1
|
Minakata N, Ikematsu H, Kiyomi F, Inoue S, Akutagawa T, Watanabe T, Yano T, Shimoda R. Usefulness of virtual scale endoscope for early gastrointestinal lesions. DEN OPEN 2025; 5:e386. [PMID: 38903962 PMCID: PMC11187934 DOI: 10.1002/deo2.386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 06/22/2024]
Abstract
Objectives For early gastrointestinal lesions, size is an important factor in the selection of treatment. Virtual scale endoscope (VSE) is a newly developed endoscope that can measure size more accurately than visual measurement. This study aimed to investigate whether VSE measurement is accurate for early gastrointestinal lesions of various sizes and morphologies. Methods This study prospectively enrolled patients with early gastrointestinal lesions ≤20 mm in size visually. Lesion sizes were measured in the gastrointestinal tract visually, on endoscopic resection specimens with VSE, and finally on endoscopic resection specimens using a ruler. The primary endpoint was the normalized difference (ND) of VSE measurement. The secondary endpoints were the ND of visual measurement and the variation between NDs of VSE and visual measurements. ND was calculated as (100 × [measured size - true size] / true size) (%). True size was defined as size measured using a ruler. Results This study included 60 lesions from April 2022 to December 2022, with 20 each in the esophagus, stomach, and colon. The lesion size was 14.0 ± 6.3 mm (mean ± standard deviation). Morphologies were protruded, slightly elevated, and flat or slightly depressed type in 8, 24, and 28 lesions, respectively. The primary endpoint was 0.3 ± 8.8%. In the secondary endpoints, the ND of visual measurement was -1.7 ± 29.3%, and the variability was significantly smaller in the ND of VSE measurement than in that of visual measurement (p < 0.001, F-test). Conclusions VSE measurement is accurate for early gastrointestinal lesions of various sizes and morphologies.
Collapse
Affiliation(s)
- Nobuhisa Minakata
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastChibaJapan
| | - Hiroaki Ikematsu
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastChibaJapan
| | - Fumiaki Kiyomi
- Department of Statistics and Data CenterClinical Research Support Center KyushuFukuokaJapan
| | - Suma Inoue
- Department of Internal MedicineDivision of GastroenterologySaga UniversitySagaJapan
| | - Takashi Akutagawa
- Department of Endoscopic Diagnostics and TherapeuticsSaga University HospitalSagaJapan
| | - Takashi Watanabe
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastChibaJapan
| | - Tomonori Yano
- Department of Gastroenterology and EndoscopyNational Cancer Center Hospital EastChibaJapan
| | - Ryo Shimoda
- Department of Endoscopic Diagnostics and TherapeuticsSaga University HospitalSagaJapan
| |
Collapse
|
2
|
Li B, Du YY, Tan WM, He DL, Qi ZP, Yu HH, Shi Q, Ren Z, Cai MY, Yan B, Cai SL, Zhong YS. Effect of computer aided detection system on esophageal neoplasm diagnosis in varied levels of endoscopists. NPJ Digit Med 2025; 8:160. [PMID: 40082585 PMCID: PMC11906877 DOI: 10.1038/s41746-025-01532-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 02/19/2025] [Indexed: 03/16/2025] Open
Abstract
A computer-aided detection (CAD) system for early esophagus carcinoma identification during endoscopy with narrow-band imaging (NBI) was evaluated in a large-scale, prospective, tandem, randomized controlled trial to assess its effectiveness. The study was registered at the Chinese Clinical Trial Registry (ChiCTR2100050654, 2021/09/01). Involving 3400 patients were randomly assigned to either routine (routine-first) or CAD-assisted (CAD-first) NBI endoscopy, followed by the other procedure, with targeted biopsies taken at the end of the second examination. The primary outcome was the diagnosis of 1 or more neoplastic lesion of esophagus during the first examination. The CAD-first group demonstrated a significantly higher neoplastic lesion detection rate (3.12%) compared to the routine-first group (1.59%) with a relative detection ratio of 1.96 (P = 0.0047). Subgroup analysis revealed a higher detection rate in junior endoscopists using CAD-first, while no significant difference was observed for senior endoscopists. The CAD system significantly improved esophageal neoplasm detection, particularly benefiting junior endoscopists.
Collapse
Affiliation(s)
- Bing Li
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Yan-Yun Du
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Wei-Min Tan
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, Shanghai, China
| | - Dong-Li He
- Endoscopy Center, Xuhui Hospital, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Zhi-Peng Qi
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Hon-Ho Yu
- Department of Gastroenterology, Kiang Wu Hospital, Macau SAR, China
| | - Qiang Shi
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Zhong Ren
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Ming-Yan Cai
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Bo Yan
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, Shanghai, China.
| | - Shi-Lun Cai
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, China.
- Endoscopy Center, Xuhui Hospital, Zhongshan Hospital of Fudan University, Shanghai, China.
| | - Yun-Shi Zhong
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, China.
- Endoscopy Center, Xuhui Hospital, Zhongshan Hospital of Fudan University, Shanghai, China.
- Endoscopy Center, Shanghai Geriatric Medical Center, Shanghai, China.
| |
Collapse
|
3
|
Liu R, Yuan X, Huang K, Peng T, Pavlov PV, Zhang W, Wu C, Feoktistova KV, Bi X, Zhang Y, Chen X, George J, Liu S, Liu W, Zhang Y, Yang J, Pang M, Hu B, Yi Z, Ye L. Artificial intelligence-based automated surgical workflow recognition in esophageal endoscopic submucosal dissection: an international multicenter study (with video). Surg Endosc 2025:10.1007/s00464-025-11644-1. [PMID: 40072547 DOI: 10.1007/s00464-025-11644-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/22/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Endoscopic submucosal dissection (ESD) is a crucial yet challenging multi-phase procedure for treating early gastrointestinal cancers. This study developed an artificial intelligence (AI)-based automated surgical workflow recognition model for esophageal ESD and proposed an innovative training program based on esophageal ESD videos with or without AI labels to evaluate its effectiveness for trainees. METHODS We retrospectively analyzed complete ESD videos collected from seven hospitals worldwide between 2016 and 2024. The ESD surgical workflow was divided into 6 phases and these videos were divided into five datasets for AI model. Trainees were invited to participate in this multimedia training program and were assigned to the AI or control group randomly. The performance of the AI model and label testing were evaluated using the accuracy. RESULTS A total of 195 ESD videos (782,488 s, 9268 phases) were included. The AI model achieved accuracy of 92.08% (95% confidence interval (CI), 91.40-92.76%), 91.71% (95% CI 90.11-93.31%), and 89.84% (95% CI 87.42-92.25%) in the training, internal, and external test dataset (esophagus), respectively. It also achieved acceptable results in the external test dataset (stomach, colorectum). For the training program, the overall label testing accuracy of the AI group learning ESD videos with AI labels was 88.73 ± 2.97%, significantly higher than the control group without AI labels (81.51 ± 4.63%, P < 0.001). CONCLUSION The AI model achieved high accuracy in the large ESD video datasets. The training program improves understanding of the complexity of ESD workflow and demonstrates the program's effectiveness for trainees.
Collapse
Affiliation(s)
- Ruide Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China
| | - Xianglei Yuan
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China
| | - Kaide Huang
- Center of Intelligent Medicine, College of Computer Science, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu City, 610065, Sichuan Province, China
| | - Tingfa Peng
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China
- Department of Gastroenterology, Armed Police Forces Hospital of Sichuan, Leshan, China
| | - Pavel V Pavlov
- Department of Diagnostic and Therapeutic Endoscopy of the University Clinical Hospital 2, Sechenov University, Moscow, Russia
| | - Wanhong Zhang
- Department of Gastroenterology, Cangxi People's Hospital, Guangyuan, China
| | - Chuncheng Wu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China
| | - Kseniia V Feoktistova
- Department of Diagnostic and Therapeutic Endoscopy of the University Clinical Hospital 2, Sechenov University, Moscow, Russia
| | - Xiaogang Bi
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, China
| | - Yan Zhang
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, China
| | - Xin Chen
- Department of Gastroenterology, Pangang Group General Hospital, Panzhihua, China
| | - Jeffey George
- Department of Gastroenterology, Medical Gastroenterology, Aster Medcity, Kochi, India
| | - Shuang Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China
| | - Wei Liu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China
| | - Yuhang Zhang
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China
| | - Juliana Yang
- Department of Gastroenterology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Maoyin Pang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, USA
| | - Bing Hu
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China.
| | - Zhang Yi
- Center of Intelligent Medicine, College of Computer Science, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu City, 610065, Sichuan Province, China.
| | - Liansong Ye
- Department of Gastroenterology and Hepatology, Digestive Endoscopy Medical Engineering Research Laboratory, Wuhou District, West China Hospital, Sichuan University, No. 37, Guo Xue Alley, Chengdu City, 610041, Sichuan Province, China.
| |
Collapse
|
4
|
Hu B. Exploring artificial intelligence-assisted diagnosis of esophageal squamous cell carcinoma: insights from a clinical trial. Endoscopy 2025; 57:218-219. [PMID: 39608407 DOI: 10.1055/a-2466-6331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Affiliation(s)
- Bing Hu
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Liu H, Zhang Y, Wang Y, Pang K, Xi W, Zou L, He K, Wang Q, Huang L. Is endoscopic submucosal tunnel dissection better than endoscopic submucosal dissection in treating large superficial esophageal neoplastic lesions? A systematic review and meta-analysis. Therap Adv Gastroenterol 2025; 18:17562848251324227. [PMID: 40028510 PMCID: PMC11869307 DOI: 10.1177/17562848251324227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Background The resection of large superficial esophageal neoplastic lesions (SENLs) presents significant challenges for traditional endoscopic submucosal dissection (ESD). Endoscopic submucosal tunnel dissection (ESTD) has emerged as an alternative that potentially reduces resection difficulty. Objectives We aimed to compare ESTD and ESD in the treatment of large SENLs. Design Meta-analysis of randomized controlled trials (RCTs). Data sources and methods We systematically searched MEDLINE, EMBASE, Cochrane Library, and Wanfang Data for RCTs comparing ESTD with ESD for large SENLs until July 1, 2024. The grading of recommendations assessment, development, and evaluation framework was used to assess the certainty of the evidence, whereas trial sequential analysis (TSA) was used to control for random errors and evaluate conclusion validity. Results Four RCTs involving 315 patients were included. The pooled analysis showed that ESTD was significantly faster than ESD (mean differences 5.06, 95% confidence interval: 3.31-6.80; p < 0.01; I 2 = 0%; low certainty of evidence). TSA indicated a desired sample size of 162, with the cumulative Z curve crossing the trial sequential monitoring boundary. ESTD also had lower rates of major complications and post-operation esophageal stricture (low certainty of evidence). No significant differences were found in en bloc and curative resection rates. Conclusion With low certainty, ESTD appears superior to ESD for large SENLs, offering faster resection and fewer complications, with similar en bloc and curative resection rates. Trial registration This meta-analysis protocol was registered on PROSPERO (CRD42024520754).
Collapse
Affiliation(s)
- Huimin Liu
- Department of Gastroenterology, Affiliated Yantai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Gastroenterology, The People’s Hospital of Xizang Autonomous Region, Lhasa, China
| | - Yueyi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yabing Wang
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical College, Beijing, China
| | - Ke Pang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenfeng Xi
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Long Zou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kun He
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, Beijing, China
- Department of Gastroenterology, The People’s Hospital of Xizang Autonomous Region, No.18 Linkuo North Road, Chengguan District, Lhasa, Tibet Autonomous Region, China
| | - Qiang Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, Beijing, China
- Department of Gastroenterology, The People’s Hospital of Xizang Autonomous Region, No.18 Linkuo North Road, Chengguan District, Lhasa, Tibet Autonomous Region, China
| | - Liuye Huang
- Department of Gastroenterology, Affiliated Yantai Yuhuangding Hospital of Qingdao University Medical College, No.20 Yudong Road, Zhifu District, Yantai, Shandong, China
| |
Collapse
|
6
|
Ohsawa M, Hamai Y, Emi M, Ibuki Y, Kurokawa T, Hirohata R, Kitasaki N, Okada M. Recurrence and prognostic predictors in pathologic T1N0 esophageal squamous cell carcinoma treated with surgery alone. Surgery 2025; 178:108863. [PMID: 39419644 DOI: 10.1016/j.surg.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 09/14/2024] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Patients diagnosed with pathologic T1N0 esophageal squamous cell carcinoma and treated with surgery alone have a good prognosis and are generally followed up without adjuvant therapy. However, recurrence has been observed in this patient group. Therefore, this study aimed to identify recurrence and prognostic factors in patients with pathologic T1N0 esophageal squamous cell carcinoma who were treated with surgery alone. METHODS Of the 532 patients who underwent esophagectomy with R0 resection at Hiroshima University Hospital between August 2003 and November 2018, 124 who underwent only esophagectomy and had pathological T1N0 esophageal squamous cell carcinoma were included in the study. Recurrence and prognostic factors were analyzed and details of recurrence were evaluated. RESULTS The 5-year recurrence-free survival and 5-year overall survival rates were 84.7% and 87.2%, respectively. Recurrence was observed in 12 (9.7%) patients. Univariate and multivariate analyses showed that the histologic type (poorly differentiated compared with others) and lymphatic and/or vascular invasion (positive compared with negative) were statistically significant for recurrence-free survival. Kaplan-Meier curves for recurrence-free survival and overall survival showed that prognosis was significantly stratified according to these factors. All patients with poorly differentiated and positive lymphatic and/or vascular invasion experienced recurrence and recurrence pattern is all distant metastases. CONCLUSIONS Poorly differentiated and lymphatic and/or vascular invasion are important recurrence and prognostic predictors in pathologic T1N0 esophageal squamous cell carcinoma treated with surgery alone. Patients with these prognostic factors experienced increased recurrence rates, often with distant metastasis. Therefore, adjuvant therapy may be beneficial for such patients and follow-ups should be performed at closer intervals.
