma (SCC). This study aimed to develop and validate a nomogram for the prediction of lymph node metastasis in patients with early SCC.
Methods
A prediction model was developed in a derivation cohort of patients with clinicopathologically confirmed early SCC. Patients who underwent oesophagectomy for pT1 SCC between January 2010 and December 2013 were identified from an institutional database. Risk factors for lymph node metastasis were assessed using a binary logistic regression modelling technique. A nomogram for the prediction of lymph node metastasis was constructed using the results of multivariable analyses. For internal validation, bootstraps with 1000 resamples were performed. The predictive performance of the nomogram was measured by Harrell's concordance index (C-index). An independent cohort from the same hospital was used to validate the nomogram. This cohort included consecutive patients with early SCC who underwent oesophagectomy from January 2014 to December 2015.
Results
The derivation cohort included 281 patients. Four variables associated with lymph node metastasis were included in the model: depth of tumour invasion (odds ratio (OR) 4·37, 95 per cent c.i. 1·59 to 12·03; P = 0·004), grade of differentiation (OR 4·47, 1·02 to 19·70; P = 0·048), tumour size (OR 2·52, 1·11 to 5·75; P = 0·028) and lymphovascular invasion (OR 6·58, 2·54 to 17·05; P < 0·001). The C-index was 0·790 (95 per cent c.i. 0·717 to 0·864) in the derivation cohort and 0·789 (0·709 to 0·869) for the validation cohort (198 patients).
Conclusion
A validated nomogram for patients with early oesophageal SCC can predict the risk of lymph node metastasis.
Collapse
Affiliation(s)
- H Zheng
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - H Tang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - H Wang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Y Fang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Y Shen
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - M Feng
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - S Xu
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - H Fan
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - D Ge
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Q Wang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - L Tan
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| |
Collapse
|
330
|
Gu MM, Li M, Gao D, Liu LH, Lang Y, Yang SM, Ou H, Huang B, Zhou PK, Shang ZF. The vanillin derivative 6-bromine-5-hydroxy-4-methoxybenzaldehyde induces aberrant mitotic progression and enhances radio-sensitivity accompanying suppression the expression of PLK1 in esophageal squamous cell carcinoma. Toxicol Appl Pharmacol 2018; 348:76-84. [PMID: 29679654 DOI: 10.1016/j.taap.2018.04.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common form of esophageal cancer in China. Since chemotherapy is the standard clinical intervention for advanced ESCC, the development of highly effective and minimal/non-toxic drugs is essential to improve the clinical outcome and prognosis of the patients. A novel derivative of vanillin, 6-bromine-5-hydroxy-4-methoxybenzaldehyde (BVAN08), has been recently reported to activate different cell death pathways in cancer cells. In this study, we demonstrate that BVAN08 exhibits a potent anti-proliferation effect on ESCC cells (TE-1 and ECA-109) by inhibiting the expression of PLK1, an important mitotic kinase. Consistent with this, BVAN08 induces mitotic arrest and chromosomal misalignment in ESCC cells. The disruption of microtubule nucleation around centrosomes is also observed in BVAN08 treated ESCC cells. Furthermore, BVAN08 enhances radio-sensitivity of ESCC cells by prolonging DNA damage repair. These findings underscore the potential value of BVAN08 in cancer therapeutics and demonstrate the underlying mechanism by which BVAN08 induces mitotic catastrophe and enhances radio-sensitivity in ESCC cells.
Collapse
Affiliation(s)
- Meng-Meng Gu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China
| | - Ming Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China
| | - Dexuan Gao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, PR China
| | - Lang-Huan Liu
- School of Public Heath, Central South University, Changsha 410078, PR China
| | - Yue Lang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China
| | - Si-Ming Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China
| | - Hongling Ou
- Department of Clinical Laboratory, The General Hospital of the PLA Rocket Force, Beijing 100088, PR China
| | - Bo Huang
- School of Public Heath, Central South University, Changsha 410078, PR China
| | - Ping-Kun Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China; Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, PR China.
| | - Zeng-Fu Shang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
331
|
Sun H, Cai X, Zhou H, Li X, Du Z, Zou H, Wu J, Xie L, Cheng Y, Xie W, Lu X, Xu L, Chen L, Li E, Wu B. The protein-protein interaction network and clinical significance of heat-shock proteins in esophageal squamous cell carcinoma. Amino Acids 2018; 50:685-697. [PMID: 29700654 DOI: 10.1007/s00726-018-2569-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/09/2018] [Indexed: 02/06/2023]
Abstract
Heat-shock proteins (HSPs), one of the evolutionarily conserved protein families, are widely found in various organisms, and play important physiological functions. Nevertheless, HSPs have not been systematically analyzed in esophageal squamous cell carcinoma (ESCC). In this study, we applied the protein-protein interaction (PPI) network methodology to explore the characteristics of HSPs, and integrate their expression in ESCC. First, differentially expressed HSPs in ESCC were identified from our previous RNA-seq data. By constructing a specific PPI network, we found differentially expressed HSPs interacted with hundreds of neighboring proteins. Subcellular localization analyses demonstrated that HSPs and their interacting proteins distributed in multiple layers, from membrane to nucleus. Functional enrichment annotation analyses revealed known and potential functions for HSPs. KEGG pathway analyses identified four significant enrichment pathways. Moreover, three HSPs (DNAJC5B, HSPA1B, and HSPH1) could serve as promising targets for prognostic prediction in ESCC, suggesting these HSPs might play a significant role in the development of ESCC. These multiple bioinformatics analyses have provided a comprehensive view of the roles of heat-shock proteins in esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Hong Sun
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Xinyi Cai
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Haofeng Zhou
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Xiaoqi Li
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Zepeng Du
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, 515041, China
| | - Haiying Zou
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Jianyi Wu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Lei Xie
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China
| | - Yinwei Cheng
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, China
| | - Wenming Xie
- Network and Information Center, Shantou University Medical College, Shantou, 515041, China
| | - Xiaomei Lu
- Tumor Hospital Affiliated to Xinjiang Medical University, Ürümqi, 830054, Xinjiang Uygur Autonomous Region, China
| | - Liyan Xu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, 515041, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Sichuan, 610041, China
| | - Enmin Li
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China.
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China.
| | - Bingli Wu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China.
