1
|
Cheung BB, Mittra R, Murray J, Wang Q, Seneviratne JA, Raipuria M, Wong IPL, Restuccia D, Gifford A, Salib A, Sutton S, Huang L, Ferdowsi PV, Tsang J, Sekyere E, Mayoh C, Luo L, Brown DL, Stow JL, Zhu S, Young RJ, Solomon BJ, Chappaz S, Kile B, Kueh A, Herold MJ, Hilton DJ, Liu T, Norris MD, Haber M, Carter DR, Parker MW, Marshall GM. Golgi-localized Ring Finger Protein 121 is necessary for MYCN-driven neuroblastoma tumorigenesis. Commun Biol 2024; 7:1322. [PMID: 39402275 PMCID: PMC11473750 DOI: 10.1038/s42003-024-06899-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
MYCN amplification predicts poor prognosis in childhood neuroblastoma. To identify MYCN oncogenic signal dependencies we performed N-ethyl-N-nitrosourea (ENU) mutagenesis on the germline of neuroblastoma-prone TH-MYCN transgenic mice to generate founders which had lost tumorigenesis. Sequencing of the mutant mouse genomes identified the Ring Finger Protein 121 (RNF121WT) gene mutated to RNFM158R associated with heritable loss of tumorigenicity. While the RNF121WT protein localised predominantly to the cis-Golgi Complex, the RNF121M158R mutation in Helix 4 of its transmembrane domain caused reduced RNF121 protein stability and absent Golgi localisation. RNF121WT expression markedly increased during TH-MYCN tumorigenesis, whereas hemizygous RNF121WT gene deletion reduced TH-MYCN tumorigenicity. The RNF121WT-enhanced growth of MYCN-amplified neuroblastoma cells depended on RNF121WT transmembrane Helix 5. RNF121WT directly bound MYCN protein and enhanced its stability. High RNF121 mRNA expression associated with poor prognosis in human neuroblastoma tissues and another MYC-driven malignancy, laryngeal cancer. RNF121 is thus an essential oncogenic cofactor for MYCN and a target for drug development.
Collapse
Affiliation(s)
- Belamy B Cheung
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia.
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia.
| | - Ritu Mittra
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Jayne Murray
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Qian Wang
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Janith A Seneviratne
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Mukesh Raipuria
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Iris Poh Ling Wong
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - David Restuccia
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Andrew Gifford
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Alice Salib
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Selina Sutton
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Libby Huang
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Parisa Vahidi Ferdowsi
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Joanna Tsang
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Eric Sekyere
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Lin Luo
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Darren L Brown
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Jennifer L Stow
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Cancer Center and Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Stephane Chappaz
- Anatomy & Developmental Biology, Monash University, Melbourne, Australia
| | - Benjamin Kile
- Faculty of Health and Medical Sciences at the University of Adelaide, Adelaide, Australia
| | - Andrew Kueh
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, 3052, Australia
| | - Marco J Herold
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, 3052, Australia
| | - Douglas J Hilton
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, 3052, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
- University of New South Wales Centre for Childhood Cancer Research, Sydney, NSW 2052, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
- University of New South Wales Centre for Childhood Cancer Research, Sydney, NSW 2052, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Daniel R Carter
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | - Michael W Parker
- ACRF Facility for Innovative Cancer Drug Discovery and Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Glenn M Marshall
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, Australia.
- Kids Cancer Centre, Sydney Children's Hospital, Sydney, 2031, NSW, Australia.
| |
Collapse
|
2
|
Zhang S, Gu J, Wang W, Sun L, Jiang T, Yang X, Yin J, Lin M, Lin D, Wang H, Tan L. Suppression of SENP3 enhances macrophage alternative activation by mediating IRF4 de-SUMOylation in ESCC progression. Cell Commun Signal 2024; 22:395. [PMID: 39123188 PMCID: PMC11312810 DOI: 10.1186/s12964-024-01770-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Esophageal cancer is common worldwide, with ESCC being the most frequent tumor in East Asia. Tumor-associated macrophages are an important component of the ESCC microenvironment. SUMOylation is a post-translational modification of proteins, and SUMO-specific proteases (SENPs) play an important role in de-SUMOylation. In human patients, we discovered that the levels of SENP3 were upregulated in the tumor-associated macrophages. Furthermore, the loss of SENP3 enhanced the alternative activation of macrophages in the 4-NQO-induced ESCC mice model. This is the first study to identify SENP3-mediated macrophage polarization via the de-SUMOylation of interferon regulatory factor 4 (IRF4) at the K349 site. Alternative activation of macrophages increases the migration and invasion potential of ESCC cells and promotes their progression in vivo. Moreover, patients with relatively low SENP3 expression in macrophages exhibit higher primary PET SUVmax value and lymph node metastasis rates. In summary, this study revealed that SENP3-mediated IRF4 de-SUMOylation is crucial for the alternative activation of macrophages and influences the progression of ESCC.