Collapse
Affiliation(s)
- Manato Ohsawa
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Yoichi Hamai
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan.
| | - Manabu Emi
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Yuta Ibuki
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Tomoaki Kurokawa
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Ryosuke Hirohata
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Nao Kitasaki
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
7
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [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: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
8
|
Li P, Gao X, Huang D, Gu X. Identification and Characterization of Prognostic Macrophage Subpopulations for Human Esophageal Carcinoma. Curr Med Chem 2025; 32:123-135. [PMID: 38362682 DOI: 10.2174/0109298673284207240108105724] [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: 10/09/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 02/17/2024]
Abstract
AIMS The aim of the present study was to investigate the relationship between the cellular ecosystem and the progression of esophageal carcinoma (ESCA) based on the evolution of macrophages and to analyze the potential of using macrophages as a new therapeutic approach in ESCA treatment. BACKGROUND Macrophage-based immunotherapy could be used for treating ESCA patients, but its clinical application is limited by the intra-tumor heterogeneity of macrophages. OBJECTIVE The objective of this study was to analyze the diversity, differentiation trajectory, and intercellular communication of macrophages in ESCA and its prognostic significance. METHODS Single-cell RNA sequencing (scRNA-seq) data in the GSE154763 dataset were downloaded from Gene Expression Omnibus (GEO) to identify cell clusters and annotate cell types using the Seurat R package. The scRNA-seq profiles of macrophages were extracted, and cluster analysis was performed to identify macrophage subsets. The differentiation trajectories of macrophage subgroups were visualized employing Monocle2. Finally, ligand-receptor pairs and communication intensity among the classified subgroups were analyzed using Cell Chat. RESULTS A total of 8 cell types were identified between ESCA tissues and paracancer tissues. The most abundant macrophages in ESCA tissues were further divided into 5 cell clusters. Compared with the normal tissues, the proportion of HSPA6+ macrophages in ESCA tissues increased the most, and the number of ligand-receptor pairs that mediated the communication of HSPA6+ macrophages with mast cells and monocytes also increased significantly. More importantly, a high proportion of HSPA6+ macrophages was inversely correlated with the survival outcomes for ESCA patients. CONCLUSIONS This study analyzed the diversity, distribution and differentiation trajectory of macrophages in ESCA tissues at single-cell level and classified a prognostic macrophage subtype (HSPA6+ macrophages) of ESCA, providing a theoretical basis for macrophage-targeted therapy in ESCA.
Collapse
Affiliation(s)
- Penghui Li
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, 471000, China
| | - Xiaohui Gao
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, 471000, China
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, 471000, China
| |
Collapse
|
9
|
Liu Y, Lan S, Duan Z. circ-TTC17 Promotes Esophagus Squamous Cell Carcinoma Cell Growth, Metastasis, and Inhibits Autophagy-Mediated Radiosensitivity Through miR-145-5p/SIRT1 Axis. Thorac Cancer 2025; 16:e15494. [PMID: 39621506 PMCID: PMC11729993 DOI: 10.1111/1759-7714.15494] [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: 08/21/2024] [Revised: 10/22/2024] [Accepted: 11/11/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Circular RNA (circRNA) plays a significant role in esophagus squamous cell carcinoma (ESCC) progression. Nevertheless, circ-TTC17 roles in ESCC have not fully understood. METHODS The levels of circ-TTC17, miR-145-5p and sirtuin 1 (SIRT1) were determined using qRT-PCR. ESCC cell functions were examined by CCK8 assay, flow cytometry, transwell assay and colony formation assay. The relative protein levels of autophagy marker and SIRT1 were determined by western blot (WB). The interactions among circ-TTC17, miR-145-5p, and SIRT1 were verified by dual-luciferase reporter assay and RIP assay. RESULTS circ-TTC17 was overexpressed and miR-145-5p was underexpressed in ESCC. circ-TTC17 knockdown restrained ESCC cell proliferation and metastasis, while enhance apoptosis and autophagy-mediated radiosensitivity. Circ-TTC17 could sponge miR-145-5p, and its inhibitor reversed the inhibitory effect of circ-TTC17 knockdown on ESCC cell progression. Additionally, SIRT1 was targeted by miR-145-5p, and SIRT1 overexpression abolished miR-145-5p-mediated the suppressive effect on ESCC cell progression. Also, circ-TTC17 interference reduced ESCC tumor growth via miR-145-5p/SIRT1 axis. CONCLUSION circ-TTC17 promoted ESCC cell growth, metastasis and inhibited autophagy-mediated radiosensitivity by miR-145-5p/SIRT1 axis.
Collapse
Affiliation(s)
- Ying Liu
- Department of Head and Neck Radiotherapy CombinedShanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Shengmin Lan
- Department of Head and Neck Radiotherapy CombinedShanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Zhihui Duan
- Department of Thoracic SurgeryShanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
10
|
Talasila P, Hedge SG, Periasamy K, Nagaraj SS, Singh H, Singh H, Gupta P. Imaging in Esophageal Cancer: A Comprehensive Review. Indian J Radiol Imaging 2025; 35:123-138. [PMID: 39697520 PMCID: PMC11651834 DOI: 10.1055/s-0044-1786871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
Esophageal cancer is one of the common cancers. Risk factors are well recognized and lead most commonly to two distinct histological subtypes (squamous cell carcinoma and adenocarcinoma). The diagnosis is based on endoscopic evaluation. The most challenging aspect of management is accurate staging as it guides appropriate management. Endoscopic ultrasound, computed tomography (CT), positron emission tomography-CT, and magnetic resonance imaging are the imaging tests employed for the staging. Each imaging test has its own merits and demerits. Imaging is also critical to evaluate posttreatment complication and for response assessment. In this review article, we discuss in detail the risk factors, anatomical aspects, and role of imaging test in staging and evaluation of complications and response after treatment.
Collapse
Affiliation(s)
- Pallavi Talasila
- Department of Radiodiagnosis and Imaging, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Swaroop G. Hedge
- Department of Radiodiagnosis and Imaging, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kannan Periasamy
- Department of Radiation Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Satish Subbiah Nagaraj
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harmandeep Singh
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harjeet Singh
- Department of Surgical Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Gupta
- Department of Radiodiagnosis and Imaging, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
11
|
Zheng Y, Xing W, BingWei, Liu X, Liang G, Yuan D, Yang K, Wang W, Chen D, Ma J. Detection of a novel DNA methylation marker panel for esophageal cancer diagnosis using circulating tumor DNA. BMC Cancer 2024; 24:1578. [PMID: 39725880 DOI: 10.1186/s12885-024-13301-7] [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: 07/22/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Esophageal cancer (ECa) is one of the most deadly cancers, with increasing incidence worldwide and poor prognosis. While endoscopy is recommended for the detection of ECa in high-risk individuals, it is not suitable for large-scale screening due to its invasiveness and inconvenience. METHODS In this study, a novel gene methylation panel was developed for a blood-based test, and its diagnostic efficacy was evaluated using a cohort of 304 participants (203 cases, 101 controls). The assessment focused on the DNA methylation levels of SEPTIN9, tissue factor pathway inhibitor 2 (TFPI2), and the fragile histidine triad gene (FHIT) in patients with ECa, benign esophageal disease, and healthy controls. The receiver operating characteristic (ROC) curve was generated for the panel to calculate the area under the curve (AUC), sensitivity, specificity, and 95% confidence intervals (CIs), along with a comparison to the gold standard of pathological examination. The consistency between biomarker and pathological diagnosis was evaluated with kappa analysis conducted with IBM SPSS Statistics. The Chi-square test or Fisher's exact test was utilized to assess the association of test positivity with demographic characteristics. RESULTS In patients with ECa, SEPTIN9, TFPI2, and FHIT DNA methylation levels were significantly higher compared to those with benign esophageal disease or healthy controls. The panel demonstrated promising potential as a noninvasive tool for distinguishing malignant tumors from both healthy controls and benign esophageal diseases, achieving an area under the ROC curve of 0.925 (95% CI: 0.889-0.952), with a sensitivity of 79.8% [95% CI 73.6-85.1%] and specificity of 95.0% [95% CI 88.8-98.4%]. In particular, the panel showed exceptional diagnostic efficiency for stage 0, I, and II cancer patients with sensitivity at 69.0, 75.5%, and 78.9%, respectively. The comparison revealed a Kappa value of 0.725 between RT-PCR testing and the established gold standard of pathological examination, indicating a high level of consistency. Additionally, there was no bias in diagnostic efficiency based on age, gender, or the presence of other malignancies (non-esophageal cancers). CONCLUSIONS The study's findings suggested that the DNA methylation biomarkers panel holds promise as a non-invasive and convenient diagnostic test for ECa. The panel's ability to distinguish malignant tumors from benign esophageal diseases, coupled with its high sensitivity and specificity, presented opportunities to enhance the over-all diagnosis of high-risk population when in conjunction with existing detection methods.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Wenqun Xing
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - BingWei
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No 127, Dongming Road, Zhengzhou, 450008, Henan, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, Henan, China
| | - Xianben Liu
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Guanghui Liang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Dongfeng Yuan
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Ke Yang
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No 127, Dongming Road, Zhengzhou, 450008, Henan, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, Henan, China
| | - Weizhen Wang
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No 127, Dongming Road, Zhengzhou, 450008, Henan, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, Henan, China
| | - Dongxu Chen
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No 127, Dongming Road, Zhengzhou, 450008, Henan, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, Henan, China
| | - Jie Ma
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No 127, Dongming Road, Zhengzhou, 450008, Henan, China.
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, Henan, China.
| |
Collapse
|
12
|
Hassan C, Antonelli G, Chiu PWY, Emura F, Goda K, Prasad I, Al Awadhi S, Al Lehibi A, Arantes V, Cerisoli CL, Draganov P, Fleischer D, Fluxá F, Gonzalez N, Inoue H, John S, Kashin S, Khashab M, Kim GH, Kothari S, Ngamruengphong S, Remes-Troche JM, Sharara AI, Shimamura Y, Villa-Gomez G, Wang KK, Wang WL, Yip HC, Sharma P. Position statement of the World Endoscopy Organization: Role of endoscopy in screening, diagnosis, and treatment of esophageal superficial squamous neoplasia. Dig Endosc 2024. [PMID: 39722219 DOI: 10.1111/den.14967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/10/2024] [Indexed: 12/28/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) remains a significant global health challenge, being the sixth leading cause of cancer mortality with pronounced geographic variability. The incidence rates range from 125 per 100,000 in northern China to 1-1.5 per 100,000 in the United States, driven by environmental and lifestyle factors such as tobacco and alcohol use, dietary habits, and pollution. Major modifiable risk factors include tobacco and alcohol consumption, with a synergistic risk increase when combined. Nonmodifiable risk factors include previous diagnoses of head and neck squamous cell carcinoma (H&N SCC), achalasia, and prior radiotherapy. Prevention strategies must be tailored to specific regional burdens to efficiently allocate medical and financial resources. Gastrointestinal endoscopy is crucial in reducing ESCC burden through early detection and characterization of neoplastic changes, such as high-grade dysplasia. Early diagnosis significantly improves survival rates, while endoscopic resection of noninvasive dysplasia can prevent ESCC onset, reducing treatment burden for advanced disease. Postresection surveillance can detect high-risk metachronous lesions. Despite these benefits, endoscopic prevention faces challenges, including the lack of high-level evidence supporting its efficacy, opportunity costs, the need for specialized training and techniques, and the requirement for advanced technology investments. This Position Statement from the World Endoscopy Organization (WEO) aims to address these challenges, supplying recommendations for the exploitation of endoscopic resources regarding the possible role of screening, quality, and training for the detection, characterization, resection, and surveillance of ESCC.