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
332
|
Shahin MI, Roy J, Hanafi M, Wang D, Luesakul U, Chai Y, Muangsin N, Lasheen DS, Abou El Ella DA, Abouzid KA, Neamati N. Synthesis and biological evaluation of novel 2-oxo-1,2-dihydroquinoline-4-carboxamide derivatives for the treatment of esophageal squamous cell carcinoma. Eur J Med Chem 2018; 155:516-530. [PMID: 29908444 DOI: 10.1016/j.ejmech.2018.05.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/21/2018] [Accepted: 05/25/2018] [Indexed: 01/07/2023]
Abstract
No new and effective treatments have been approved for the treatment of esophageal squamous cell carcinoma (ESCC) in the past decade. Cisplatin and 5-fluoruracil are the most commonly used drugs for this disease. In order to develop a new class of drugs effective in our ESCC phenotypic screens, we began a systematic approach to generate novel compounds based on the 2-oxo-1,2-dihydroquinoline-4-carboxamide fragment. Herein, we report on the synthesis and initial assessment of 55 new analogues in two ESCC cell lines. Some of the active analogues with IC50 values around 10 μM were tested in three additional cell lines. Our structure-activity relationships revealed remarkable alterations in the anti proliferative activities upon modest chemical modifications and autophagy modulation is a suggested mechanism of action.
Collapse
Affiliation(s)
- Mai I Shahin
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, MI, 48109, United States; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Joyeeta Roy
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, MI, 48109, United States
| | - Maha Hanafi
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, MI, 48109, United States; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Dongyao Wang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, MI, 48109, United States; School of Pharmacy, Second Military Medical University, 200433, Shanghai, China
| | - Urarika Luesakul
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, MI, 48109, United States; Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, 200433, Shanghai, China
| | - Nongnuj Muangsin
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Deena S Lasheen
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Dalal A Abou El Ella
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Nahda University, 62511, Beni Suef, Egypt
| | - Khaled A Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, North Campus Research Complex, 1600 Huron Parkway, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
333
|
Wu Y, Chen X, Liang Y, Li J, Zhang K, Dai L, Guan X, Wang K, Bai Y. Overexpression of long non-coding RNA SOX2OT promotes esophageal squamous cell carcinoma growth. Cancer Cell Int 2018; 18:76. [PMID: 29849506 PMCID: PMC5970475 DOI: 10.1186/s12935-018-0570-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/12/2018] [Indexed: 11/10/2022] Open
Abstract
Background SOX2 overlapping transcript (SOX2OT) has been reported to be an important lncRNA in various cancers. SOX2 is embedded in an intron of the SOX2OT gene. But the role of SOX2OT in esophageal squamous cell carcinoma (ESCC) and the association between SOX2OT and SOX2 remain unclear. Methods Quantitative PCR (qPCR) was used to detect the expression of SOX2OT and SOX2 in ESCC tissues and cells. The isoforms of SOX2OT were identified by PCR and confirmed by sequencing. CCK-8 and Edu assays were performed to investigate the effects of SOX2OT on cell growth. The relationship between SOX2OT and SOX2 was explored by luciferase reporter assay. Results Both SOX2OT and SOX2 were upregulated in ESCC tissues and cells. SOX2OT expression was positively associated with SOX2 expression in ESCC tissues. NR_004053 was one of the major SOX2OT transcripts aberrantly expressed in ESCC tissues and cells. Overexpression of SOX2OT (NR_004053) promoted ESCC cell growth, antagonized the effect of DDP and increased cell proliferation ratio. Ectopic expression of SOX2 could increase the luciferase activity of SOX2OT-pGL3/Basic and SOX2OT expression, while overexpression of SOX2OT (NR_004053) had no effect on SOX2 expression. Conclusion Our study demonstrates that the major isoform of SOX2OT in ESCC, SOX2OT (NR_004053) contributes to cell growth. SOX2 promotes SOX2OT expression at transcriptional level.
Collapse
Affiliation(s)
- Yuanyuan Wu
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Xuedan Chen
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Yan Liang
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Juan Li
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Kun Zhang
- 2Department of Pathgenic Biology, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Limeng Dai
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Xingying Guan
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Kai Wang
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| | - Yun Bai
- 1Department of Medical Genetics, College of Basic Medical Science, Third Military Medical University or Army Medical University, Chongqing, China
| |
Collapse
|
334
|
Liu X, Zhao Y, Zhang W, Gao Y, Huo M, Liu M, Xiao Z, Liang S, Xu N, Zhu H. Inhibition of survivin enhances radiosensitivity of esophageal cancer cells by switching radiation-induced senescence to apoptosis. Onco Targets Ther 2018; 11:3087-3100. [PMID: 29872320 PMCID: PMC5975611 DOI: 10.2147/ott.s166798] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose Strategies to increase radiosensitivity are urgently needed. Combining radiosensitizing reagents with radiotherapy could improve the outcome of cancer treatment. Some preclinical studies showed that sepantronium bromide (YM155) could sensitize cancer cells to radiation by inhibiting the survivin protein. In this study, we try to investigate the function of YM155 on radiosensitivity of esophageal squamous cell carcinoma (ESCC) cells. Materials and methods ESCC cell lines were treated with radiation and YM155, and the radiation efficacy was evaluated by cell counting kit-8 assay and clonogenic survival assay. Cell senescence was measured by senescence-associated β-galactosidase staining. Terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay, fluorescein isothiocyanate-labeled Annexin V/propidium iodide assay, and poly ADP-ribose polymerase cleavage were used to detect apoptosis. KYSE150 xenografts model was used to test the efficacy of radiation combined with YM155. Results YM155 could inhibit the upregulation of survivin induced by radiation in all ESCC cell lines, but the efficacy of radiosensitization varied in different cell lines. Radiation-induced senescence in KYSE150 and KYSE410 cells, and the combination with YM155 inhibited senescence and promoted apoptosis of ESCC cells, thereby enhancing radiosensitivity. Combination with YM155 and radiation delayed the growth of KYSE150 xenografts in nude mice by switching radiation-induced senescence to apoptosis. When p21 was inhibited in KYSE150 cells, radiation did not induce senescence, and the radiosensitization of YM155 was also attenuated. In KYSE510 and KYSE180 cells, radiation did not induce senescence, and YM155 could not enhance the radiosensitivity. Conclusion Our results suggest a new mechanism that YM155 might sensitize ESCC cells to radiation by switching radiation-induced senescence to apoptosis. The major determinant of radiosensitization by YM155 might be the induction of senescence by radiation.