Collapse
Affiliation(s)
- Shaoyuan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jianmin Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wenhan Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Linyi Sun
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Tian Jiang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xinyu Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jun Yin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Miao Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Dong Lin
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Hongkou District, No. 100, Haining Road, Shanghai, 200080, People's Republic of China.
| | - Hao Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Xuhui District, No. 180, Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
3
|
Fan Z, Zhou J, Tian Y, Qin Y, Liu Z, Gu L, Dawsey SM, Wei W, Deng D. Somatic CDKN2A copy number variations are associated with the prognosis of esophageal squamous cell dysplasia. Chin Med J (Engl) 2024; 137:980-989. [PMID: 38445358 PMCID: PMC11046026 DOI: 10.1097/cm9.0000000000002982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Somatic copy number variations (SCNVs) in the CDKN2A gene are among the most frequent events in the dysplasia-carcinoma sequence of esophageal squamous cell carcinoma. However, whether CDKN2A SCNVs are useful biomarkers for the risk stratification and management of patients with esophageal squamous cell dysplasia (ESCdys) is unknown. This study aimed to investigate the characteristics and prognostic value of CDKN2A SCNVs in patients with mild or moderate (m/M) ESCdys. METHODS This study conducted a prospective multicenter study of 205 patients with a baseline diagnosis of m/M ESCdys in five high-risk regions of China (Ci County, Hebei Province; Yanting, Sichuan Province; Linzhou, Henan Province; Yangzhong, Jiangsu Province; and Feicheng, Shandong Province) from 2005 to 2019. Genomic DNA was extracted from paraffin biopsy samples and paired peripheral white blood cells from patients, and a quantitative polymerase chain reaction assay, P16-Light, was used to detect CDKN2A copy number. The cumulative regression and progression rates of ESCdys were evaluated using competing risk models. RESULTS A total of 205 patients with baseline m/M ESCdys were enrolled. The proportion of ESCdys regression was significantly lower in the CDKN2A deletion cohort than in the diploid and amplification cohorts (18.8% [13/69] vs. 35.0% [28/80] vs. 51.8% [29/56], P <0.001). In the univariable competing risk analysis, the cumulative regression rate was statistically significantly lower ( P = 0.008), while the cumulative progression rate was higher ( P = 0.017) in ESCdys patients with CDKN2A deletion than in those without CDKN2A deletion. CDKN2A deletion was also an independent predictor of prognosis in ESCdys ( P = 0.004) in the multivariable analysis. CONCLUSION The results indicated that CDKN2A SCNVs are associated with the prognosis of ESCdys and may serve as potential biomarkers for risk stratification.
Collapse
Affiliation(s)
- Zhiyuan Fan
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jing Zhou
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Yuan Tian
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Yu Qin
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhaojun Liu
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Liankun Gu
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Sanford M. Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wenqiang Wei
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
4
|
Cheng M, Xin Q, Ma S, Ge M, Wang F, Yan X, Jiang B. Advances in the Theranostics of Oesophageal Squamous Carcinoma. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202200251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Indexed: 01/04/2025]
Abstract
AbstractOesophageal squamous carcinoma (ESCC) is one of the most lethal human malignancies, and it is a more aggressive form of oesophageal cancer (EC) that comprises over 90% of all EC cases in China compared with oesophageal adenocarcinoma (EAC). The high mortality of ESCC is attributed to the late‐stage diagnosis, chemoradiotherapy resistance, and lack of appropriate therapeutic targets and corresponding therapeutic formulations. Recently, emerging clinical and translational investigations have involved genome analyses, diagnostic biomarkers, and targeted therapy for ESCC, and these studies provide a new horizon for improving the clinical outcomes of patients with ESCC. Here, the latest research advances in the theranostics of ESCC are reviewed and the unique features of ESCC (including differences from EAC, genomic alterations, and microbe infections), tissue and circulating biomarkers, chemoradiotherapy resistance, clinical targeted therapy for ESCC, identification of novel therapeutic targets, and designation of nanotherapeutic systems for ESCC are particularly focused on. Finally, the perspectives for future clinical and translational theranostic research of ESCC are discussed and the obstacles that must be overcome in ESCC theranostics are described.