Collapse
Affiliation(s)
- Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Giulio Antonelli
- Gastroenterology and Digestive Endoscopy Unit, Ospedale dei Castelli Hospital, Rome, Italy
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Philip Wai-Yan Chiu
- Division of Upper Gastrointestinal and Metabolic Surgery, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Fabian Emura
- Digestive Health and Liver Diseases, University of Miami, Miami, USA
- Interventional Endoscopy Center, Jackson Memorial Hospital, Miami, USA
| | - Kenichi Goda
- Gastrointestinal Endoscopy Center, Dokkyo Medical University Hospital, Tochigi, Japan
| | - Iyer Prasad
- Esophageal Interest Group, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | - Sameer Al Awadhi
- Rashid Hospital, Dubai Health Authority, Dubai, United Arab Emirates
| | - Abed Al Lehibi
- Gastroenterology and Hepatology Department, King Fahad Medical City, Riyad, Saudi Arabia
| | - Vitor Arantes
- Endoscopy Unit, Alfa Institute of Gastroenterology, School of Medicine, Federal University of Minas Gerais, Hospital Mater Dei Contorno, Belo Horizonte, Brazil
| | - Cecilio L Cerisoli
- Therapeutic and Diagnostic Gastroenterology (GEDYT) Center, Buenos Aires, Argentina
| | | | - David Fleischer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, USA
| | - Fernando Fluxá
- Gastroenterology Department Clinica Meds, Santiago, Chile
| | | | - Haruhiro Inoue
- Digestive Diseases Center, Showa University Koto Toyosu Hospital, Tokyo, Japan
| | - Sneha John
- Endoscopy Unit, Gold Coast University Hospital, Southport, Australia
| | - Sergey Kashin
- Endoscopy Department, Yaroslavl State Medical University, Yaroslavl, Russia
| | - Mouen Khashab
- Therapeutic Endoscopy, Johns Hopkins Hospital, Baltimore, USA
| | - Gwang Ha Kim
- Department of Internal Medicine, Pusan National University College of Medicine, Busan, South Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Shivangi Kothari
- Division of Gastroenterology and Hepatology, University of Rochester Medical Center, Rochester, USA
| | | | | | - Ala I Sharara
- Division of Gastroenterology, American University of Beirut Medical Center, Beirut, Lebanon
| | | | - Guido Villa-Gomez
- Gastroenterology and Digestive Endoscopy Unit, WGO La Paz Training Center, La Paz, Bolivia
| | - Kenneth K Wang
- Russ and Kathy Van Cleve Professor of Gastroenterology, Mayo Clinic, Rochester, USA
| | - Wen-Lun Wang
- Department of Internal Medicine, E-Da Hospital/I-Shou University, Kaohsiung, Taiwan
| | - Hon-Chi Yip
- Division of Upper Gastrointestinal and Metabolic Surgery, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Prateek Sharma
- University of Kansas School of Medicine and VA Medical Center, Kansas City, USA
| |
Collapse
|
13
|
Qi L, Sun M, Liu W, Zhang X, Yu Y, Tian Z, Ni Z, Zheng R, Li Y. Global esophageal cancer epidemiology in 2022 and predictions for 2050: A comprehensive analysis and projections based on GLOBOCAN data. Chin Med J (Engl) 2024; 137:3108-3116. [PMID: 39668405 PMCID: PMC11706580 DOI: 10.1097/cm9.0000000000003420] [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: 05/05/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND The burden of esophageal cancer varies across different regions of the world. The aim of this study is to analyze the current burden of esophageal cancer in 185 countries in 2022 and to project the trends up to the year 2050. METHODS We extracted data on primary esophageal cancer cases and deaths from the GLOBOCAN 2022 database, which includes data from 185 countries. Age-standardized incidence rates (ASIR) and mortality rates (ASMR) per 100,000 person-years were calculated by stratifying by Human Development Index (HDI) levels and regions. Considering changes in population size and age structure, we assumed that the risks of incidence and mortality remain constant at the levels of 2022 to forecast the number of new cases and deaths from esophageal cancer globally by 2050. RESULTS In 2022, an estimated 511,054 people were diagnosed with esophageal cancer globally, and 445,391 died from the disease. The global ASIR and ASMR for esophageal cancer were 5.00 and 4.30 per 100,000, respectively. The highest rates were observed in East Africa (7.60 for incidence, 7.20 for mortality per 100,000), East Asia (7.60 for incidence, 5.90 for mortality per 100,000), Southern Africa (6.30 for incidence, 5.90 for mortality per 100,000), and South Central Asia (5.80 for incidence, 5.50 for mortality per 100,000). Among the 185 countries worldwide, esophageal cancer was among the top five causes of cancer incidence in 18 countries and among the top five causes of cancer mortality in 25 countries. In 2022, China had 224,012 new cases and 187,467 deaths from esophageal cancer, accounting for approximately 43.8% and 42.1% of the global total, respectively, which is higher than the proportion of China's population to the global population (17.9%). ASIR was 8.30 per 100,000, and ASMR was 6.70 per 100,000. The highest burden of esophageal cancer was in high HDI countries, with new cases and deaths accounting for 51.3% and 50.0% of the global total, respectively. The ASIR and ASMR were highest in the high HDI group (6.10 and 5.10 per 100,000, respectively), also exceeding the global averages. There was a trend of decreasing mortality to incidence ratio with increasing HDI, but no correlation was observed between HDI and ASIR or ASMR. In all regions worldwide, the incidence and mortality rates were higher in males than in females (with a male-to-female ASR ratio ranging from 1.10 to 28.7). Compared to 2022, it is projected that by 2050, the number of new esophageal cancer cases will increase by approximately 80.5%, and deaths will increase by 85.4% due to population growth and aging. CONCLUSIONS The burden of esophageal cancer remains heavy. Adopting a healthy lifestyle, including reducing tobacco and alcohol intake, avoiding moldy foods, and increasing intake of fresh fruits and vegetables, can help reduce the risk of stomach and esophageal cancer. In addition, the development and implementation of evidence-based and effective public health policies are critical to reducing the global disease burden of esophageal cancer.
Collapse
Affiliation(s)
- Ling Qi
- Department of Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Mengfei Sun
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, China
| | - Weixin Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Xuefeng Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Yongjun Yu
- Department of Thoracic Surgery, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010110, China
- Department of Cardio-Thoracic Surgery, The Second Hospital of Chifeng, Chifeng, Inner Mongolia Autonomous Region 024099, China
| | - Ziqiang Tian
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 110000, China
| | - Zhiyu Ni
- Central Laboratory, Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
- Medical College, Hebei University of Engineering, Handan, Hebei 056038, China
- Affiliated Hospital of Hebei Engineering University, Handan, Hebei 056002, China
| | - Rongshou Zheng
- 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
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
- Central Laboratory, Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| |
Collapse
|
14
|
Sun Y, Ying K, Sun J, Qiu L, Wang Y, Ji M, Zhou L, Chen J. Curcumin mediates glutathione depletion via metal-organic framework nanocarriers to enhance cisplatin chemosensitivity on esophageal cancer. DISCOVER NANO 2024; 19:200. [PMID: 39661226 PMCID: PMC11635067 DOI: 10.1186/s11671-024-04168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Cisplatin (CDDP) is the primary drug used in the initial treatment of esophageal cancer (EC). However, its side effects and resistance can limit its effectiveness in clinical therapy. Curcumin (Cur)-mediated glutathione (GSH) depletion can reverse resistance, enhance the chemosensitivity of CDDP, and further improve the efficacy of platinum-containing chemotherapy in the treatment of esophageal cancer. However, it is also faced with problems of poor water solubility and low bioavailability in vivo, which severely hinders cancer treatments. In order to address these issues, we developed a novel nanotherapeutic system called CDCZA, combining Cur/CDDP/Cu/ZIF8@Au to enhance chemotherapy through GSH depletion and chemodynamic therapy through self-produced H2O2. Cu and CDDP were precisely co-loaded into Cu/ZIF8 nanoparticles using a one-pot method, then ultra-small gold nanoparticles mimicking glucose oxidase (Au nanoparticles) were embedded in the outer shell to create the CDCZA nano system. The released Cur could notably decrease intracellular GSH content and thus improve the chemosensitivity of CDDP, resulting in severe cellular apoptosis. And the Au nanoparticles effectively enabled chemodynamic therapy enhancement by accelerating the depletion of β-D-glucose into H2O2. As a result, the CDCZA nanoparticles showed increased tumor accumulation and improved antitumor effectiveness in a model of EC. Taken together, this work provides a new idea for the clinical design of efficient treatment reagents for EC.
Collapse
Affiliation(s)
- Yunhao Sun
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China.
| | - Kaijun Ying
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Jian Sun
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Limin Qiu
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Yao Wang
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Mingming Ji
- Department of Thoracic Surgery, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China
| | - Lulu Zhou
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, People's Republic of China.
| | - Jinjin Chen
- Department of Oncology, The First People's Hospital of Yancheng City, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224005, Jiangsu, People's Republic of China.
| |
Collapse
|
15
|
Tang Z, Feng S, Liu Q, Ban Y, Zhang Y. Using Pathomics-Based Model for Predicting Positive Surgical Margins in Patients with Esophageal Squamous Cell Carcinoma: A Comparative Study of Decision Tree and Nomogram. Int J Gen Med 2024; 17:5869-5882. [PMID: 39659511 PMCID: PMC11629666 DOI: 10.2147/ijgm.s495296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
Objective Esophageal squamous cell carcinoma (ESCC) has a high incidence and mortality rate. Postoperative positive surgical margins (PSM) often correlate with poor prognosis. This study aims to develop and validate a predictive model for PSM positivity in ESCC patients, with the potential to guide preoperative planning and improve patient outcomes. Methods We conducted a retrospective analysis of 1776 patients who underwent esophageal cancer surgery at the First Affiliated Hospital of Jilin University between January 2015 and December 2023. Patients with visible residual tumors (R2) or microscopic residual tumors (R1) at the surgical margins were classified as having PSM. High-dimensional pathological features were extracted from digital pathological sections using CellProfiler software. The selected features were used to develop a predictive model based on decision trees and generalized linear regression, and the model was validated in an independent cohort. Clinically significant pathological factors (P < 0.05) were included in multivariate logistic regression for further validation. The model's performance was assessed using calibration curves and receiver operating characteristic (ROC) curves, generated with the Bootstrap method. Decision curve analysis (DCA) was employed to evaluate the clinical utility of the predictive model. Results A total of 229 patients (12.89%) were diagnosed with PSM. Logistic regression analysis identified multifocal lesions, vascular invasion, and pathomics-based features as independent predictors of PSM. The predictive model, represented by a decision tree, demonstrated good discrimination with an area under the ROC curve of 0.899 (95% CI: 0.842-0.956, P < 0.001), and a strong calibration curve between the predicted probability and the actual probability. Additionally, the nomogram demonstrated slightly inferior discrimination with an area under the ROC curve of 0.803 (95% CI: 0.734-0.872, P < 0.001) in the training cohort. Conclusion Our study successfully established and validated a pathology-based predictive model for PSM risk, which could enhance preoperative evaluation and inform treatment strategies for ESCC.
Collapse
Affiliation(s)
- Ze Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Shiyun Feng
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Qing Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Yunze Ban
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Yan Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
16
|
Wang YK, Karmakar R, Mukundan A, Men TC, Tsao YM, Lu SC, Wu IC, Wang HC. Computer-aided endoscopic diagnostic system modified with hyperspectral imaging for the classification of esophageal neoplasms. Front Oncol 2024; 14:1423405. [PMID: 39687890 PMCID: PMC11646837 DOI: 10.3389/fonc.2024.1423405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 11/04/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction The early detection of esophageal cancer is crucial to enhancing patient survival rates, and endoscopy remains the gold standard for identifying esophageal neoplasms. Despite this fact, accurately diagnosing superficial esophageal neoplasms poses a challenge, even for seasoned endoscopists. Recent advancements in computer-aided diagnostic systems, empowered by artificial intelligence (AI), have shown promising results in elevating the diagnostic precision for early-stage esophageal cancer. Methods In this study, we expanded upon traditional red-green-blue (RGB) imaging by integrating the YOLO neural network algorithm with hyperspectral imaging (HSI) to evaluate the diagnostic efficacy of this innovative AI system for superficial esophageal neoplasms. A total of 1836 endoscopic images were utilized for model training, which included 858 white-light imaging (WLI) and 978 narrow-band imaging (NBI) samples. These images were categorized into three groups, namely, normal esophagus, esophageal squamous dysplasia, and esophageal squamous cell carcinoma (SCC). Results An additional set comprising 257 WLI and 267 NBI images served as the validation dataset to assess diagnostic accuracy. Within the RGB dataset, the diagnostic accuracies of the WLI and NBI systems for classifying images into normal, dysplasia, and SCC categories were 0.83 and 0.82, respectively. Conversely, the HSI dataset yielded higher diagnostic accuracies for the WLI and NBI systems, with scores of 0.90 and 0.89, respectively. Conclusion The HSI dataset outperformed the RGB dataset, demonstrating an overall diagnostic accuracy improvement of 8%. Our findings underscored the advantageous impact of incorporating the HSI dataset in model training. Furthermore, the application of HSI in AI-driven image recognition algorithms significantly enhanced the diagnostic accuracy for early esophageal cancer.