Collapse
Affiliation(s)
- Xianghe Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yahui Zhao
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weina Zhang
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Gao
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miaomiao Huo
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zefen Xiao
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
335
|
Xu J, Pan X, Hu Z. MiR-502 mediates esophageal cancer cell TE1 proliferation by promoting AKT phosphorylation. Biochem Biophys Res Commun 2018; 501:119-123. [PMID: 29709473 DOI: 10.1016/j.bbrc.2018.04.188] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/24/2018] [Indexed: 01/08/2023]
Abstract
Esophageal cancer is one of the most common cancers in the world and esophageal squamous cell carcinoma is one of the two main types in esophageal cancer. MicroRNA is a small non-coding RNA molecule functions in many different cancers including esophageal cancer. We found miR-502 was up-regulated in esophageal tissues, which indicated miRNA-502 may play important roles in esophageal cancer. In this study, we used esophageal cancer cell line TE1 as an in vitro model for investigating the role of miR-502 in promoting the proliferation of the cancer cells. We found that overexpressing miR-502 in TE1 cells promoted the proliferation and inhibited the apoptosis induced by dox. Down-regulating miR-502 made the opposite phenomenon. Furthermore, western blot showed that miR-502 enhanced the phosphorylation levels of AKT pathways, which may be the mechanism of the overgrowth for esophageal cancer cell. Our data provide the evidence of a role for miR-502 in the regulation the proliferation of esophageal cancer cell through promoting the phosphorylation of AKT signaling. Due to its ability to promote the overgrowth of esophageal cancer cell, miR-502 may be a novel target for esophageal cancer therapeutic.
Collapse
Affiliation(s)
- Jing Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, PR China.
| | - Xiongxiong Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, PR China.
| | - Zhendong Hu
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, 210000, PR China.
| |
Collapse
|
336
|
Shi H, Yu F, Mao Y, Ju Q, Wu Y, Bai W, Wang P, Xu R, Jiang M, Shi J. EphA2 chimeric antigen receptor-modified T cells for the immunotherapy of esophageal squamous cell carcinoma. J Thorac Dis 2018; 10:2779-2788. [PMID: 29997940 DOI: 10.21037/jtd.2018.04.91] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background It is urgent to explore an effective potential therapeutic strategy for ESCC. In recent years, cell-based cancer immunotherapy has become a potentially close for carcinoma therapy. Chimeric antigen receptor (CAR) T cell technology is a kind of adoptive cell therapy technique which has been developed rapidly. We sought to obtain EphA2.CAR-T cell and revealed the ability of EphA2.CAR-T cells to kill esophageal squamous cell carcinoma (ESCC) cells in vitro. Methods Firstly, the expression and location of EphA2 in ESCC tissues and cells was tested by immunohistochemistry staining and Western blot. Secondly, the second generation of EphA2.CAR was constructed via molecular biology technology, and transduced into T cells to obtain the EphA2.CAR-T cell. The transduction efficacies were assessed using flow cytometry (FCM). Thirdly, the effect of cell killing of EphA2.CAR-T cell on ESCC cells in vitro was detected by co-culture experiments. The productions of cytokines (TNF-α and IFN-γ) by EphA2.CAR-T cell after co-culture with ESCC cells were analyzed by ELISA assay. Results The expression of EphA2 was significantly upregulated in ESCC tissues and cells (P<0.05). EphA2 was expressed on the membrane of ESCC cells, so it could be served as tumor-associated surface antigens (TAA) of CAR for ESCC treatment. The EphA2.CAR-T cell was obtained successfully, and its' transduction efficacies was 61.4% by FCM. The ability of cell killing of EphA2.CAR-T cell was better than that of T cells (P<0.01), and demonstrated a dose-dependent cell killing. The results of ELISA assay showed that the levels of TNF-α and IFN-γ in EphA2.CAR-T cells were notably raised compared with T cells (P<0.05). Conclusions We firstly constructed the second generation of EphA2.CAR and established EphA2.CAR-T cells. The EphA2.CAR-T cells showed a dose-dependent cell killing of ESCC cells, and promoted the production of cytokines in vitro. These findings open a new way for treatment of ESCC by immunotherapy in the future.
Collapse
Affiliation(s)
- Hui Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Feng Yu
- Institute of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Yinting Mao
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Qianqian Ju
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yingcheng Wu
- Medical College, Nantong University, Nantong 226001, China
| | - Wen Bai
- Medical College, Nantong University, Nantong 226001, China
| | - Peiwen Wang
- Medical College, Nantong University, Nantong 226001, China
| | - Ran Xu
- Medical College, Nantong University, Nantong 226001, China
| | - Maorong Jiang
- Laboratory Animals Center, Nantong University, Nantong 226001, China
| | - Jiahai Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
337
|
Jesinghaus M, Steiger K, Slotta-Huspenina J, Drecoll E, Pfarr N, Meyer P, Konukiewitz B, Bettstetter M, Wieczorek K, Ott K, Feith M, Langer R, Weichert W, Specht K, Boxberg M. Increased intraepithelial CD3+ T-lymphocytes and high PD-L1 expression on tumor cells are associated with a favorable prognosis in esophageal squamous cell carcinoma and allow prognostic immunogenic subgrouping. Oncotarget 2018; 8:46756-46768. [PMID: 28657901 PMCID: PMC5564521 DOI: 10.18632/oncotarget.18606] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/08/2017] [Indexed: 11/25/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common esophageal cancer associated with poor prognosis and additional therapeutic strategies must be implemented to optimize ESCC treatment. Meanwhile, the important biologic role and potential prognostic and therapeutic implications of a tumors immunologic microenvironment (IM) have been recognized in various cancers.In order to investigate the contexture and the prognostic relevance of the IM in ESCC, we immunohistochemically evaluated the extent of overall/intraepithelial TILs (CD3+/CD8+) and of PD-1 / PD-L1 expression in a cohort of 125 therapy-naive ESCCs, additionally assessing PD-L1 copy number status via fluorescence in-situ hybridization.High intraepithelial CD3+ TILs (CD3ihigh) and high PD-L1 expression on tumor cells (PD-L1high) were each significantly associated with improved overall- (OS) (CD3+: p = 0.019; PD-L1: p = 0.028), disease specific- (DSS) (CD3+: p = 0.05; PD-L1: p = 0.006) and disease free survival (DFS) (CD3+: p = 0.009; PD-L1: p < 0.001). CD3ihigh- and PD-L1high cases were significantly associated with one another (p < 0.001). Subgrouping of ESCC revealed decreased OS (p = 0.031), DSS (p = 0.012) and DFS (p < 0.001) for CD3ilow/PD-L1low cancers.Our data not only associate CD3ihigh- and PD-L1high ESCC with a beneficial outcome, but also demonstrate PD-L1high- and CD3ihigh status to be closely intertwined. Furthermore, our study demarcates a prognostically unfavorable, "non-immunoreactive" CD3ilow / PD-L1low ESCC-subgroup, potentially forming the basis for an immune-based stratification of ESCC.