Collapse
Affiliation(s)
- Miaomiao Cheng
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Qi Xin
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Saiyu Ma
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Mengyue Ge
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Feng Wang
- Oncology Department The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450000 China
| | - Xiyun Yan
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Zhengzhou Henan 450001 China
- CAS Engineering Laboratory for Nanozyme Key Laboratory of Protein and Peptide Pharmaceuticals Institute of Biophysics Chinese Academy of Sciences Beijing 100101 China
| | - Bing Jiang
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Zhengzhou Henan 450001 China
| |
Collapse
|
5
|
Ke S, Wang J, Lu J, Fang M, Li R. Long intergenic non-protein coding RNA 00858 participates in the occurrence and development of esophageal squamous cell carcinoma through the activation of the FTO-m6A-MYC axis by recruiting ZNF184. Genomics 2023; 115:110593. [PMID: 36868327 DOI: 10.1016/j.ygeno.2023.110593] [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: 11/20/2022] [Revised: 02/10/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023]
Abstract
OBJECTIVES We aimed at probing impact of LINC00858 on esophageal squamous cell carcinoma (ESCC) progression via ZNF184-FTO-m6A-MYC axis. METHODS Expression of related genes (LINC00858, ZNF184, FTO, and MYC) was detected in ESCC tissues or cells and their relationships were assessed. After expression alterations in ESCC cells, cell proliferation, invasion, migration, and apoptosis were detected. Tumor formation in nude mice was conducted. RESULTS LINC00858, ZNF184, FTO, and MYC were overexpressed in ESCC tissues and cells. LINC00858 enhanced ZNF184 expression to upregulate FTO, which augmented MYC expression. LINC00858 knockdown diminished ESCC cell proliferative, migratory, and invasive properties while elevating apoptosis, which was negated by FTO overexpression. FTO knockdown exerted similar functions of LINC00858 knockdown on ESCC cell movements, which was annulled by MYC upregulation. Silencing LINC00858 repressed tumor growth and related gene expression in nude mice. CONCLUSIONS LINC00858 modulated MYC m6A modification via FTO by recruiting ZNF184, thus facilitating ESCC progression.
Collapse
Affiliation(s)
- Shun Ke
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Jing Wang
- Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430071, PR China
| | - Jun Lu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Minghao Fang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Ruichao Li
- Department of General Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
6
|
Liu K, Lu H, Jiang D, Guan Y, Xu H, Sun Q, Jiang Q, Zheng J, Chen H, Zhang F, Luo R, Huang Y, Xu J, Hou Y. Prognostic Significance of CDK6 Amplification in Esophageal Squamous Cell Carcinoma. Cancer Treat Res Commun 2023; 35:100698. [PMID: 37023643 DOI: 10.1016/j.ctarc.2023.100698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Dysregulation of CDK6 plays crucial roles in the carcinogenesis of many kinds of human malignancies. However, the role of CDK6 in esophageal squamous cell carcinoma (ESCC) is not well known. We investigated the frequency and prognostic value of CDK6 amplification to improve the risk stratification in patients with ESCC. Pan-cancer analysis of CDK6 was conducted on The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx) and Gene Expression Omnibus (GEO) databases. CDK6 amplification was detected in 502 ESCC samples by Fluorescence in situ hybridization (FISH) through tissue microarrays (TMA). Pan-cancer analysis revealed that CDK6 mRNA level was much higher in multiple kinds of cancers and higher CDK6 mRNA level indicated a better prognosis in ESCC. In this study, CDK6 amplification was detected in 27.5% (138/502) of patients with ESCC. CDK6 amplification was significantly correlated with tumor size (p = 0.044). Patients with CDK6 amplification tended to have a longer disease-free survival (DFS) (p = 0.228) and overall survival (OS) (p = 0.200) compared with patients without CDK6 amplification but of no significance. When further divided into I-II and III-IV stage, CDK6 amplification was significantly associated with longer DFS and OS in III-IV stage group (DFS, p = 0.036; OS, p = 0.022) rather than in I-II stage group (DFS, p = 0.776; OS, p = 0.611). On univariate and multivariate analysis of Cox hazard model, differentiation, vessel invasion, nerve invasion, invasive depth, lymph node metastasis and clinical stage were significantly associated with DFS and OS. Moreover, invasion depth was an independent factor for ESCC prognosis. Taken together, for ESCC patients in III-IV stage, CDK6 amplification indicated a better prognosis.