Collapse
Affiliation(s)
- Yao-Kuang Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Riya Karmakar
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - Arvind Mukundan
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - Ting-Chun Men
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - Yu-Ming Tsao
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - Song-Cun Lu
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - I-Chen Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiang-Chen Wang
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi, Taiwan
- Department of Medical Research, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Technology Development, Hitspectra Intelligent Technology Co., Ltd., Kaohsiung, Taiwan
| |
Collapse
|
17
|
Zhou Y, Zhou J, Cai X, Ge S, Sang S, Yang Y, Zhang B, Deng S. Integrating 18F-FDG PET/CT radiomics and body composition for enhanced prognostic assessment in patients with esophageal cancer. BMC Cancer 2024; 24:1402. [PMID: 39543534 PMCID: PMC11566154 DOI: 10.1186/s12885-024-13157-x] [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: 07/30/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND This study aimed to develop a predictive model utilizing radiomics and body composition features derived from 18F-FDG PET/CT scans to forecast progression-free survival (PFS) and overall survival (OS) outcomes in patients with esophageal squamous cell carcinoma (ESCC). METHODS We analyzed data from 91 patients who underwent baseline 18F-FDG PET/CT imaging. Radiomic features extracted from PET and CT images and subsequent radiomics scores (Rad-scores) were calculated. Body composition metrics were also quantified, including muscle and fat distribution at the L3 level from CT scans. Multiparametric survival models were constructed using Cox regression analysis, and their performance was assessed using the area under the time-dependent receiver operating characteristic (ROC) curve (AUC) and concordance index (C-index). RESULTS Multivariate analysis identified Rad-scorePFS (P = 0.003), sarcopenia (P < 0.001), and visceral adipose tissue index (VATI) (P < 0.001) as independent predictors of PFS. For OS, Rad-scoreOS (P = 0.001), sarcopenia (P = 0.002), VATI (P = 0.037), stage (P = 0.042), and body mass index (BMI) (P = 0.008) were confirmed as independent prognostic factors. Integration of the Rad-score with clinical variables and body composition parameters enhanced predictive accuracy, yielding C-indices of 0.810 (95% CI: 0.737-0.884) for PFS and 0.806 (95% CI: 0.720-0.891) for OS. CONCLUSIONS This study underscored the potential of combining Rad-score with clinical and body composition data to refine prognostic assessment in ESCC patients.
Collapse
Affiliation(s)
- Yeye Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jin Zhou
- Department of Nuclear Medicine, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Suqian, China
| | - Xiaowei Cai
- Department of Nuclear Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Shushan Ge
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang, 621099, China
| | - Shibiao Sang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yi Yang
- Department of Nuclear Medicine, Affiliated Hospital of Medical School, Suzhou Hospital, Nanjing University, Suzhou, China.
| | - Bin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang, 621099, China.
| |
Collapse
|
18
|
Zhang C, Li T, Zhao Q, Ma R, Hong Z, Huang X, Gao P, Liu J, Zhao J, Wang Z. Advances and Prospects in Liquid Biopsy Techniques for Malignant Tumor Diagnosis and Surveillance. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404709. [PMID: 39082395 DOI: 10.1002/smll.202404709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/07/2024] [Indexed: 11/02/2024]
Abstract
Liquid biopsy technology provides invaluable support for the early diagnosis of tumors and surveillance of disease course by detecting tumor-related biomarkers in bodily fluids. Currently, liquid biopsy techniques are mainly divided into two categories: biomarker and label-free. Biomarker liquid biopsy techniques utilize specific antibodies or probes to identify and isolate target cells, exosomes, or molecules, and these techniques are widely used in clinical practice. However, they have certain limitations including dependence on tumor markers, alterations in cell biological properties, and high cost. In contrast, label-free liquid biopsy techniques directly utilize physical or chemical properties of cells, exosomes, or molecules for detection and isolation. These techniques have the advantage of not needing labeling, not impacting downstream analysis, and low detection cost. However, most are still in the research stage and not yet mature. This review first discusses recent advances in liquid biopsy techniques for early tumor diagnosis and disease surveillance. Several current techniques are described in detail. These techniques exploit differences in biomarkers, size, density, deformability, electrical properties, and chemical composition in tumor components to achieve highly sensitive tumor component identification and separation. Finally, the current research progress is summarized and the future research directions of the field are discussed.
Collapse
Affiliation(s)
- Chengzhi Zhang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Tenghui Li
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Qian Zhao
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Rui Ma
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Zhengchao Hong
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Xuanzhang Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Peng Gao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Jingjing Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Junhua Zhao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, 155 N Nanjing Street, Shenyang, Liaoning, 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
- Institute of Health Sciences, China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China
| |
Collapse
|
19
|
Zhang WY, Chang YJ, Shi RH. Artificial intelligence enhances the management of esophageal squamous cell carcinoma in the precision oncology era. World J Gastroenterol 2024; 30:4267-4280. [PMID: 39492825 PMCID: PMC11525855 DOI: 10.3748/wjg.v30.i39.4267] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common histological type of esophageal cancer with a poor prognosis. Early diagnosis and prognosis assessment are crucial for improving the survival rate of ESCC patients. With the advancement of artificial intelligence (AI) technology and the proliferation of medical digital information, AI has demonstrated promising sensitivity and accuracy in assisting precise detection, treatment decision-making, and prognosis assessment of ESCC. It has become a unique opportunity to enhance comprehensive clinical management of ESCC in the era of precision oncology. This review examines how AI is applied to the diagnosis, treatment, and prognosis assessment of ESCC in the era of precision oncology, and analyzes the challenges and potential opportunities that AI faces in clinical translation. Through insights into future prospects, it is hoped that this review will contribute to the real-world application of AI in future clinical settings, ultimately alleviating the disease burden caused by ESCC.
Collapse
Affiliation(s)
- Wan-Yue Zhang
- School of Medicine, Southeast University, Nanjing 221000, Jiangsu Province, China
| | - Yong-Jian Chang
- School of Cyber Science and Engineering, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Rui-Hua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
20
|
Cui Y, Luo Z, Wang X, Liang S, Hu G, Chen X, Zuo J, Zhou L, Guo H, Wang X. Analyzing risk factors and constructing a predictive model for superficial esophageal carcinoma with submucosal infiltration exceeding 200 micrometers. BMC Gastroenterol 2024; 24:350. [PMID: 39370515 PMCID: PMC11457335 DOI: 10.1186/s12876-024-03442-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
OBJECTIVE Submucosal infiltration of less than 200 μm is considered an indication for endoscopic surgery in cases of superficial esophageal cancer and precancerous lesions. This study aims to identify the risk factors associated with submucosal infiltration exceeding 200 micrometers in early esophageal cancer and precancerous lesions, as well as to establish and validate an accompanying predictive model. METHODS Risk factors were identified through least absolute shrinkage and selection operator (LASSO) and multivariate logistic regression. Various machine learning (ML) classification models were tested to develop and evaluate the most effective predictive model, with Shapley Additive Explanations (SHAP) employed for model visualization. RESULTS Predictive factors for early esophageal invasion into the submucosa included endoscopic ultrasonography or magnifying endoscopy> SM1(P<0.001,OR = 3.972,95%CI 2.161-7.478), esophageal wall thickening(P<0.001,OR = 12.924,95%CI,5.299-33.96), intake of pickled foods(P=0.04,OR = 1.837,95%CI,1.03-3.307), platelet-lymphocyte ratio(P<0.001,OR = 0.284,95%CI,0.137-0.556), tumor size(P<0.027,OR = 2.369,95%CI,1.128-5.267), the percentage of circumferential mucosal defect(P<0.001,OR = 5.286,95%CI,2.671-10.723), and preoperative pathological type(P<0.001,OR = 4.079,95%CI,2.254-7.476). The logistic regression model constructed from the identified risk factors was found to be the optimal model, demonstrating high efficacy with an area under the curve (AUC) of 0.922 in the training set, 0.899 in the validation set, and 0.850 in the test set. CONCLUSION A logistic regression model complemented by SHAP visualizations effectively identifies early esophageal cancer reaching 200 micrometers into the submucosa.
Collapse
Affiliation(s)
- Yutong Cui
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Zichen Luo
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Xiaobo Wang
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Shiqi Liang
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Guangbing Hu
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Xinrui Chen
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Ji Zuo
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Lu Zhou
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Haiyang Guo
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China
| | - Xianfei Wang
- Department of Gastroenterology, Digestive endoscopy center, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 63700, Sichuan, China.
| |
Collapse
|
21
|
Shao L, Li B. Synaptotagmin 13 Could Drive the Progression of Esophageal Squamous Cell Carcinoma Through Upregulating ACRV1. DNA Cell Biol 2024; 43:452-462. [PMID: 39046915 DOI: 10.1089/dna.2024.0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Abstract
SYT13 is one of the atypical members of the synaptotagmin (SYT) family whose function has attracted considerable attention in recent years. Although SYT13 has been studied in several types of human cancers, such as lung cancer, its role in esophageal squamous cell carcinoma (ESCC) is still unclear. It was demonstrated that SYT13 is significantly upregulated in ESCC tissues compared with normal ones and correlated with higher degree of malignancy. Knockdown of SYT13 could inhibit ESCC cell proliferation and migration, while promoting cell apoptosis. Meanwhile, ESCC cells with relatively lower SYT13 expression grew slower in vivo and finally formed smaller xenografts. Furthermore, acrosomal vesicular protein 1 was identified as a potential downstream target of SYT13, which regulates cell phenotypes of ESCC cells in cooperation with SYT13. All the in vitro and in vivo results in this study identified that SYT13 silencing could be an effective strategy to inhibit the development of ESCC, which could be considered as a promising therapeutic target in the treatment of ESCC.
Collapse
Affiliation(s)
- Longlong Shao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bin Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Chu LY, Wu FC, Fang WK, Hong CQ, Huang LS, Zou HY, Peng YH, Chen H, Xie JJ, Xu YW. Secreted proteins encoded by super enhancer-driven genes could be promising biomarkers for early detection of esophageal squamous cell carcinoma. Biomed J 2024; 47:100662. [PMID: 37774793 PMCID: PMC11340493 DOI: 10.1016/j.bj.2023.100662] [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: 04/22/2023] [Revised: 08/25/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Early detection of cancer remains an unmet need in clinical practice, and high diagnostic sensitivity and specificity biomarkers are urgently required. Here, we attempted to identify secreted proteins encoded by super-enhancer (SE)-driven genes as diagnostic biomarkers for esophageal squamous cell carcinoma (ESCC). METHODS We conducted an integrative analysis of multiple data sets including ChIP-seq data, secretome data, CCLE data and GEO data to screen secreted proteins encoded by SE-driven genes. Using ELISA, we further identified up-regulated secreted proteins through a small size of clinical samples and verified in a multi-centre validation stage (345 in test cohort and 231 in validation cohort). Receiver operating characteristic curves were used to calculate diagnostic accuracy. Artificial intelligence (AI) method named gradient boosting machine (GBM) were applied for model construction to enhance diagnostic accuracy. RESULTS Serum EFNA1 and MMP13 were identified, and showed significantly higher levels in ESCC patients compared to normal controls. An integrated Five-Biomarker Panel (iFBPanel) established by combining EFNA1, MMP13, carcino-embryonic antigen, Cyfra21-1 and squmaous cell carcinoma antigen had AUCs of 0.881 and 0.880 for ESCC in test and validation cohorts, respectively. Importantly, the iFBPanel also exhibited good performance in detecting early-stage ESCC patients (0.872 and 0.864). Furthermore, the iFBPanel was further empowered by AI technology which showed excellent diagnostic performance in early-stage ESCC (0.927 and 0.907). CONCLUSIONS Our study suggested that serum EFNA1 and MMP13 could potentially assist ESCC detection, and provided an easy-to-use detection model that might help the diagnosis of early-stage ESCC.