Collapse
Affiliation(s)
- Moritz Jesinghaus
- Institute of Pathology, Technical University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | | | - Enken Drecoll
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Nicole Pfarr
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Petra Meyer
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Björn Konukiewitz
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | | | - Kathrin Wieczorek
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Katja Ott
- RoMED Surgical Clinic, Rosenheim, Germany
| | - Markus Feith
- Department of Surgery, Technical University of Munich, Munich, Germany
| | - Rupert Langer
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Munich, Germany
| | - Katja Specht
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Melanie Boxberg
- Institute of Pathology, Technical University of Munich, Munich, Germany
| |
Collapse
|
338
|
Li X, Chen D, Li M, Gao X, Shi G, Zhao H. The CADM2/Akt pathway is involved in the inhibitory effect of miR-21-5p downregulation on proliferation and apoptosis in esophageal squamous cell carcinoma cells. Chem Biol Interact 2018; 288:76-82. [PMID: 29680210 DOI: 10.1016/j.cbi.2018.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/27/2018] [Accepted: 04/18/2018] [Indexed: 02/08/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC), the main subtype of esophageal cancer, is the eighth most common cancer worldwide. Cell adhesion molecule 2 (CADM2) has been reported to be a tumor suppressor and is usually downregulated in several cancers. However, the role of CADM2 in ESCC remains unknown. The aim of the present study was to evaluate the potential role and underlying action mechanism of CADM2 in ESCC. Herein, we found that CADM2 was low-expressed in ESCC tissues and cell lines. CADM2 overexpression inhibited proliferation and induced apoptosis of ESCC cells. Moreover, CADM2 overexpression also suppressed the Akt signaling pathway in ESCC cells. MiR-21-5p down-regulation inhibited cell proliferation and induced cell apoptosis, while CADM2 knockdown attenuated the effect of anti-miR-21-5p. The expression of p-Akt was decreased in the cells transfected with anti-miR-21. However, the expression of p-Akt was increased in the cells co-transfected with anti-miR-21-5p and si-CADM2 compared with that in anti-miR-21-5p-transfecting cells. In summary, the CADM2/Akt pathway is involved in the inhibitory effect of miR-21-5p downregulation on proliferation and apoptosis in ESCC cells. These findings indicated that the miR-21-5p/CADM2/Akt axis might be a new approach for the treatment of ESCC.
Collapse
Affiliation(s)
- Xiaohui Li
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Dong Chen
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Mengfei Li
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Xiang Gao
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Gongning Shi
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifeng 475000, PR China
| | - Hui Zhao
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University, Kaifeng 475000, PR China.
| |
Collapse
|
339
|
Essential role of Na+/Ca2+ exchanger 1 in smoking-induced growth and migration of esophageal squamous cell carcinoma. Oncotarget 2018; 7:63816-63828. [PMID: 27588478 PMCID: PMC5325406 DOI: 10.18632/oncotarget.11695] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/24/2016] [Indexed: 12/22/2022] Open
Abstract
Tobacco-derived carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is a major environmental risk factor for the pathogenesis of human esophageal squamous cell carcinoma (ESCC). However, the molecular mechanisms by which tobacco induces ESCC are not well understood. Na+/Ca2+ exchanger 1 (NCX1) is a plasma membrane transporter protein that plays an essential role in maintaining cytosolic Ca2+ ([Ca2+]cyt) homeostasis under physiological conditions and is implicated in tumorigenesis as well. In this study, we found that NCX1 expression was significantly higher in ESCC primary tissues compared to the noncancerous tissues and was overexpressed in tumor samples from the smoking patients. The expression of NCX1 proteins was also significantly higher in human ESCC cell lines compared to normal esophageal epithelial cell line. Moreover, NNK potentiated the [Ca2+]cyt signaling induced by removal of extracellular Na+, which was abolished by KB-R7943 or SN-6. NNK dose-dependently promoted proliferation and migration of human ESCC cells induced by NCX1 activation. Therefore, NCX1 expression correlates with the smoking status of ESCC patients, and NNK activates the Ca2+ entry mode of NCX1 in ESCC cells, leading to cell proliferation and migration. Our findings suggest NCX1 protein is a novel potential target for ESCC therapy.
Collapse
|
340
|
Okata H, Hatta W, Iijima K, Asanuma K, Tsuruya A, Asano N, Koike T, Hamada S, Nakayama T, Masamune A, Shimosegawa T. Detection of Acetaldehyde in the Esophageal Tissue among Healthy Male Subjects after Ethanol Drinking and Subsequent L-Cysteine Intake. TOHOKU J EXP MED 2018; 244:317-325. [PMID: 29681559 DOI: 10.1620/tjem.244.317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ethanol is oxidized by alcohol dehydrogenase to acetaldehyde, a recognized carcinogen for the esophagus. However, no previous study has measured the acetaldehyde levels in the esophageal tissue. L-cysteine has been shown to reduce the acetaldehyde levels in the saliva; however, it is unknown whether L-cysteine intake affects the acetaldehyde concentration in the esophageal tissue. The aim of this study was to measure the acetaldehyde concentration in the esophageal tissue after ethanol drinking and evaluate the effect of L-cysteine intake on the acetaldehyde levels in the esophagus. We enrolled 10 male subjects with active acetaldehyde dehydrogenase-2*1/*1 (ALDH2*1/*1) genotype and 10 male subjects with the inactive acetaldehyde dehydrogenase-2*1/*2 (ALDH2*1/*2) genotype, the mean ages of whom were 25.6 and 27.9 years, respectively. In this prospective, single-blind, placebo-controlled study using L-cysteine and placebo lozenges (first and second examination), saliva and blood were collected before and after ethanol drinking. Esophageal tissue was obtained by endoscopic biopsy at 60 minutes after drinking, and the acetaldehyde and ethanol concentrations were measured. The acetaldehyde concentration of the saliva was significantly lower in those taking L-cysteine than in those taking the placebo. Acetaldehyde in the esophageal tissue was detected only in those taking L-cysteine lozenges. There were no correlations between the acetaldehyde concentrations in the esophageal tissue and saliva or blood. In conclusion, we detected acetaldehyde in the human esophageal tissue after ethanol drinking. Unexpectedly, intake of L-cysteine lozenges appears to contribute to detection of acetaldehyde in the esophageal tissue.