Collapse
Affiliation(s)
- Kun Liu
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Huadong Lu
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingying Guan
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Huijuan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Qi Sun
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Qiuli Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Jingmei Zheng
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Huan Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Fuhan Zhang
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Ruichen Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Ying Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China
| | - Jianfang Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China; Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Xiamen Branch, Xiamen City, Fujian Province, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen City, Fujian Province, China; Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Testa U, Castelli G, Pelosi E. The Molecular Characterization of Genetic Abnormalities in Esophageal Squamous Cell Carcinoma May Foster the Development of Targeted Therapies. Curr Oncol 2023; 30:610-640. [PMID: 36661697 PMCID: PMC9858483 DOI: 10.3390/curroncol30010048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Esophageal cancer is among the most common tumors in the world and is associated with poor outcomes, with a 5-year survival rate of about 10-20%. Two main histological subtypes are observed: esophageal squamous cell carcinoma (ESCC), more frequent among Asian populations, and esophageal adenocarcinoma (EAC), the predominant type in Western populations. The development of molecular analysis techniques has led to the definition of the molecular alterations observed in ESCC, consistently differing from those observed in EAC. The genetic alterations observed are complex and heterogeneous and involve gene mutations, gene deletions and gene amplifications. However, despite the consistent progress in the definition of the molecular basis of ESCC, precision oncology for these patients is still virtually absent. The recent identification of molecular subtypes of ESCC with clinical relevance may foster the development of new therapeutic strategies. It is estimated that about 40% of the genetic alterations observed in ESCC are actionable. Furthermore, the recent introduction of solid tumor immunotherapy with immune checkpoint inhibitors (ICIs) showed that a minority of ESCC patients are responsive, and the administration of ICIs, in combination with standard chemotherapy, significantly improves overall survival over chemotherapy in ESCC patients with advanced disease.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | | | | |
Collapse
|
8
|
Naa10p promotes cell invasiveness of esophageal cancer by coordinating the c-Myc and PAI1 regulatory axis. Cell Death Dis 2022; 13:995. [PMID: 36433943 PMCID: PMC9700753 DOI: 10.1038/s41419-022-05441-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022]
Abstract
N-α-acetyltransferase 10 protein, Naa10p, is involved in various cellular functions impacting tumor progression. Due to its capacity to acetylate a large spectrum of proteins, both oncogenic and tumor-suppressive roles of Naa10p have been documented. Here, we report an oncogenic role of Naa10p in promoting metastasis of esophageal cancer. NAA10 is more highly expressed in esophageal cancer tissues compared to normal tissues. Higher NAA10 expression also correlates with poorer survival of esophageal cancer patients. We found that NAA10 expression was transcriptionally regulated by the critical oncogene c-Myc in esophageal cancer. Furthermore, activation of the c-Myc-Naa10p axis resulted in upregulated cell invasiveness of esophageal cancer. This increased cell invasiveness was also elucidated to depend on the enzymatic activity of Naa10p. Moreover, Naa10p cooperated with Naa15p to interact with the protease inhibitor, PAI1, and prevent its secretion. This inhibition of PAI1 secretion may derive from the N-terminal acetylation effect of the Naa10p/Naa15p complex. Our results establish the significance of Naa10p in driving metastasis in esophageal cancer by coordinating the c-Myc-PAI1 axis, with implications for its potential use as a prognostic biomarker and therapeutic target for esophageal cancer.
Collapse
|
9
|
Savva L, Fossépré M, Keramidas O, Themistokleous A, Rizeq N, Panagiotou N, Leclercq M, Nicolaidou E, Surin M, Hayes SC, Georgiades SN. Gaining Insights on the Interactions of a Class of Decorated (2-([2,2'-Bipyridin]-6-yl)phenyl)platinum Compounds with c-Myc Oncogene Promoter G-Quadruplex and Other DNA Structures. Chemistry 2022; 28:e202201497. [PMID: 35726630 PMCID: PMC9804160 DOI: 10.1002/chem.202201497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Indexed: 01/05/2023]
Abstract
Organometallic molecules offer some of the most promising scaffolds for interaction with G-quadruplex nucleic acids. We report the efficient synthesis of a family of organoplatinum(II) complexes, featuring a 2-([2,2'-bipyridin]-6-yl)phenyl tridentate (N∧ N∧ C) ligand, that incorporates peripheral side-chains aiming at enhancing and diversifying its interaction capabilities. These include a di-isopropyl carbamoyl amide, a morpholine ethylenamide, two enantiomeric proline imides and an oxazole. The binding affinities of the Pt-complexes were evaluated via UV-vis and fluorescence titrations, against 5 topologically-distinct DNA structures, including c-myc G-quadruplex, two telomeric (22AG) G-quadruplexes, a duplex (ds26) and a single-stranded (polyT) DNA. All compounds exhibited binding selectivity in favour of c-myc, with association constants (Ka ) in the range of 2-5×105 M-1 , lower affinity for both folds of 22AG and for ds26 and negligible affinity for polyT. Remarkable emission enhancements (up to 200-fold) upon addition of excess DNA were demonstrated by a subset of the compounds with c-myc, providing a basis for optical selectivity, since optical response to all other tested DNAs was low. A c-myc DNA-melting experiment showed significant stabilizing abilities for all compounds, with the most potent binder, the morpholine-Pt-complex, exhibiting a ΔTm >30 °C, at 1 : 5 DNA-to-ligand molar ratio. The same study implied contributions of the diverse side-chains to helix stabilization. To gain direct evidence of the nature of the interactions, mixtures of c-myc with the four most promising compounds were studied via UV Resonance Raman (UVRR) spectroscopy, which revealed end-stacking binding mode, combined with interactions of side-chains with loop nucleobase residues. Docking simulations were conducted to provide insights into the binding modes for the same four Pt-compounds, suggesting that the binding preference for two alternative orientations of the c-myc G-quadruplex thymine 'cap' ('open' vs. 'closed'), as well as the relative contributions to affinity from end-stacking and H-bonding, are highly dependent on the nature of the interacting Pt-complex side-chain.