Collapse
Affiliation(s)
- Ling-Yu Chu
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Fang-Cai Wu
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shanto, China; Guangdong Esophageal Cancer Institute, Cancer Hospital of Shantou University Medical College, Shanto, China; Esophageal Cancer Prevention and Control Research Centre, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Chao-Qun Hong
- Department of Oncological Laboratory Research, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Li-Sheng Huang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shanto, China
| | - Hai-Ying Zou
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China; Guangdong Esophageal Cancer Institute, Cancer Hospital of Shantou University Medical College, Shanto, China
| | - Hao Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Centre, Guangzhou, China.
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China.
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China; Guangdong Esophageal Cancer Institute, Cancer Hospital of Shantou University Medical College, Shanto, China; Esophageal Cancer Prevention and Control Research Centre, Cancer Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
23
|
Zhang H, Jin T, Peng Y, Luan S, Li X, Xiao X, Yuan Y. Association between plasma circulating tumor DNA and the prognosis of esophageal cancer patients: a meta-analysis. Int J Surg 2024; 110:4370-4381. [PMID: 38526514 PMCID: PMC11254190 DOI: 10.1097/js9.0000000000001373] [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: 12/27/2023] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND The application of liquid biopsy analysis utilizing circulating tumor DNA (ctDNA) has gained prominence as a biomarker in specific cancer types. Nevertheless, the correlation between ctDNA and the prognostic outcomes of patients with esophageal cancer (EC) remains a subject of controversy. This meta-analysis aims to assess the correlation between ctDNA and the prognosis of EC patients. METHODS The authors systematically explored Embase, PubMed, and the Cochrane Database to identify studies reporting on the prognostic value of ctDNA in EC patients before November 2023. The primary outcome involved the determine of associations between ctDNA with overall survival (OS), disease-free survival (DFS)/recurrence-free survival (RFS), as well asprogression-free survival (PFS) among EC patients. Secondary outcomes encompassed a detailed subgroup analysis in the setting of EC, including parameters such as detection time, histological subtypes, treatment modalities, regions, anatomic locations, and detection methods. Publication bias was assessed utilizing Begg's test, Egger's test, and funnel plots. A sensitivity analysis was conducted by systematically excluding individual studies to evaluate the stability of the results. RESULTS A total of 1203 studies were initially screened, from which 13 studies underwent further analysis, encompassing 604 patients diagnosed with EC. The comprehensive pooled analysis indicated a significant association between the detection of ctDNA and poor OS (HR: 3.65; 95% CI: 1.97-6.75, P <0.001), DFS/RFS (HR: 6.08; 95% CI: 1.21-30.50, P <0.001), and PFS (HR: 2.84; 95% CI: 1.94-4.16, P <0.001). Subgroup analysis showed that ctDNA remained a consistent negative predictor of OS when stratified by different detection time, histological subtypes, regions, anatomic locations, and detection methods. Furthermore, subgroup analysis stratified by regions and study types demonstrated an association between ctDNA detection and poor PFS in EC patients. CONCLUSION Our results indicate plasma ctDNA may serve as robust prognostic markers for OS, DFS/RFS, and PFS among EC patients. This finding suggests that plasma ctDNA could offer a highly effective approach for risk stratification and personalized medicine.
Collapse
Affiliation(s)
- Haowen Zhang
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University
| | - Tao Jin
- Gastric Cancer Center, West China Hospital, Sichuan University, People’s Republic of China; Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, People’s Republic of China
| | - Yuhao Peng
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University
| | - Siyuan Luan
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University
| | - Xiaokun Li
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University
| | - Xin Xiao
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University
| | - Yong Yuan
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University
| |
Collapse
|
24
|
Zhou S, Chen Z, Jiao Y, Cheng Z, Gao Y, Wang T, Xin L, Wan R, Wang L. Development of esophagogastroduodenoscopy in China: results from the national census in 2013 and 2020. Front Oncol 2024; 14:1366706. [PMID: 38912062 PMCID: PMC11190165 DOI: 10.3389/fonc.2024.1366706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Background Given the significant burden of upper digestive diseases, there has been a substantial increase in the utilization of esophagogastroduodenoscopy (EGD) in China from 2012 to 2019. The objective of this study is to investigate the development, practice, and factors influencing the widespread use of EGD during this period. Methods Two national censuses were conducted among all hospitals in mainland China that perform gastrointestinal endoscopy. These censuses aimed to extract information on the infrastructure, volume, and quality of EGD. The analysis of potential factors influencing EGD practice was based on real-world data from open access sources. Results From 2012 to 2019, the number of hospitals performing EGD in mainland China increased from 1,518 to 2,265 (1.49-fold) in tertiary hospitals and from 3,633 to 4,097 (1.12-fold) in secondary hospitals, respectively. The national utilization rate of EGD also increased from 1,643.53 to 2,018.06 per 100,000 inhabitants, indicating a 1.23-fold increase. Regions with more endoscopists per 100,000 inhabitants (OR 9.61, P<0.001), more tertiary hospitals performing EGD per million inhabitants (OR 2.43, P<0.001), higher incidence of esophageal and gastric cancer (OR 2.09, P=0 016), and higher number of hospitals performing EGD per million inhabitants (OR 1.77, P=0.01) tended to provided more numerous and qualitied EGD. And hospital grading, regional GDP, incidence of esophageal and gastric cancer and the volume of EGD were observed as the significantly relevant factors of malignant dictation rate (MDR) (P<0.05), but not the number and educational background of endoscopists. Conclusion Over the past seven years, China has made significant progress in EGD. However, challenges persist in terms of quality and inequality.
Collapse
Affiliation(s)
- Siwei Zhou
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- National Digestive Endoscopy Improvement System, Shanghai, China
| | - Zheran Chen
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- National Digestive Endoscopy Improvement System, Shanghai, China
| | - Yunfei Jiao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- National Digestive Endoscopy Improvement System, Shanghai, China
| | - Zhiyuan Cheng
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- National Digestive Endoscopy Improvement System, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- National Digestive Endoscopy Improvement System, Shanghai, China
| | - Tianjiao Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- National Digestive Endoscopy Improvement System, Shanghai, China
| | - Lei Xin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- National Digestive Endoscopy Improvement System, Shanghai, China
| | - Rong Wan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luowei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
- National Digestive Endoscopy Improvement System, Shanghai, China
| |
Collapse
|
25
|
Li Y, Liu B, Zhou X, Yang H, Han T, Hong Y, Wang C, Huang M, Yan S, Li S, Li J, Liu Y, Zhang E, Ni Y, Shen N, Chen W, Huang YS, Wu N. Genome-Scale Multimodal Analysis of Cell-Free DNA Whole-Methylome Sequencing for Noninvasive Esophageal Cancer Detection. JCO Precis Oncol 2024; 8:e2400111. [PMID: 38976830 DOI: 10.1200/po.24.00111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/19/2024] [Accepted: 05/22/2024] [Indexed: 07/10/2024] Open
Abstract
PURPOSE Simultaneous profiling of cell-free DNA (cfDNA) methylation and fragmentation features to improve the performance of cfDNA-based cancer detection is technically challenging. We developed a method to comprehensively analyze multimodal cfDNA genomic features for more sensitive esophageal squamous cell carcinoma (ESCC) detection. MATERIALS AND METHODS Enzymatic conversion-mediated whole-methylome sequencing was applied to plasma cfDNA samples extracted from 168 patients with ESCC and 251 noncancer controls. ESCC characteristic cfDNA methylation, fragmentation, and copy number signatures were analyzed both across the genome and at accessible cis-regulatory DNA elements. To distinguish ESCC from noncancer samples, a first-layer classifier was developed for each feature type, the prediction results of which were incorporated to construct the second-layer ensemble model. RESULTS ESCC plasma genome displayed global hypomethylation, altered fragmentation size, and chromosomal copy number alteration. Methylation and fragmentation changes at cancer tissue-specific accessible cis-regulatory DNA elements were also observed in ESCC plasma. By integrating multimodal genomic features for ESCC detection, the ensemble model showed improved performance over individual modalities. In the training cohort with a specificity of 99.2%, the detection sensitivity was 81.0% for all stages and 70.0% for stage 0-II. Consistent performance was observed in the test cohort with a specificity of 98.4%, an all-stage sensitivity of 79.8%, and a stage 0-II sensitivity of 69.0%. The performance of the classifier was associated with the disease stage, irrespective of clinical covariates. CONCLUSION This study comprehensively profiles the epigenomic landscape of ESCC plasma and provides a novel noninvasive and sensitive ESCC detection approach with genome-scale multimodal analysis.
Collapse
Affiliation(s)
- Yulong Li
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xuantong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hechuan Yang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Tiancheng Han
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yuanyuan Hong
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Ciran Wang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Miao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shaolei Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jingjing Li
- The Precision Medicine Centre, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yanfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Enli Zhang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yang Ni
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Ning Shen
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Weizhi Chen
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yu S Huang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Nan Wu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
26
|
Zhang H, Xiao X, Luan S, Li X, Sun S, Yuan Y. Impact of change in the Naples prognostic score after neoadjuvant chemoradiotherapy on survival in esophageal squamous cell carcinoma patients. Saudi Med J 2024; 45:481-489. [PMID: 38734428 PMCID: PMC11147560 DOI: 10.15537/smj.2024.45.5.20230908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
OBJECTIVES To assess the clinical relevance and prognostic value of changes in the Naples prognostic score (NPS) after neoadjuvant chemoradiotherapy (NACR) among esophageal squamous cell carcinoma (ESCC) patients. METHODS We studied 232 locally advanced ESCC patients who received NACR before undergoing esophagectomy retrospectively. Categorizing individuals into the elevated NPS group and the non-elevated NPS group based on the change in NPS after NACR (ΔNPS > 0 or ∆NPS ≤ 0), we examined and compared the clinicopathological characteristics, survival rates, and postoperative complications between these 2 groups (∆NPS = post-NACR NPS - pre-NACR NPS). RESULTS Results: Out of the 232 patients enrolled, 105 exhibited elevated NPS levels, while 127 showed non-elevated NPS levels. Survival analyses indicated inferior overall survival (OS) (p=0.024) and recurrence-free survival (RFS) (p=0.047) in the elevated NPS cohort compared to the non-elevated NPS cohort. Subsequent cox regression analyses identified the post-NACR change in NPS as an independent prognostic indicator for RFS (p=0.029) and OS (p=0.036). CONCLUSION Elevated NPS post-NACR emerged as a significant indicator of worse prognosis for locally advanced ESCC patients who underwent NACR. This finding has great potential to be useful for recognizing high-risk ESCC patients who received NACR before undergoing esophagectomy and making individualized subsequent therapeutic decisions in clinical practice.
Collapse
Affiliation(s)
- Haowen Zhang
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xin Xiao
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Siyuan Luan
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaokun Li
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Shangwei Sun
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yong Yuan
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
27
|
Zhang Y, Liu Y, Wu L, Chen T, Jiao H, Ruan Y, Zhou P, Zhang Y. Expression of SOX4 Significantly Predicts the Risk of Lymph Node Metastasis for Patients With Early-Stage Esophageal Squamous Cell Carcinoma. J Transl Med 2024; 104:102042. [PMID: 38431117 DOI: 10.1016/j.labinv.2024.102042] [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: 11/27/2023] [Revised: 02/12/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
Esophageal squamous cell carcinoma stands as a notably aggressive malignancy within the digestive system. In cases of early esophageal cancer without lymph node metastasis, endoscopic surgical resection offers a viable alternative, often resulting in improved patient quality of life. However, the paucity of methods to preoperatively ascertain lymph node involvement complicates surgical planning. SOX4 gene was previously found to be highly associated with invasive metastasis in our work through single-cell RNA sequencing on 5 paired tumor/peritumor tissues. This research included the collection of 124 tissue samples from 106 patients (106 tumor and 18 lymph node specimens). Samples were methodically arranged into a tissue microarray and treated with immunohistochemical staining. Statistical analysis was conducted to assess the relationship between them. In the univariate analysis, 3 factors were identified as statistically significant in relation to lymph node metastasis: T category (P = .014), vascular invasion (P < .001), and SOX4 intensity (P = .001). Additionally, when evaluating SOX4 intensity alongside other clinical indicators, SOX4 was shown to independently influence lymph node metastasis. Further, the multivariate analysis revealed that vascular invasion (P < .001) and SOX4 intensity (P = .003) were significantly associated with lymph node metastasis, exhibiting hazard ratios of 10.174 and 7.142, respectively. The results of our study indicate that both SOX4 expression and vascular invasion serve as predictors of lymph node metastasis in patients diagnosed with category T1 esophageal squamous cell carcinoma, underscoring the potential utility of SOX4 in prognostic evaluations.