Collapse
Affiliation(s)
- Hideki Okata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| | - Katsunori Iijima
- Department of Gastroenterology and Neurology, Akita University Graduate School of Medicine
| | - Kiyotaka Asanuma
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| | - Atsuki Tsuruya
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University
| | - Naoki Asano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| | - Shin Hamada
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| | - Toru Nakayama
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine
| |
Collapse
|
341
|
Moghadam AR, Mehramiz M, Entezari M, Aboutalebi H, Kohansal F, Dadjoo P, Fiuji H, Nasiri M, Aledavood SA, Anvari K, Simab SA, Khorrami MS, Moradi A, Hassanian SM, Ferns GA, Sales SS, Avan A. A genetic polymorphism in the CYP1B1 gene in patients with squamous cell carcinoma of the esophagus: an Iranian Mashhad cohort study recruited over 10 years. Pharmacogenomics 2018; 19:539-546. [DOI: 10.2217/pgs-2018-0197] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aim: Esophageal cancer is the eighth most common cancer globally and the seventh most common cause of cancer-related deaths in men. Recent studies have shown that CYP450, family 1, subfamily B, polypeptide 1, which plays a role in the metabolism of xenobiotics, is associated with several cancers. Therefore, in the present study we investigated the association between a genetic variant, CYP1B1-rs1056836 gene, with the clinical characteristics of patients with squamous cell carcinoma of the esophagus (ESCC). Method: In this study, 117 patients with ESCC and 208 healthy controls were recruited. DNA was extracted and genotyped using real-time PCR-based TaqMan. Kaplan–Meier curves were utilized to assess overall and progression-free survival. To evaluate the relationship between clinicopathological data, genotypic frequencies, disease prognosis and survival, Pearson's χ2 and t-test were used. Logistic regression was utilized to assess the association between the risk of ESCC and genotypes. Results: The genotypic frequency for GG, GC and CC were 58.6, 29.8 and 11.5%, respectively, in the healthy subjects and 51.8, 36.14 and 12% in the ESCC group. An association between the GG genotype and stage of ESCC was found. Also, statistically significant results were not found for this variation and risk of ESCC. Conclusion: Our findings suggest a relationship between the CYP1B1-rs1056836 genetic polymorphism and clinical features of ESCC, supporting further studies in larger populations in different ethnic groups, taking into account potentially important environmental factors such as diet.
Collapse
Affiliation(s)
- Azam Rastgar Moghadam
- Department of Genetics, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
- Department of Biology, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrane Mehramiz
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Entezari
- Department of Biology, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Hamideh Aboutalebi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Kohansal
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parisa Dadjoo
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Fiuji
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nasiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Iran
| | | | - Kazem Anvari
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohammad Sadegh Khorrami
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Moradi
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | | | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
342
|
Integrative analysis of mRNA and lncRNA profiles identified pathogenetic lncRNAs in esophageal squamous cell carcinoma. Gene 2018; 661:169-175. [PMID: 29604464 DOI: 10.1016/j.gene.2018.03.066] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/02/2018] [Accepted: 03/20/2018] [Indexed: 01/11/2023]
Abstract
Systems biology approaches can help understand pathogenesis of complex human diseases like cancers for identification of potential new therapeutic targets. Here in this study, we performed genome-wide screening for mRNA and lncRNA profiles in esophageal cancer to identify the novel cancer-related mRNA and lncRNA in esophageal squamous cell carcinoma (ESCC). We identified 1260 up-regulated/1445 down-regulated mRNAs and 402 up-regulated/741 down-regulated lncRNAs. Further functional analysis revealed differentially expressed genes (DEGs) of mRNA and lncRNA are related to different pathways. mRNA pathways are mainly involved in cell cycles while lncRNA pathways are for regulation and metabolic procession. Differentially expressed mRNAs/lncRNAs were validated with qPCR. At last, mRNA and lncRNA co-expression network were built and highly-connected networks were identified, which may provide a mechanism of mRNA expression regulation by lncRNA. In together, we used next generation sequencing data to explore the co-expression networks of lncRNA and mRNA, which may elucidate the functions and mechanisms of lncRNA in ESCC.
Collapse
|
343
|
Activation of the Dickkopf1-CKAP4 pathway is associated with poor prognosis of esophageal cancer and anti-CKAP4 antibody may be a new therapeutic drug. Oncogene 2018; 37:3471-3484. [PMID: 29563607 DOI: 10.1038/s41388-018-0179-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/15/2017] [Accepted: 11/08/2017] [Indexed: 12/17/2022]
Abstract
Aberrant expression of the secretory protein Dickkopf1 (DKK1) is associated with poor prognosis of esophageal squamous cell carcinoma (ESCC), but the underlying mechanism of how DKK1 is involved in aggressiveness of ESCC is not clear. In this study, we show that cytoskeleton-associated protein 4 (CKAP4) functions as a DKK1 receptor in ESCC cells. Immunohistochemical analyses of ESCC revealed that both DKK1 and CKAP4 are minimally expressed in associated normal esophageal squamous mucosa of non-tumor regions, but strongly expressed in tumor lesions. Forty-six of 119 cases (38.7%) were positive for both DKK1 and CKAP4. Those expressing both proteins showed poor prognosis and relapse-free survival. Multivariate analysis demonstrated that expression of both proteins was the poor prognostic factor. The Cancer Genome Atlas data set indicated that the mRNA levels of DKK1 and CKAP4 are significantly elevated in the tumor lesions compared to non-tumor regions. DKK1 bound to CKAP4 at endogenous levels. DKK1 induced the internalization of CKAP4 from and its recycling to the plasma membrane. AKT was activated in ESCC cells in which DKK1 was highly expressed and CKAP4 was localized to the plasma membrane. Knockdown of either DKK1 or CKAP4 inhibited AKT activity and cell proliferation in vitro and xenograft tumor formation. Wild-type CKAP4 or DKK1, but not a DKK1 mutant that was unable to bind to CKAP4, rescued phenotypes induced by CKAP4 or DKK1 knockdown, respectively. The anti-CKAP4 antibody also inhibited AKT activity and suppressed xenograft tumor formation. In contrast, in ESCC cells in which DKK1 was marginally expressed, knockdown of CKAP4 or anti-CKAP4 antibody affected neither AKT activity nor cell proliferation. These findings suggest that the DKK1-CKAP4 pathway promotes ESCC cell proliferation and that CKAP4 might represent a novel therapeutic target for ESCCs expressing both DKK1 and CKAP4.