Collapse
Affiliation(s)
- Loukiani Savva
- Department of ChemistryUniversity of Cyprus1 Panepistimiou Avenue, Aglandjia2109NicosiaCyprus
| | - Mathieu Fossépré
- Laboratory for Chemistry of Novel MaterialsUniversity of Mons – UMONS20 Place du ParcB-7000MonsBelgium
| | - Odysseas Keramidas
- Department of ChemistryUniversity of Cyprus1 Panepistimiou Avenue, Aglandjia2109NicosiaCyprus
| | | | - Natalia Rizeq
- Department of ChemistryUniversity of Cyprus1 Panepistimiou Avenue, Aglandjia2109NicosiaCyprus
| | - Nikos Panagiotou
- Department of ChemistryUniversity of Cyprus1 Panepistimiou Avenue, Aglandjia2109NicosiaCyprus
| | - Maxime Leclercq
- Laboratory for Chemistry of Novel MaterialsUniversity of Mons – UMONS20 Place du ParcB-7000MonsBelgium
| | - Eliana Nicolaidou
- Department of ChemistryUniversity of Cyprus1 Panepistimiou Avenue, Aglandjia2109NicosiaCyprus
| | - Mathieu Surin
- Laboratory for Chemistry of Novel MaterialsUniversity of Mons – UMONS20 Place du ParcB-7000MonsBelgium
| | - Sophia C. Hayes
- Department of ChemistryUniversity of Cyprus1 Panepistimiou Avenue, Aglandjia2109NicosiaCyprus
| | - Savvas N. Georgiades
- Department of ChemistryUniversity of Cyprus1 Panepistimiou Avenue, Aglandjia2109NicosiaCyprus
| |
Collapse
|
10
|
Liu T, Wang X, Guo W, Shao F, Li Z, Zhou Y, Zhao Z, Xue L, Feng X, Li Y, Tan F, Zhang K, Xue Q, Gao S, Gao Y, He J. RNA Sequencing of Tumor-Educated Platelets Reveals a Three-Gene Diagnostic Signature in Esophageal Squamous Cell Carcinoma. Front Oncol 2022; 12:824354. [PMID: 35615147 PMCID: PMC9124963 DOI: 10.3389/fonc.2022.824354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/29/2022] [Indexed: 12/24/2022] Open
Abstract
There is no cost-effective, accurate, and non-invasive method for the detection of esophageal squamous cell carcinoma (ESCC) in clinical practice. We aimed to investigate the diagnostic potential of tumor-educated platelets in ESCC. In this study, seventy-one ESCC patients and eighty healthy individuals were enrolled and divided into a training cohort (23 patients and 27 healthy individuals) and a validation cohort (48 patients and 53 healthy individuals). Next-generation RNA sequencing was performed on platelets isolated from peripheral blood of all participants, and a support vector machine/leave-one-out cross validation (SVM/LOOCV) approach was used for binary classification. A diagnostic signature composed of ARID1A, GTF2H2, and PRKRIR discriminated ESCC patients from healthy individuals with 91.3% sensitivity and 85.2% specificity in the training cohort and 87.5% sensitivity and 81.1% specificity in the validation cohort. The AUC was 0.924 (95% CI, 0.845–0.956) and 0.893 (95% CI, 0.821–0.966), respectively, in the training cohort and validation cohort. This 3-gene platelet RNA signature could effectively discriminate ESCC from healthy control. Our data highlighted the potential of tumor-educated platelets for the noninvasive diagnosis of ESCC. Moreover, we found that keratin and collagen protein families and ECM-related pathways might be involved in tumor progression and metastasis of ESCC, which might provide insights to understand ESCC pathobiology and advance novel therapeutics.