Collapse
Affiliation(s)
- Yifei Zhang
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, China
| | - Yanbo Liu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, China
| | - Linfeng Wu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, China
| | - Tianyin Chen
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, China
| | - Heng Jiao
- Department of Thoracic Surgery, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanyuan Ruan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pinghong Zhou
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, China.
| | - Yiqun Zhang
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Shanghai, China.
| |
Collapse
|
28
|
Wang S, Li Z, Zhou Z, Kang M. Causal analysis of gastroesophageal reflux disease and esophageal cancer. Medicine (Baltimore) 2024; 103:e37433. [PMID: 38489737 PMCID: PMC10939529 DOI: 10.1097/md.0000000000037433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/01/2024] [Accepted: 02/08/2024] [Indexed: 03/17/2024] Open
Abstract
Patients with gastroesophageal reflux disease (GERD) are more likely to develop esophageal cancer (EC). However, a causal relationship between the 2 has been difficult to determine. Therefore, this study aimed to evaluate the impact of GERD on EC using the Mendelian randomization (MR) method. The causal association between GERD and EC was analyzed based on 2 publicly available genetic summary datasets for the GERD cohort (129,080 cases vs 473,524 controls) and the EC cohort (740 cases vs 372,016 controls). The causal inference was mainly evaluated by the inverse variance weighted MR. The MR-Egger regression, MR Pleiotropy Residual Sum and Outlier test, and leave-one-out test were used to confirm the sensitivity of the MR results. Possible interfering factors were excluded by multivariate MR (MVMR) analysis. We used 73 single nucleotide polymorphisms as instrumental variables. GERD was associated with increasing EC risk (odds ratio [OR], 1.001; 95% confidence interval, 1.001-1.002; P < .001), which was identified using the inverse variance weighted method. The sensitivity analysis also demonstrated similar results with the causal explanation, and major bias in genetic pleiotropy was not identified (intercept, 0.001; standard error, 0.001; P = .418). The multivariate MR analysis demonstrated the effect of GERD on EC even after excluding possible mediating factors (OR, 1.003; 95% confidence interval, 1.001-1.005; P = .012). This study confirmed that GERD has a causal effect on EC. Therefore, interventional measures are recommended to prevent EC.
Collapse
Affiliation(s)
- Shuangyue Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Tumor Radiation Therapy Clinical Medical Research Center, Nanning, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| | - Zhiru Li
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Tumor Radiation Therapy Clinical Medical Research Center, Nanning, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| | - Ziyan Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Tumor Radiation Therapy Clinical Medical Research Center, Nanning, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| | - Min Kang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Tumor Radiation Therapy Clinical Medical Research Center, Nanning, Guangxi, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| |
Collapse
|
29
|
Sun Y, Liu W, Su M, Zhang T, Li X, Liu W, Cai Y, Zhao D, Yang M, Zhu Z, Wang J, Yu J. Purine salvage-associated metabolites as biomarkers for early diagnosis of esophageal squamous cell carcinoma: a diagnostic model-based study. Cell Death Discov 2024; 10:139. [PMID: 38485739 PMCID: PMC10940714 DOI: 10.1038/s41420-024-01896-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) remains an important health concern in developing countries. Patients with advanced ESCC have a poor prognosis and survival rate, and achieving early diagnosis remains a challenge. Metabolic biomarkers are gradually gaining attention as early diagnostic biomarkers. Hence, this multicenter study comprehensively evaluated metabolism dysregulation in ESCC through an integrated research strategy to identify key metabolite biomarkers of ESCC. First, the metabolic profiles were examined in tissue and serum samples from the discovery cohort (n = 162; ESCC patients, n = 81; healthy volunteers, n = 81), and ESCC tissue-induced metabolite alterations were observed in the serum. Afterward, RNA sequencing of tissue samples (n = 46) was performed, followed by an integrated analysis of metabolomics and transcriptomics. The potential biomarkers for ESCC were further identified by censoring gene-metabolite regulatory networks. The diagnostic value of the identified biomarkers was validated in a validation cohort (n = 220), and the biological function was verified. A total of 457 dysregulated metabolites were identified in the serum, of which 36 were induced by tumor tissues. The integrated analyses revealed significant alterations in the purine salvage pathway, wherein the abundance of hypoxanthine/xanthine exhibited a positive correlation with HPRT1 expression and tumor size. A diagnostic model was developed using two purine salvage-associated metabolites. This model could accurately discriminate patients with ESCC from normal individuals, with an area under the curve (AUC) (95% confidence interval (CI): 0.680-0.843) of 0.765 in the external cohort. Hypoxanthine and HPRT1 exerted a synergistic effect in terms of promoting ESCC progression. These findings are anticipated to provide valuable support in developing novel diagnostic approaches for early ESCC and enhance our comprehension of the metabolic mechanisms underlying this disease.
Collapse
Affiliation(s)
- Yawen Sun
- Department of Medical Epidemiology and Biostatistics, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Wenjuan Liu
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Mu Su
- Berry Oncology Corporation, Beijing, 102206, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xia Li
- Department of Public Health, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong, 250013, China
| | - Wenbin Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yuping Cai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Deli Zhao
- Tumor Preventative and Therapeutic Base of Shandong Province, Feicheng People's Hospital, Feicheng, Shandong, 271600, China
| | - Ming Yang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Jinan, Shandong, 250117, China
| | - Zhengjiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Jialin Wang
- Department of Medical Epidemiology and Biostatistics, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
30
|
Arai T, Ono S, Takubo K. Squamous Neoplastic Precursor Lesions of the Esophagus. Gastroenterol Clin North Am 2024; 53:25-38. [PMID: 38280749 DOI: 10.1016/j.gtc.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2024]
Abstract
Clinicopathological and molecular studies have demonstrated that dysplasia is a precancerous and/or neoplastic lesion with malignant potential. Further, it is subclassified into two grades: high-grade and low-grade dysplasia. High-grade dysplasia is a clinically significant lesion requiring resection or ablation. Low-grade dysplasia has a much lower risk of carcinoma; thus, it should be followed by endoscopic surveillance. Because squamous dysplasia may progress to squamous cell carcinoma, periodic endoscopy is useful to detect the lesion in patients with risk factors. Squamous dysplasia is diagnosed histopathologically by evaluating both cytologic and structural changes.
Collapse
Affiliation(s)
- Tomio Arai
- Department of Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan.
| | - Satoshi Ono
- Department of Gastroenterology and Gastrointestinal Endoscopy, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Kaiyo Takubo
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
31
|
Gao M, Wu X, Jiao X, Hu Y, Wang Y, Zhuo N, Dong F, Wang Y, Wang F, Cao Y, Liu C, Li J, Shen L, Zhang H, Lu Z. Prognostic and predictive value of angiogenesis-associated serum proteins for immunotherapy in esophageal cancer. J Immunother Cancer 2024; 12:e006616. [PMID: 38302415 PMCID: PMC10836376 DOI: 10.1136/jitc-2022-006616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have significantly improved patient survival in multiple cancers. However, therapy response in esophageal cancer is limited to subgroups of patients and clinically useful predictive biomarkers are lacking. METHODS We collected a series of plasma samples from 91 patients with esophageal cancer before and after ICI treatment. The Olink Immuno-Oncology panel (92 proteins) with proximity extension assays was used to detect the dynamic changes in plasma and potential biomarkers associated with treatment outcomes. We screened all survival-related proteins and established a risk score model to better predict the prognosis and treatment response in patients with esophageal cancer immunotherapy. RESULTS We found that 47 out of 92 quantified proteins had significant changes in plasma levels during ICI treatment (p<0.050), and these changed proteins were involved in immune-related reactions, such as intercellular adhesion and T-cell activation. Notably, the baseline levels of three angiogenesis-related proteins (IL-8, TIE2, and HGF) were significantly associated with the survival outcomes of patients treated with ICIs (p<0.050). According to these prognostic proteins, we established an angiogenesis-related risk score, which could be a superior biomarker for ICI response prediction. In addition, antiangiogenic therapy combined with ICIs significantly improved overall survival compared with ICI monotherapy (p=0.044). CONCLUSIONS An angiogenesis-related risk score based on three proteins (IL-8, TIE2, and HGF) could predict ICI response and prognosis in patients with esophageal cancer, which warrants verification in the future. Our study highlights the potential application of combining ICIs and antiangiogenic therapy and supports Olink plasma protein sequencing as a liquid biopsy method for biomarker exploration.
Collapse
Affiliation(s)
- Mengting Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xueying Wu
- Biomedical Innovation Center, Beijing Shijitan Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing Shijitan Hospital Capital Medical University, Beijing, China
| | - Xi Jiao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ying Hu
- Biomedical Innovation Center, Beijing Shijitan Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing Shijitan Hospital Capital Medical University, Beijing, China
| | - Yanni Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Na Zhuo
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fengxiao Dong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yujiao Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Fengyuan Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yanshuo Cao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chang Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jian Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Henghui Zhang
- Biomedical Innovation Center, Beijing Shijitan Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing Shijitan Hospital Capital Medical University, Beijing, China
- Beijing Engineering Research Center of Immunocellular therapy, Beijing, China
| | - Zhihao Lu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
32
|
Sun W, Kou H, Fang Y, Xu F, Xu Z, Wang X, Yin R, Zhang Q, Jiang Q, Xu Y. FOXO3a-regulated arginine metabolic plasticity adaptively promotes esophageal cancer proliferation and metastasis. Oncogene 2024; 43:216-223. [PMID: 38049565 DOI: 10.1038/s41388-023-02906-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 12/06/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor with a poor prognosis due to a lack of early detection. Indeed, the mechanisms underlying ESCC progression remain unclear. Here, we discovered that abnormal arginine metabolism contributes to ESCC progression. Based on transcriptomic and metabolomic analyses, we found that argininosuccinate synthetase 1 (ASS1) and argininosuccinate lyase (ASL) levels were increased in primary tumor tissues but decreased in lymph-metastatic tumor tissues. Intriguingly, FOXO3a was inversely correlated with ASS1 and ASL in primary and metastatic tumor tissues, suggesting that FOXO3a dissimilarly regulates ASS1 and ASL at different stages of ESCC. Silencing ASS1/ASL inhibited primary tumor growth and promoted metastasis. Conversely, overexpression of ASS1/ASL or increased arginine supply promoted tumor proliferation but suppressed metastasis. In addition, FOXO3a activation inhibited primary tumor growth by repressing ASS1 and ASL transcription, whereas inactivation of FOXO3a impeded metastasis by releasing ASS1 and ASL transcription. Together, the finding sheds light on metastatic reprogramming in ESCC.
Collapse
Affiliation(s)
- Wenbo Sun
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
- Department of Thoracic Surgery, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hengyuan Kou
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China
| | - Yao Fang
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China
| | - Fan Xu
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Zhi Xu
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China
| | - Xiumei Wang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China
| | - Rong Yin
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Qin Zhang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China.
| | - Qin Jiang
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China.
| | - Yong Xu
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China.
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China.
| |
Collapse
|
33
|
Agarwal S, Bell MG, Dhaliwal L, Codipilly DC, Dierkhising RA, Lansing R, Gibbons EE, Leggett CL, Kisiel JB, Iyer PG. Population Based Time Trends in the Epidemiology and Mortality of Gastroesophageal Junction and Esophageal Adenocarcinoma. Dig Dis Sci 2024; 69:246-253. [PMID: 37914889 PMCID: PMC10926253 DOI: 10.1007/s10620-023-08126-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/02/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Limited data are available on the epidemiology of gastroesophageal junction adenocarcinoma (GEJAC), particularly in comparison to esophageal adenocarcinoma (EAC). With the advent of molecular non-endoscopic Barrett's esophagus (BE) detection tests which sample the esophagus and gastroesophageal junction, early detection of EAC and GEJAC has become a possibility and their epidemiology has gained importance. AIMS We sought to evaluate time trends in the epidemiology and survival of patients with EAC and GEJAC in a population-based cohort. METHODS EAC and GEJAC patients from 1976 to 2019 were identified using ICD 9 and 10 diagnostic codes from the Rochester Epidemiology Project (REP). Clinical data and survival status were abstracted. Poisson regression was used to calculate incidence rate ratios (IRR). Survival analysis and Cox proportional models were used to assess predictors of survival. RESULTS We included 443 patients (287 EAC,156 GEJAC). The incidence of EAC and GEJAC during 1976-2019 was 1.40 (CI 1.1-1.74) and 0.83 (CI 0.61-1.11) per 100,000 people, respectively. There was an increase in the incidence of EAC (IRR = 2.45, p = 0.011) and GEJAC (IRR = 3.17, p = 0.08) from 2000 to 2004 compared to 1995-1999, plateauing in later time periods. Most patients had associated BE and presented at advanced stages, leading to high 5-year mortality rates (66% in EAC and 59% in GEJAC). Age and stage at diagnosis were predictors of mortality. CONCLUSION The rising incidence of EAC/GEJAC appears to have plateaued somewhat in the last decade. However, both cancers present at advanced stages with persistently poor survival, underscoring the need for early detection.