Collapse
|
344
|
A six-microRNA signature in plasma was identified as a potential biomarker in diagnosis of esophageal squamous cell carcinoma. Oncotarget 2018; 8:34468-34480. [PMID: 28380431 PMCID: PMC5470983 DOI: 10.18632/oncotarget.16519] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/15/2017] [Indexed: 12/22/2022] Open
Abstract
The differential expression of microRNAs (miRNAs) in plasma of esophageal squamous cell carcinoma (ESCC) patients may serve as a diagnostic biomarker. A four-stage study was conducted to identify plasma miRNAs with potential in detecting ESCC. Exiqon panels (2 ESCC pools vs. 1 normal control (NC) pool) were applied in the screening phase to obtain miRNA profiles. The identified miRNAs were further evaluated through training (36 ESCC VS. 42 NCs) and testing stages (101 ESCC VS. 113 NCs) with qRT-PCR assays. A six-miRNA signature including up-regulated miR-106a, miR-18a, miR-20b, miR-486-5p, miR-584 and down-regulated miR-223-3p in ESCC was identified. The signature could accurately discriminate ESCC patients from NCs with areas under the receiver operating characteristic curve of 0.935, 0.959 and 0.966 for the training, testing and the additional validation stage (41 ESCC VS. 50 NCs), respectively. MiR-106a and miR-584 were significantly up-regulated in tumor tissues with qRT-PCR assays. And miR-584 was also up-regulated in ESCC tissues from TCGA database. In addition, exosomal miR-223-3p and miR-584 were consistently dysregulated with those in plasma and could also act as biomarkers in diagnosis of ESCC. In conclusion, we identified a six-miRNA signature in plasma which could act as a non-invasive biomarker in detection of ESCC.
Collapse
|
345
|
Wang H, Yu J, Guo Y, Zhang Z, Liu G, Li J, Zhang X, Jin T, Wang Z. Genetic variants in the ZNF208 gene are associated with esophageal cancer in a Chinese Han population. Oncotarget 2018; 7:86829-86835. [PMID: 27907911 PMCID: PMC5349957 DOI: 10.18632/oncotarget.13468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/29/2016] [Indexed: 01/23/2023] Open
Abstract
Previous studies showed an association between the ZNF208 gene and gastric cancer. In this study, we investigated the association between single nucleotide polymorphisms (SNPs) in ZNF208 and the risk of esophageal cancer in a Chinese Han population. We conducted a case-control study that included 386 cases and 495 controls. Five SNPs were selected from previous genome-wide association studies and genotyped using the Sequenom MassARRAY platform. Unconditional logistic regression was used to calculate odds ratios and 95% confidence intervals after adjustment for age and gender. Logistic regressionl analysis showed that two SNPs (rs8103163 and rs7248488) were associated with an increased risk of esophageal cancer under different inheritance models after Bonferroni correction. Haplotype analysis suggested that the four variants comprised one block, and that the Grs2188972Crs2188971Crs8103163Crs7248488 haplotype was significantly correlated with an increased risk of esophageal cancer. Our data indicate that variants in ZNF208 are contribute to the susceptibility to esophageal cancer in a Chinese Han population.
Collapse
Affiliation(s)
- Huijie Wang
- Department of Intergrated Traditional Chinese and Western Medicine in Oncology, Affiliated Luoyang Central Hospital, Zhengzhou University, Luoyang 471000, China
| | - Jianzhong Yu
- Department of Neurology, Haikou People's Hospital, Haikou 570208, Hainan, China
| | - Yanling Guo
- Department of Intergrated Traditional Chinese and Western Medicine in Oncology, Affiliated Luoyang Central Hospital, Zhengzhou University, Luoyang 471000, China
| | - Zhengxing Zhang
- Department of Intergrated Traditional Chinese and Western Medicine in Oncology, Affiliated Luoyang Central Hospital, Zhengzhou University, Luoyang 471000, China
| | - Guoqi Liu
- Department of Intergrated Traditional Chinese and Western Medicine in Oncology, Affiliated Luoyang Central Hospital, Zhengzhou University, Luoyang 471000, China
| | - Jingjie Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Xiyang Zhang
- Xi'an Tiangen Precision Medical Institute, Xi'an, Shaanxi 710075, China
| | - Tianbo Jin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Zhaoxia Wang
- Department of Intergrated Traditional Chinese and Western Medicine in Oncology, Affiliated Luoyang Central Hospital, Zhengzhou University, Luoyang 471000, China
| |
Collapse
|
346
|
Wang M, An S, Wang D, Ji H, Geng M, Guo X, Wang Z. Quantitative Proteomics Identify the Possible Tumor Suppressive Role of Protease-Activated Receptor-4 in Esophageal Squamous Cell Carcinoma Cells. Pathol Oncol Res 2018; 25:937-943. [PMID: 29502225 DOI: 10.1007/s12253-018-0395-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/21/2018] [Indexed: 12/21/2022]
Abstract
Exposure to carcinogens of tobacco smoke may result in methylation of protease-activated receptors-4 (PAR4) gene and further induces the loss of PAR4 expression, which is considered to be involved in carcinogenesis of esophageal squamous cell carcinoma (ESCC). Here we employed a TMT-based quantitative proteomic approach to identify PAR4-regulated changes of proteomic profiles in ESCC cells and to identify potentially therapeutic value. A total of 33 proteins were found significantly changed with 15 up-regulated and 18 down-regulated in PAR4-activating peptide (PAR4-AP) treated ESCC cells compared with controls. Bioinformatics analysis showed that key higher expressed proteins included those associated with apoptosis and tumor suppressor (e.g. CASP9), and lower expressed proteins included those associated with anti-apoptosis, autophagy and promoting cell proliferation (e.g. CHMP1B, PURA, PARG and HIST1H2AH). Western blot verified changes in five representative proteins including CASP9, CHMP1B, PURA, PARG and HIST1H2AH. Immunohistochemistry analysis showed that CHMP1B, PURA, PARG and HIST1H2AH expression in ESCC tissues were significantly higher than those in adjacent nontumorous tissues. Our findings will be helpful in further investigations into the functions and molecular mechanisms of PAR4 in ESCC.