Collapse
Affiliation(s)
- Tiejun Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Shao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Cancer Institute of the Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, Qingdao, China
| | - Zitong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihong Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Zhang
- Department of Medical Examination for Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- 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, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- 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, China
| | - Yibo Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- 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, China
- *Correspondence: Yibo Gao, ; Jie He,
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- 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, China
- *Correspondence: Yibo Gao, ; Jie He,
| |
Collapse
|
11
|
Hirata H, Niida A, Kakiuchi N, Uchi R, Sugimachi K, Masuda T, Saito T, Kageyama SI, Motomura Y, Ito S, Yoshitake T, Tsurumaru D, Nishimuta Y, Yokoyama A, Hasegawa T, Chiba K, Shiraishi Y, Du J, Miura F, Morita M, Toh Y, Hirakawa M, Shioyama Y, Ito T, Akimoto T, Miyano S, Shibata T, Mori M, Suzuki Y, Ogawa S, Ishigami K, Mimori K. The Evolving Genomic Landscape of Esophageal Squamous Cell Carcinoma Under Chemoradiotherapy. Cancer Res 2021; 81:4926-4938. [PMID: 34413060 DOI: 10.1158/0008-5472.can-21-0653] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/22/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) often recurs after chemoradiotherapy, and the prognosis of ESCC after chemoradiotherapy has not improved over the past few decades. The mutation process in chemoradiotherapy-resistant clones and the functional relevance of genetic alterations remain unclear. To address these problems, we performed whole-exome sequencing of 52 tumor samples from 33 patients with ESCC who received radiotherapy combined with 5-fluorouracil/platinum. In multiregion analyses of pretreatment and locally recurrent lesions from five cases, most driver gene-altered clones remained under chemoradiotherapy selection pressure, while few driver gene alterations were acquired at recurrence. The mutation signatures of recurrent ESCC, including increased deletion frequency and platinum dose-dependent base substitution signatures, were substantially different from those of primary ESCC and reflected the iatrogenic impacts of chemoradiotherapy. Single-region analysis of 28 pretreatment tumors indicated that focal copy-number gain at the MYC locus was significantly associated with poor progression-free survival and overall survival after chemoradiotherapy. MYC gain remained throughout the chemoradiotherapy course and potentially contributes to intrinsic resistance to chemoradiotherapy. Consistent with these findings, MYC copy number and mRNA and protein levels in ESCC cell lines correlated positively with resistance to radiotherapy, and MYC knockdown improved sensitivity to radiotherapy. Overall, these data characterize the clonal evolution process induced by chemoradiotherapy and clinically relevant associations for genetic alterations in ESCC. These findings increase our understanding of therapeutic resistance and support the rationale for precision chemoradiotherapy. SIGNIFICANCE: Whole-exome sequencing reveals the genetic evolution of ESCC during chemoradiotherapy, highlighting MYC gain in pretreatment tumors as a potential marker of therapy resistance.
Collapse
Affiliation(s)
- Hidenari Hirata
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.,Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan.,Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Atsushi Niida
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Division of Health Medical Computational Science, Health Intelligence Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Nobuyuki Kakiuchi
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryutaro Uchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Keishi Sugimachi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Tomoko Saito
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Shun-Ichiro Kageyama
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan.,Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Yushi Motomura
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.,Department of Radiology, Kyushu University Beppu Hospital, Beppu, Japan
| | - Shuhei Ito
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Tadamasa Yoshitake
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daisuke Tsurumaru
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Nishimuta
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Yokoyama
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takanori Hasegawa
- Division of Health Medical Computational Science, Health Intelligence Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kenichi Chiba
- Laboratory of DNA Information Analysis, Human Genome Centre, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Section of Genome Analysis Platform, Center for Cancer Genomic and Advanced Therapeutics, National Cancer Center, Tokyo, Japan
| | - Yuichi Shiraishi
- Laboratory of DNA Information Analysis, Human Genome Centre, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Section of Genome Analysis Platform, Center for Cancer Genomic and Advanced Therapeutics, National Cancer Center, Tokyo, Japan
| | - Junyan Du
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Fumihito Miura
- Department of Biochemistry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaru Morita
- Department of Gastroenterological Surgery, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Yasushi Toh
- Department of Gastroenterological Surgery, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Masakazu Hirakawa
- Department of Radiology, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yoshiyuki Shioyama
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Ion Beam Therapy Center, SAGA HIMAT Foundation, Tosu, Japan
| | - Takashi Ito
- Department of Biochemistry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuo Akimoto
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan.,Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Centre, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiro Shibata
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yutaka Suzuki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kousei Ishigami
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan.