Collapse
Affiliation(s)
- Siddharth Agarwal
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Matthew G Bell
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lovekirat Dhaliwal
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - D Chamil Codipilly
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Ross A Dierkhising
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Ramona Lansing
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Erin E Gibbons
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Cadman L Leggett
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - John B Kisiel
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA
| | - Prasad G Iyer
- Division of Gastroenterology and Hepatology, Barrett's Esophagus Unit, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
34
|
Yuan XL, Liu W, Lin YX, Deng QY, Gao YP, Wan L, Zhang B, Zhang T, Zhang WH, Bi XG, Yang GD, Zhu BH, Zhang F, Qin XB, Pan F, Zeng XH, Chaudhry H, Pang MY, Yang J, Zhang JY, Hu B. Effect of an artificial intelligence-assisted system on endoscopic diagnosis of superficial oesophageal squamous cell carcinoma and precancerous lesions: a multicentre, tandem, double-blind, randomised controlled trial. Lancet Gastroenterol Hepatol 2024; 9:34-44. [PMID: 37952555 DOI: 10.1016/s2468-1253(23)00276-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Despite the usefulness of white light endoscopy (WLE) and non-magnified narrow-band imaging (NBI) for screening for superficial oesophageal squamous cell carcinoma and precancerous lesions, these lesions might be missed due to their subtle features and interpretation variations among endoscopists. Our team has developed an artificial intelligence (AI) system to detect superficial oesophageal squamous cell carcinoma and precancerous lesions using WLE and non-magnified NBI. We aimed to evaluate the auxiliary diagnostic performance of the AI system in a real clinical setting. METHODS We did a multicentre, tandem, double-blind, randomised controlled trial at 12 hospitals in China. Eligible patients were aged 18 years or older and underwent sedated upper gastrointestinal endoscopy for screening, investigation of gastrointestinal symptoms, or surveillance. Patients were randomly assigned (1:1) to either the AI-first group or the routine-first group using a computerised random number generator. Patients, pathologists, and statistical analysts were masked to group assignment, whereas endoscopists and research assistants were not. The same endoscopist at each centre did tandem upper gastrointestinal endoscopy for each eligible patient on the same day. In the AI-first group, the endoscopist did the first examination with the assistance of the AI system and the second examination without it. In the routine-first group, the order of examinations was reversed. The primary outcome was the miss rate of superficial oesophageal squamous cell carcinoma and precancerous lesions, calculated on a per-lesion and per-patient basis. All analyses were done on a per-protocol basis. This trial is registered with the Chinese Clinical Trial Registry (ChiCTR2100052116) and is completed. FINDINGS Between Oct 19, 2021, and June 8, 2022, 5934 patients were randomly assigned to the AI-first group and 5912 to the routine-first group, of whom 5865 and 5850 were eligible for analysis. Per-lesion miss rates were 1·7% (2/118; 95% CI 0·0-4·0) in the AI-first group versus 6·7% (6/90; 1·5-11·8) in the routine-first group (risk ratio 0·25, 95% CI 0·06-1·08; p=0·079). Per-patient miss rates were 1·9% (2/106; 0·0-4·5) in AI-first group versus 5·1% (4/79; 0·2-9·9) in the routine-first group (0·37, 0·08-1·71; p=0·40). Bleeding after biopsy of oesophageal lesions was observed in 13 (0·2%) patients in the AI-first group and 11 (0·2%) patients in the routine-first group. No serious adverse events were reported by patients in either group. INTERPRETATION The observed effect of AI-assisted endoscopy on the per-lesion and per-patient miss rates of superficial oesophageal squamous cell carcinoma and precancerous lesions under WLE and non-magnified NBI was consistent with substantial benefit through to a neutral or small negative effect. The effectiveness and cost-benefit of this AI system in real-world clinical settings remain to be further assessed. FUNDING National Natural Science Foundation of China, 1·3·5 project for disciplines of excellence, West China Hospital, Sichuan University, and Chengdu Science and Technology Project. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Xiang-Lei Yuan
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Liu
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi-Xiu Lin
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Qian-Yi Deng
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan-Ping Gao
- Department of Gastroenterology, Meishan People's Hospital, Meishan, China
| | - Ling Wan
- Department of Gastroenterology, Shimian People's Hospital, Ya'an, China
| | - Bin Zhang
- Department of Gastroenterology, Nanbu People's Hospital, Nanchong, China
| | - Tao Zhang
- Department of Gastroenterology, Nanchong Central Hospital, Nanchong, China
| | - Wan-Hong Zhang
- Department of Gastroenterology, Cangxi People's Hospital, Guangyuan, China
| | - Xiao-Gang Bi
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, China
| | - Guo-Dong Yang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Bi-Hui Zhu
- Department of Gastroenterology, Zizhong People's Hospital, Neijiang, China
| | - Fan Zhang
- Department of Gastroenterology, The Third People's Hospital of Yunnan Province, Kunming, China
| | - Xiao-Bo Qin
- Department of Gastroenterology, The First Veterans Hospital of Sichuan Province, Chengdu, China
| | - Feng Pan
- Department of Gastroenterology, Huai'an First People's Hospital, Huai'an, China
| | - Xian-Hui Zeng
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Hunza Chaudhry
- Department of Internal Medicine, University of California San Francisco-Fresno, CA, USA
| | - Mao-Yin Pang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Juliana Yang
- Department of Gastroenterology and Hepatology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jing-Yu Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Bing Hu
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
35
|
Lu D, Wu X, Wu W, Wu S, Li H, Zhang Y, Yan X, Zhai J, Dong X, Feng S, Zhang X, Sun F, Wang S, Cai K. Plasma cell-free DNA 5-hydroxymethylcytosine and whole-genome sequencing signatures for early detection of esophageal cancer. Cell Death Dis 2023; 14:843. [PMID: 38114477 PMCID: PMC10730877 DOI: 10.1038/s41419-023-06329-3] [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: 04/24/2023] [Revised: 11/05/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Esophageal cancer is a highly incidence and deadly disease with a poor prognosis, especially in developing countries. Owing to the lack of specific symptoms and early diagnostic biomarkers, most patients are diagnosed with advanced disease, leading to a 5-year survival rate of less than 15%. Early (n = 50) and middle-advanced (n = 50) esophageal squamous cell carcinoma (ESCC) patients, as well as 71 healthy individuals, underwent 5-hydroxymethylcytosine (5hmC) sequencing on their plasma cell-free DNA (cfDNA). A Northern Chinese cohort of cfDNA 5hmC dataset of 150 ESCC patients and 183 healthy individuals were downloaded for validation. A diagnostic model was developed using cfDNA 5hmC signatures and then improved by low-pass whole genome sequencing (WGS) features of cfDNA. Conserved cfDNA 5hmC modification motifs were observed in the two independent ESCC cohorts. The diagnostic model with 5hmC features achieved an AUC of 0.810 and 0.862 in the Southern and Northern cohorts, respectively, with sensitivities of 69.3-74.3% and specificities of 82.4-90.7%. The performance was well maintained in Stage I to Stage IV, with accuracy of 70-100%, but low in Stage 0, 33.3%. Low-pass WGS of cfDNA improved the AUC to 0.934 with a sensitivity of 82.4%, a specificity of 88.2%, and an accuracy of 84.3%, particularly significantly in Stage 0, with an accuracy up to 80%. 5hmC and WGS could efficiently differentiate very early ESCC from healthy individuals. These findings imply a non-invasive and convenient method for ESCC detection when clinical treatments are available and may eventually prolong survival.
Collapse
Affiliation(s)
- Di Lu
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xuanzhen Wu
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wendy Wu
- Berry Oncology Corporation, Beijing, 100102, China
| | - Shuangxiu Wu
- Berry Oncology Corporation, Beijing, 100102, China
| | - Hui Li
- Berry Oncology Corporation, Beijing, 100102, China
| | - Yuhong Zhang
- Berry Oncology Corporation, Beijing, 100102, China
| | - Xuebin Yan
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jianxue Zhai
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoying Dong
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Siyang Feng
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | | | - Fuming Sun
- Berry Oncology Corporation, Beijing, 100102, China
| | - Shaobo Wang
- Berry Oncology Corporation, Beijing, 100102, China
| | - Kaican Cai
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
36
|
Chang J, Zhao X, Wang Y, Liu T, Zhong C, Lao Y, Zhang S, Liao H, Bai F, Lin D, Wu C. Genomic alterations driving precancerous to cancerous lesions in esophageal cancer development. Cancer Cell 2023; 41:2038-2050.e5. [PMID: 38039962 DOI: 10.1016/j.ccell.2023.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/26/2023] [Accepted: 11/04/2023] [Indexed: 12/03/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) develops through a series of increasingly abnormal precancerous lesions. Previous studies have revealed the striking differences between normal esophageal epithelium and ESCC in copy number alterations (CNAs) and mutations in genes driving clonal expansion. However, due to limited data on early precancerous lesions, the timing of these transitions and which among them are prerequisites for malignant transformation remained unclear. Here, we analyze 1,275 micro-biopsies from normal esophagus, early and late precancerous lesions, and esophageal cancers to decipher the genomic alterations at each stage. We show that the frequency of TP53 biallelic inactivation increases dramatically in early precancerous lesion stage while CNAs and APOBEC mutagenesis substantially increase at late stages. TP53 biallelic loss is the prerequisite for the development of CNAs of genes in cell cycle, DNA repair, and apoptosis pathways, suggesting it might be one of the earliest steps initiating malignant transformation.
Collapse
Affiliation(s)
- Jiang Chang
- Department of Health Toxicology, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xuan Zhao
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Yichen Wang
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Hinxton, UK
| | - Tianyuan Liu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Ce Zhong
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Yueqiong Lao
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Han Liao
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing 100871, China; Center for Translational Cancer Research, Peking University First Hospital, Beijing 100034, China.
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100021, China; Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China; CAMS Oxford Institute, Chinese Academy of Medical Sciences, Beijing 100006, China.
| |
Collapse
|
37
|
Sun G, Chen H, Xia J, Li T, Ye H, Li J, Zhang X, Cheng Y, Wang K, Shi J, Wang P. Diagnostic performance of anti-MAGEA family protein autoantibodies in esophageal squamous cell carcinoma. Int Immunopharmacol 2023; 125:111041. [PMID: 37866309 DOI: 10.1016/j.intimp.2023.111041] [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: 08/03/2023] [Revised: 09/27/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023]
Abstract
MAGEA family proteins are immunogenic and can produce corresponding autoantibodies, and we aim to evaluate the diagnostic value of anti-MAGEA family protein autoantibodies in esophageal squamous cell carcinoma (ESCC). Protein chip was used to detect the expression level of anti-MAGEA autoantibodies (IgG and IgM) in 20 mixed serum samples. Enzyme linked immunosorbent assay was adopted to determine the expression level of autoantibodies in 1019 serum samples (423 ESCC, 423 healthy control (HC), 173 benign esophageal disease (BED)), and stepwise logistic regression analysis was used for developing a diagnostic model. Eight anti-MAGEA autoantibodies were screened out based on the protein chip. The levels of 7 autoantibodies (MAGEA1-IgG, MAGEA3-IgG, MAGEA3-IgM, MAGEA4-IgG, MAGEA6-IgG, MAGEA10-IgG, MAGEA12-IgG) in ESCC were significantly higher than that in HC, and the levels of anti-MAGEA1 IgG, anti-MAGEA3-IgG, anti-MAGEA4-IgG, anti-MAGEA10-IgG and anti-MAGEA12-IgG autoantibodies in ESCC group were significantly higher than those in BED group. The area under curve (AUC), sensitivity and specificity of the logistic regression model (MAGEA1-IgG, MAGEA4-IgG, MAGEA6-IgG, MAGEA12-IgG) in the training set and the validation set were 0.725 and 0.698, 55.2% and 51.8%, 80.4% and 84.5%, respectively, in distinguishing ESCC and HC. The model also could distinguish between ESCC and BED, with the AUC of 0.743, sensitivity of 55.4% and specificity of 89.0%. The positive rate of the model combined with cytokeratin 19 fragment to diagnose ESCC reached 78.0%. The study identified anti-MAGEA autoantibodies with potential diagnostic value for ESCC, which may provide new promising for the detection of the disease.