Collapse
Affiliation(s)
- Ming Wang
- Department of Human Anatomy, Taishan Medical University, 2 Ying Sheng Dong Lu, Tai'an, 271000, China
| | - Shuhong An
- Department of Human Anatomy, Taishan Medical University, 2 Ying Sheng Dong Lu, Tai'an, 271000, China
| | - Diyi Wang
- Department of Pathology, Affiliated Hospital of Taishan Medical University, Tai'an, 271000, China
| | - Haizhen Ji
- Department of Physiology, Taishan Medical University, Tai'an, China
| | - Min Geng
- Department of Human Anatomy, Taishan Medical University, 2 Ying Sheng Dong Lu, Tai'an, 271000, China
| | - Xingjing Guo
- Department of Physiology, Taishan Medical University, Tai'an, China
| | - Zhaojin Wang
- Department of Human Anatomy, Taishan Medical University, 2 Ying Sheng Dong Lu, Tai'an, 271000, China.
| |
Collapse
|
347
|
Zhang B, Zhang C, Yang X, Chen Y, Zhang H, Liu J, Wu Q. Cytoplasmic collagen XIαI as a prognostic biomarker in esophageal squamous cell carcinoma. Cancer Biol Ther 2018; 19:364-372. [PMID: 29333916 DOI: 10.1080/15384047.2018.1423915] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Stromal/cytoplasmic collagen XIαI (COL11A1) has been highlighted in the process of neoplastic transformation, including epithelial mesenchymal transition (EMT), metastasis and invasiveness. In this study, we aim to illuminate the clinical significance and biological role of COL11A1 in esophageal squamous cell carcinoma (ESCC). Herein, we investigated COL11A1 expression in 16 pairs of ESCC and adjacent normal tissues by RT-PCR and western blotting analysis. Correlations of COL11A1 expression with clinicopathologic parameters and survival status were then determined by immunohistochemistry in 116 ESCC and 50 normal specimens. Furthermore, bioinformatics was used for mechanisms exploration. And in vitro knockdown experiments were also performed. We found that COL11A1 expression was significantly higher in ESCC than in paired normal tissues at both mRNA and protein level. Immunohistochemistry showed that COL11A1 was predominantly localized to the cytoplasm rather than tumor stroma, patients with high COL11A1 expression had a poorer overall survival (OS) rate than those with low COL11A1 expression. Besides, increased COL11A1 expression was dramatically correlated with advanced clinical stage, invasion depth and lymph node metastases and served as an independent prognostic marker for ESCC. Likewise, COL11A1 dependent nomogram predicted a more precise survival outcome than traditional staging system. Moreover, COL11A1 silencing resulted in impaired cell proliferation and EMT, and subdued EMT inhibited cells aggressiveness. These biological processes (BPs) might be modulated by COL11A1 via the intracellular AKT/ERK/c-Myc cascades.
Collapse
Affiliation(s)
- Bohan Zhang
- a Department of Cardiothoracic Surgery , The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China.,b Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China
| | - Cheng Zhang
- a Department of Cardiothoracic Surgery , The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China
| | - Xuetao Yang
- a Department of Cardiothoracic Surgery , The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China.,b Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China
| | - Yue Chen
- a Department of Cardiothoracic Surgery , The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China.,b Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China
| | - Hongqi Zhang
- a Department of Cardiothoracic Surgery , The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China
| | - Jingshu Liu
- b Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China
| | - Qingchen Wu
- a Department of Cardiothoracic Surgery , The First Affiliated Hospital of Chongqing Medical University , Chongqing , P. R. China
| |
Collapse
|
348
|
Xiong Z, Ren S, Chen H, Liu Y, Huang C, Zhang YL, Odera JO, Chen T, Kist R, Peters H, Garman K, Sun Z, Chen X. PAX9 regulates squamous cell differentiation and carcinogenesis in the oro-oesophageal epithelium. J Pathol 2018; 244:164-175. [PMID: 29055049 PMCID: PMC5842438 DOI: 10.1002/path.4998] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/05/2017] [Accepted: 10/10/2017] [Indexed: 12/28/2022]
Abstract
PAX9 is a transcription factor of the PAX family characterized by a DNA-binding paired domain. Previous studies have suggested a potential role of PAX9 in squamous cell differentiation and carcinogenesis of the oro-oesophageal epithelium. However, its functional roles in differentiation and carcinogenesis remain unclear. In this study, Pax9 deficiency in mouse oesophagus promoted cell proliferation, delayed cell differentiation, and altered the global gene expression profile. Ethanol exposure downregulated PAX9 expression in human oesophageal epithelial cells in vitro and mouse forestomach and tongue in vivo. We further showed that PAX9 was downregulated in human oro-oesophageal squamous cell carcinoma (OESCC), and its downregulation was associated with alcohol drinking and promoter hypermethylation. Moreover, ad libitum feeding with a liquid diet containing ethanol for 40 weeks or Pax9 deficiency promoted N-nitrosomethylbenzylamine-induced squamous cell carcinogenesis in mouse tongue, oesophagus, and forestomach. In conclusion, PAX9 regulates squamous cell differentiation in the oro-oesophageal epithelium. Alcohol drinking and promoter hypermethylation are associated with PAX9 silencing in human OESCC. PAX9 downregulation may contribute to alcohol-associated oro-oesophageal squamous cell carcinogenesis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Zhaohui Xiong
- Department of Oral Medicine, Beijing Hospital for Stomatology, Capital Medical University, 4 Tian-Tan-Xi-Li, Beijing 100050, China
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Shuang Ren
- Department of Oral Medicine, Beijing Hospital for Stomatology, Capital Medical University, 4 Tian-Tan-Xi-Li, Beijing 100050, China
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Hao Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Yao Liu
- Department of Oral Medicine, Beijing Hospital for Stomatology, Capital Medical University, 4 Tian-Tan-Xi-Li, Beijing 100050, China
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Caizhi Huang
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Yawan Lyvia Zhang
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Joab Otieno Odera
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
| | - Tong Chen
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, 410 West 12 Avenue, Columbus, OH 43210, USA
| | - Ralf Kist
- Centre for Oral Health Research, School of Dental Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4BW, UK
- Institute of Human Genetics, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Heiko Peters
- Institute of Human Genetics, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Katherine Garman
- Division of Gastroenterology, Department of Medicine, Duke University, DUMC 3913, Durham, NC 27710, USA
| | - Zheng Sun
- Department of Oral Medicine, Beijing Hospital for Stomatology, Capital Medical University, 4 Tian-Tan-Xi-Li, Beijing 100050, China