| |
Collapse
|
12
|
Erkizan HV, Sukhadia S, Natarajan TG, Marino G, Notario V, Lichy JH, Wadleigh RG. Exome sequencing identifies novel somatic variants in African American esophageal squamous cell carcinoma. Sci Rep 2021; 11:14814. [PMID: 34285259 PMCID: PMC8292420 DOI: 10.1038/s41598-021-94064-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Esophageal cancer has a strikingly low survival rate mainly due to the lack of diagnostic markers for early detection and effective therapies. In the U.S., 75% of individuals diagnosed with esophageal squamous cell carcinoma (ESCC) are of African descent. African American ESCC (AA ESCC) is particularly aggressive, and its biological underpinnings remain poorly understood. We sought to identify the genomic abnormalities by conducting whole exome sequencing of 10 pairs of matched AA esophageal squamous cell tumor and control tissues. Genomic analysis revealed diverse somatic mutations, copy number alterations (SCNAs), and potential cancer driver genes. Exome variants created two subgroups carrying either a high or low tumor mutation burden. Somatic mutational analysis based on the Catalog of Somatic Mutations in Cancer (COSMIC) detected SBS16 as the prominent signature in the high mutation rate group suggesting increased DNA damage. SBS26 was also detected, suggesting possible defects in mismatch repair and microsatellite instability. We found SCNAs in multiple chromosome segments, encoding MYC on 8q24.21, PIK3CA and SOX2 on 3q26, CCND1, SHANK2, CTTN on 11q13.3, and KRAS on 12p12. Amplifications of EGFRvIII and EGFRvIVa mutants were observed in two patients, representing a novel finding in ESCC that has potential clinical relevance. This present exome sequencing, which to our knowledge, represents the first comprehensive exome analysis exclusively in AA ESCC, and highlights novel mutated loci that might explain the aggressive nature of AA ESCC and lead to the development of diagnostic and prognostic markers as well as therapeutic targets.
Collapse
Affiliation(s)
- Hayriye Verda Erkizan
- Institute for Clinical Research, Veterans Affairs Medical Center, Washington, DC, USA.
| | | | | | - Gustavo Marino
- Hepatology and Gastroenterology, Veterans Affairs Medical Center, Washington, DC, USA
| | - Vicente Notario
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Jack H Lichy
- Pathology and Laboratory Service, Veterans Affairs Medical Center, Washington, DC, USA
| | - Robert G Wadleigh
- Institute for Clinical Research, Veterans Affairs Medical Center, Washington, DC, USA.,Hematology and Medical Oncology, Veterans Affairs Medical Center, Washington, DC, USA
| |
Collapse
|
13
|
Yu Z, Wang H, Song Q, Huang J, Xu J, Su J, Wang H, Tan L, Wang X, Jiang Z, Chen W, Jiang D, Hou Y. Prognostic value and characterization of NTRK1 variation by fluorescence in situ hybridization in esophageal squamous cell carcinoma. J Cancer Res Clin Oncol 2021; 147:3113-3121. [PMID: 33963905 DOI: 10.1007/s00432-021-03578-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/20/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Rearrangement of the neurotrophic tyrosine kinase receptor (NTRK) 1 gene is a target of tropomyosin receptor kinase A (TRKA) inhibitors, and its targeted drug (larotrectinib) has been approved by the US Food and Drug Administration. We investigated the existence and prognostic importance of NTRK1 variation in esophageal squamous cell carcinoma (ESCC). METHODS Fluorescence in situ hybridization of a NTRK1 rearrangement was conducted on 523 ESCC samples through tissue microarrays. Kaplan-Meier curves with log-rank tests were used to evaluate survival. RESULTS We identified 8 (1.5%), 35(6.7%) and 109 (20.8%) cases with a NTRK1 rearrangement using 15%, 10% and 5% as cut-off values, respectively. We observed copy number (CN) variation of NTRK1 in some cases: 79 (15.1%) cases had a gain in NTRK1 CN ≥ 3, and 24 (4.6%) cases had NTRK1 CN ≥ 4. A NTRK1 rearrangement at the above-mentioned thresholds was not related to disease-free survival (DFS, P = 0.45, 0.47, 0.87) and overall survival (OS, P = 0.80, 0.74, 0.57), respectively. Gain in NTRK1 CN was associated with a poor prognosis irrespective of whether NTRK1 CN ≥ 4 (DFS, P = 0.015; OS, P = 0.035) or NTRK1 CN ≥ 3 (DFS, P = 0.039; OS, P = 0.025). CONCLUSION A NTRK1 rearrangement occurred rarely in ESCC. The increased CN of NTRK1 might be a prognostic indicator for DFS and OS in patients with ESCC.