| | - Xiaoxin Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, 700 George Street, Durham, NC 27707, USA
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
349
|
Hisano O, Nonoshita T, Hirata H, Sasaki T, Watanabe H, Wakiyama H, Ono M, Ohga S, Honda H. Additional radiotherapy following endoscopic submucosal dissection for T1a-MM/T1b-SM esophageal squamous cell carcinoma improves locoregional control. Radiat Oncol 2018; 13:14. [PMID: 29378603 PMCID: PMC5789550 DOI: 10.1186/s13014-018-0960-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 01/15/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Endoscopic submucosal dissection (ESD) can be used as a less invasive treatment option for superficial esophageal cancer involving the muscularis mucosae (T1a-MM) or upper third of the submucosa (T1b-SM1). Additional treatment after ESD is needed to prevent lymph node metastasis. However, the efficacy of radiotherapy following ESD has not been well evaluated. Moreover, the clinical outcomes of patients with large mucosal defects of the esophagus who received radiotherapy after ESD have not been reported. This study aimed to clarify the efficacy of additional radiotherapy following ESD for esophageal squamous cell cancer involving T1a-MM or T1b-SM1. METHODS We analyzed twenty-seven patients with pathologically confirmed T1a-MM or T1b-SM1 esophageal squamous cell cancer treated by ESD. Thirteen patients received additional radiotherapy (RT group), and the remaining patients did not (non-RT group). Locoregional control (LRC), overall survival, cause-specific survival, and adverse events including treatment-related esophageal strictures were evaluated. RESULTS The three-year LRC was significantly better for the RT than the non-RT group (100% vs. 57.8%, respectively; p = 0.022). Chemotherapy following ESD did not improve LRC. Multivariate analysis showed that radiotherapy was an independent prognostic factor for better LRC (p = 0.0022). Contrary to the results in LRC, overall and cause-specific survival were not significantly different between the RT and non-RT groups. A subgroup analysis of patients with mucosal defects involving ≥ 3/4 of the esophageal circumference after ESD showed that LRC of the RT group was better than that of the non-RT group (p = 0.049). Treatment-related esophageal strictures were observed in 2 of 6 patients in the RT group with large mucosal defects after ESD. No patients with mucosal defects involving less than 3/4 of the circumference after ESD developed treatment-related strictures. CONCLUSIONS Radiotherapy after ESD contributed to better LRC in esophageal squamous cell cancer involving pT1a-MM and pT1b-SM1. Esophageal strictures were observed in some patients with large mucosal defects after ESD. Despite leading to better LRC, radiotherapy after ESD should be undertaken after careful consideration for patients with large mucosal defects after ESD.
Collapse
Affiliation(s)
- Osamu Hisano
- Department of Clinical Radiology, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Radiology, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan
| | - Takeshi Nonoshita
- Department of Radiology, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan
| | - Hidenari Hirata
- Department of Clinical Radiology, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Tomonari Sasaki
- Department of Clinical Radiology, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hideyuki Watanabe
- Department of Radiology, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan
| | - Hiroaki Wakiyama
- Department of Clinical Radiology, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Minoru Ono
- Department of Radiology, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan
| | - Saiji Ohga
- Department of Clinical Radiology, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroshi Honda
- Department of Clinical Radiology, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
350
|
Huang J, Xu B, Mo H, Zhang W, Chen X, Wu D, Qu D, Wang X, Lan B, Yang B, Wang P, Zhang H, Yang Q, Jiao Y. Safety, Activity, and Biomarkers of SHR-1210, an Anti-PD-1 Antibody, for Patients with Advanced Esophageal Carcinoma. Clin Cancer Res 2018; 24:1296-1304. [PMID: 29358502 DOI: 10.1158/1078-0432.ccr-17-2439] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/30/2017] [Accepted: 01/03/2018] [Indexed: 12/14/2022]
Abstract
Purpose: The current management of advanced esophageal squamous cell carcinoma (ESCC) remains unsatisfactory. We investigated the safety, efficacy, and biomarkers of SHR-1210, an anti-PD-1 antibody, in patients with recurrent or metastatic ESCC.Experimental Design: This study was part of a phase I trial in China. Patients with advanced ESCC who were refractory or intolerant to previous chemotherapy were enrolled. Eligible patients received intravenous SHR-1210 at a dose of 60 mg, with escalation to 200 and 400 mg (4-week interval after first dose followed by a 2-week schedule) until disease progression or intolerable toxicity. The associations between candidate biomarkers (PD-L1 and somatic mutation load) and the efficacy of SHR-1210 were also explored.Results: Between May 11, 2016, and December 9, 2016, a total of 30 patients from one site in China were enrolled. Ten patients (33.3%) had an independently assessed objective response. Median progression-free survival was 3.6 months (95% CI, 0-7.2). Three (10.0%) treatment-related grade 3 adverse events were reported: two (6.7%) pneumonitis and one (3.3%) increased cardiac troponin I. No grade 4 or grade 5 treatment-related adverse events were reported. The exome sequencing and analysis showed that the mutational burden and the potential mutation-associated neoantigen count were associated with better responses. An objective response was more common in patients with PD-L1-positive tumors as defined by ≥5% staining (7 of 15 patients) than in those with PD-L1-negative tumors (1 of 9 patients).Conclusions: In this population of ESCC patients, SHR-1210 had a manageable safety profile and promising antitumor activity. Clin Cancer Res; 24(6); 1296-304. ©2018 AACR.
Collapse
Affiliation(s)
- Jing Huang
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hongnan Mo
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weilong Zhang
- Laboratory of Cell and Molecular Biology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuelian Chen
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dawei Wu
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Qu
- Department of Diagnostic Radiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingyuan Wang
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Lan
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Beibei Yang
- Laboratory of Cell and Molecular Biology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Wang
- Laboratory of Cell and Molecular Biology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongtu Zhang
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Yang
- Jiangsu Hengrui Medicine Co., Ltd. Shanghai, China
| | - Yuchen Jiao
- Laboratory of Cell and Molecular Biology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|