Collapse
Affiliation(s)
- Zixiang Yu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Haixing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jianfang Xu
- Department of Pathology, Xiamen Branch of Zhongshan Hospital, Fudan University, Xiamen, Fujian, 361015, People's Republic of China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Hao Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xin Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Zhengzeng Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Weijie Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China. .,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China. .,Department of Pathology, Xiamen Branch of Zhongshan Hospital, Fudan University, Xiamen, Fujian, 361015, People's Republic of China.
| |
Collapse
|
14
|
Huang J, Wang X, Zhang X, Chen W, Luan L, Song Q, Wang H, Liu J, Xu L, Xu Y, Shen L, Tan L, Jiang D, Su J, Hou Y. CDK4 Amplification in Esophageal Squamous Cell Carcinoma Associated With Better Patient Outcome. Front Genet 2021; 12:616110. [PMID: 33995474 PMCID: PMC8116700 DOI: 10.3389/fgene.2021.616110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/26/2021] [Indexed: 01/01/2023] Open
Abstract
In the present study, we aimed to investigate the clinical and prognostic values of CDK4 amplification and improve the risk stratification in patients with esophageal squamous cell carcinoma. CDK4 amplification was analyzed by fluorescence in situ hybridization using tissue microarray consisting of representative tissues of 520 patients with esophageal squamous cell carcinoma, and its correlation with clinicopathological features and clinical outcomes were evaluated. CDK4 amplification was found in 8.5% (44/520) of patients with esophageal squamous cell carcinoma. CDK4 amplification was negatively correlated with disease progression (P = 0.003) and death (P = 0.006). Patients with CDK4 amplification showed a significantly better disease-free survival (P = 0.016) and overall survival (P = 0.023) compared with those patients without CDK4 amplification. When patients were further stratified into I–II stage groups and III–IV stage groups, CDK4 amplification was significantly associated with both better disease-free survival (P = 0.023) and overall survival (P = 0.025) in the I–II stage group rather than the III–IV stage group. On univariate and multivariate analysis, invasive depth and CDK4 amplification were associated with disease-free survival and overall survival. Taken together, CDK4 amplification was identified as an independent prognostic factor for survival, which could be incorporated into the tumor–node–metastasis staging system to refine risk stratification of patients with esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiang Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xue Zhang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weijie Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lijuan Luan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Liu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Licheng Shen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Wang X, Langer EM, Daniel CJ, Janghorban M, Wu V, Wang XJ, Sears RC. Altering MYC phosphorylation in the epidermis increases the stem cell population and contributes to the development, progression, and metastasis of squamous cell carcinoma. Oncogenesis 2020; 9:79. [PMID: 32895364 PMCID: PMC7477541 DOI: 10.1038/s41389-020-00261-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/06/2020] [Accepted: 08/13/2020] [Indexed: 12/31/2022] Open
Abstract
cMYC (MYC) is a potent oncoprotein that is subject to post-translational modifications that affect its stability and activity. Here, we show that Serine 62 phosphorylation, which increases MYC stability and oncogenic activity, is elevated while Threonine 58 phosphorylation, which targets MYC for degradation, is decreased in squamous cell carcinoma (SCC). The oncogenic role of MYC in the development of SCC is unclear since studies have shown in normal skin that wild-type MYC overexpression can drive loss of stem cells and epidermal differentiation. To investigate whether and how altered MYC phosphorylation might affect SCC development, progression, and metastasis, we generated mice with inducible expression of MYCWT or MYCT58A in the basal layer of the skin epidermis. In the T58A mutant, MYC is stabilized with constitutive S62 phosphorylation. When challenged with DMBA/TPA-mediated carcinogenesis, MYCT58A mice had accelerated development of papillomas, increased conversion to malignant lesions, and increased metastasis as compared to MYCWT mice. In addition, MYCT58A-driven SCC displayed stem cell gene expression not observed with MYCWT, including increased expression of Lgr6, Sox2, and CD34. In support of MYCT58A enhancing stem cell phenotypes, its expression was associated with an increased number of BrdU long-term label-retaining cells, increased CD34 expression in hair follicles, and increased colony formation from neonatal keratinocytes. Together, these results indicate that altering MYC phosphorylation changes its oncogenic activity—instead of diminishing establishment and/or maintenance of epidermal stem cell populations like wild-type MYC, pS62-MYC enhances these populations and, under carcinogenic conditions, pS62-MYC expression results in aggressive tumor phenotypes.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Ellen M Langer
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Colin J Daniel
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Mahnaz Janghorban
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Vivian Wu
- Department of Otolaryngology-HNS, Henry Ford Health System, Detroit, MI, USA
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA.,Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO, USA
| | - Rosalie C Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA. .,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|