Collapse
Affiliation(s)
- Guiying Sun
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Huili Chen
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Junfen Xia
- Office of Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Tiandong Li
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hua Ye
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jiaxin Li
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Xiaoyue Zhang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yifan Cheng
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Keyan Wang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jianxiang Shi
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Peng Wang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| |
Collapse
|
38
|
Zhu J, Liang X, Chen S, Qin Y, Shen D, Yang X. Endoscopic therapy versus esophagectomy for T1bN0M0 esophageal cancer: A population-based study using propensity score matching. Heliyon 2023; 9:e22189. [PMID: 38045191 PMCID: PMC10692814 DOI: 10.1016/j.heliyon.2023.e22189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023] Open
Abstract
Background Endoscopic therapy is an optional strategy for the treatment of esophageal cancer (EC) under an early stage, especially stage T1a. However, its efficacy in the treatment of T1b EC has not been thoroughly assessed. We investigated the efficacy of esophagectomy, endoscopic therapy, as well as chemoradiotherapy in patients with T1bN0M0 EC. Methods The Surveillance, Epidemiology, and End Results database (SEER) was employed to identify patients diagnosed with T1bN0M0 EC. Patient demographics were compared among the endoscopic therapy, esophagectomy, and chemoradiotherapy groups. Our study employed Kaplan-Meier analysis and Cox regression model to evaluate patient outcomes and long-term survival rates. The overall survival (OS) and cancer-specific survival (CSS) rates were compared among patients with EC who underwent endoscopic therapy or esophagectomy, employing propensity score matching (PSM). Results A total of 820 patients diagnosed with T1bN0M0 EC were identified. The number of patients who received endoscopic therapy, esophagectomy, and chemoradiotherapy was 173, 556, and 91, respectively. Patients subjected to endoscopic therapy and esophagectomy had greatly longer OS and CSS than those who underwent chemoradiotherapy. Patients treated with esophagectomy had longer OS than endoscopic therapy patients, but there were no differences in CSS between the two groups. PSM generated 153 patient pairs among T1bN0M0 patients, demonstrating that both the esophagectomy and endoscopic therapy groups exhibited comparable OS and CSS rates. Conclusion Endoscopic therapy and esophagectomy were associated with a significant survival advantage compared with chemoradiotherapy in patients with T1bN0M0 EC. In contrast, after PSM, among the EC patients with stage T1bN0M0, OS and CSS did not differ after endoscopic therapy or esophagectomy. These results indicate that endoscopic therapy could be a viable alternative to esophagectomy in patients diagnosed with T1bN0M0 EC.
Collapse
Affiliation(s)
- Jiamin Zhu
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin, 214400, China
| | - Xiao Liang
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin, 214400, China
| | - Shusen Chen
- Department of Radiation Oncology, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, 226321, China
| | - Ya Qin
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin, 214400, China
| | - Dong Shen
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, 163 Shoushan Road, Jiangyin, 214400, China
| | - Xi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai Clinical Research Center for Radiation Oncology, Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| |
Collapse
|
39
|
Acharya R, Mahapatra A, Verma HK, Bhaskar LVKS. Unveiling Therapeutic Targets for Esophageal Cancer: A Comprehensive Review. Curr Oncol 2023; 30:9542-9568. [PMID: 37999111 PMCID: PMC10670555 DOI: 10.3390/curroncol30110691] [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: 09/12/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 11/25/2023] Open
Abstract
Esophageal cancer is a highly aggressive and deadly disease, ranking as the sixth leading cause of cancer-related deaths worldwide. Despite advances in treatment, the prognosis remains poor. A multidisciplinary approach is crucial for achieving complete remission, with treatment options varying based on disease stage. Surgical intervention and endoscopic treatment are used for localized cancer, while systemic treatments like chemoradiotherapy and targeted drug therapy play a crucial role. Molecular markers such as HER2 and EGFR can be targeted with drugs like trastuzumab and cetuximab, and immunotherapy drugs like pembrolizumab and nivolumab show promise by targeting immune checkpoint proteins. Epigenetic modifications offer new avenues for targeted therapy. Treatment selection depends on factors like stage, tumor location, and patient health, with post-operative and rehabilitation care being essential. Early diagnosis, appropriate treatment, and supportive care are key to improving outcomes. Continued research is needed to develop effective targeted drugs with minimal side effects. This review serves as a valuable resource for clinicians and researchers dedicated to enhancing esophageal cancer treatment outcomes.
Collapse
Affiliation(s)
- Rakesh Acharya
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur 495009, India; (R.A.); (A.M.)
| | - Ananya Mahapatra
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur 495009, India; (R.A.); (A.M.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany;
| | - L. V. K. S. Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur 495009, India; (R.A.); (A.M.)
| |
Collapse
|
40
|
Cui R, Wang L, Lin L, Li J, Lu R, Liu S, Liu B, Gu Y, Zhang H, Shang Q, Chen L, Tian D. Deep Learning in Barrett's Esophagus Diagnosis: Current Status and Future Directions. Bioengineering (Basel) 2023; 10:1239. [PMID: 38002363 PMCID: PMC10669008 DOI: 10.3390/bioengineering10111239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/26/2023] Open
Abstract
Barrett's esophagus (BE) represents a pre-malignant condition characterized by abnormal cellular proliferation in the distal esophagus. A timely and accurate diagnosis of BE is imperative to prevent its progression to esophageal adenocarcinoma, a malignancy associated with a significantly reduced survival rate. In this digital age, deep learning (DL) has emerged as a powerful tool for medical image analysis and diagnostic applications, showcasing vast potential across various medical disciplines. In this comprehensive review, we meticulously assess 33 primary studies employing varied DL techniques, predominantly featuring convolutional neural networks (CNNs), for the diagnosis and understanding of BE. Our primary focus revolves around evaluating the current applications of DL in BE diagnosis, encompassing tasks such as image segmentation and classification, as well as their potential impact and implications in real-world clinical settings. While the applications of DL in BE diagnosis exhibit promising results, they are not without challenges, such as dataset issues and the "black box" nature of models. We discuss these challenges in the concluding section. Essentially, while DL holds tremendous potential to revolutionize BE diagnosis, addressing these challenges is paramount to harnessing its full capacity and ensuring its widespread application in clinical practice.
Collapse
Affiliation(s)
- Ruichen Cui
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| | - Lei Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
- West China School of Nursing, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China
| | - Lin Lin
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
- West China School of Nursing, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China
| | - Jie Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
- West China School of Nursing, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China
| | - Runda Lu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| | - Shixiang Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| | - Bowei Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| | - Yimin Gu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| | - Hanlu Zhang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| | - Qixin Shang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| | - Dong Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu 610041, China; (R.C.); (L.W.); (L.L.); (J.L.); (R.L.); (S.L.); (B.L.); (Y.G.); (H.Z.); (Q.S.)
| |
Collapse
|
41
|
Xue M, Tong Y, Xiong Y, Yu C. Role of cancer-associated fibroblasts in the progression, therapeutic resistance and targeted therapy of oesophageal squamous cell carcinoma. Front Oncol 2023; 13:1257266. [PMID: 37927475 PMCID: PMC10623436 DOI: 10.3389/fonc.2023.1257266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/11/2023] [Indexed: 11/07/2023] Open
Abstract
Oesophageal squamous cell carcinoma (ESCC) is one of the most aggressive malignant tumours with high morbidity and mortality. Although surgery, radiotherapy and chemotherapy are common treatment options available for oesophageal cancer, the 5-year survival rate remains low after treatment. On the one hand, many oesophageal cancers are are discovered at an advanced stage and, on the other hand, treatment resistance is a major obstacle to treating locally advanced ESCC. Cancer-associated fibroblasts (CAFs), the main type of stromal cell in the tumour microenvironment, enhance tumour progression and treatment resistance and have emerged as a major focus of study on targeted therapy of oesophageal cancer.With the aim of providing potential, prospective targets for improving therapeutic efficacy, this review summarises the origin and activation of CAFs and their specific role in regulating tumour progression and treatment resistance in ESCC. We also emphasize the clinical potential and emerging trends of ESCC CAFs-targeted treatments.
Collapse
Affiliation(s)
| | | | | | - Changhua Yu
- Department of Radiotherapy, The Affiliated Huaian No.1 People’s Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
42
|
Shen R, Wang J, Wang R, Tian Y, Guo P, Shen S, Liu D, Zou T. The Role of Cancer in the Risk of Cardiovascular and All-Cause Mortality: A Nationwide Prospective Cohort Study. Int J Public Health 2023; 68:1606088. [PMID: 37927387 PMCID: PMC10620309 DOI: 10.3389/ijph.2023.1606088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Objectives: Evidence on cardiovascular-related and all-cause mortality risks in a wide range of cancer survivors is scarce but needed to inform prevention and management. Methods: We performed a nationwide prospective cohort study using information from the Continuous National Health and Nutrition Examination Survey (NHANES) in the United States and the linked mortality follow-up files, available for public access. A propensity score-matched analysis with a 1:1 ratio was conducted to reduce the baseline differences between participants with and without cancer. The relationship between cancer status and the cardiovascular-related and all-cause mortality risk was examined using weighted Cox proportional hazards regression. Independent stratification analysis and cancer-specific analyses were also performed. Results: The study sample included 44,342 participants, aged 20-85, interviewed between 1999 and 2018. Of these, 4,149 participants had cancer. All-cause death occurred in 6,655 participants, of whom 2,053 died from cardiovascular causes. Propensity-score matching identified 4,149 matched pairs of patients. A fully adjusted Cox proportional hazards regression showed that cancer was linked to an elevated risk of cardiovascular-related and all-cause mortality both before and after propensity score matching. Stratification analysis and cancer-specific analyses confirmed robustness of results. Conclusion: Our study confirmed that cancer was strongly linked to cardiovascular-related and all-cause mortality, even after adjusting for other factors that could impact a risk, including the American Heart Association (AHA)'s Life's Simple 7 cardiovascular health score, age, sex, ethnicity, marital status, income, and education level.
Collapse
Affiliation(s)
- Ruihuan Shen
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Jia Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Peking University Fifth School of Clinical Medicine, Beijing, China
| | - Rui Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Yuqing Tian
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Peiyao Guo
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Peking University Fifth School of Clinical Medicine, Beijing, China
| | - Shuhui Shen
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Donghao Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Peking University Fifth School of Clinical Medicine, Beijing, China
| | - Tong Zou
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
Su Z, Chen W, Cao X, Deng L, Zhang Y. Exploratory Study of a New Technique of Pixelated Chromoendoscopy in the Diagnosis of Early Esophageal Cancer. Surg Laparosc Endosc Percutan Tech 2023; 33:522-526. [PMID: 37585390 DOI: 10.1097/sle.0000000000001206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/24/2022] [Indexed: 08/18/2023]
Abstract
BACKGROUND Chromoendoscopy is an effective method for early screening of esophageal cancer, but diagnosis can depend on subjective judgment. The study aimed to explore a new technique of pixelated chromoendoscopy in the diagnosis of early esophageal cancer. PATIENTS AND METHODS The study included patients with symptoms of esophageal cancer who attended Jiangyin People's Hospital between January 2015 and July 2021. Chromoendoscopy was performed on each patient. The images then underwent digital analysis; the lesion area (the sensitive region) was pixelated by dividing it into the smallest image unit and the red, green, and blue color components. The diagnostic performance of pixelated chromoendoscopy was evaluated by calculating the area under the receiver operating characteristic. RESULTS The study finally enrolled 86 patients (aged 51.34 ± 5.82 y), including 54 males and 32 females. Pathologic diagnosis identified 54 cases in the cancer group and 32 cases in the non-cancer group. Traditional judgment had a diagnostic sensitivity of 70.73% and specificity was 75.00%. Pixelated chromoendoscopy sensitivity was 80.49%, and specificity was 83.33%. The area under the receiver operating characteristic was 0.814, at a cutoff value of 0.625, indicating a good prediction effect. CONCLUSIONS These results showed that pixelated chromoendoscopy might improve the rate of esophageal cancer diagnoses from early screening.
Collapse
Affiliation(s)
- Zhe Su
- Department of Gastroenterology
| | - Wei Chen
- Department of Oncology, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin
| | - Xiangming Cao
- Department of Digestive Disease, Dongtai Hospital Affiliated to Nantong Medical University, Yancheng, Jiangsu, China
| | - Lichun Deng
- Department of Digestive Disease, Dongtai Hospital Affiliated to Nantong Medical University, Yancheng, Jiangsu, China
| | | |
Collapse
|
44
|
Liu Y, Wu M. Deep learning in precision medicine and focus on glioma. Bioeng Transl Med 2023; 8:e10553. [PMID: 37693051 PMCID: PMC10486341 DOI: 10.1002/btm2.10553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 04/13/2023] [Accepted: 05/08/2023] [Indexed: 09/12/2023] Open
Abstract
Deep learning (DL) has been successfully applied to different fields for a range of tasks. In medicine, DL methods have been also used to improve the efficiency of disease diagnosis. In this review, we first summarize the history of the development of artificial intelligence models, demonstrate the features of the subtypes of machine learning and different DL networks, and then explore their application in the different fields of precision medicine, such as cardiology, gastroenterology, ophthalmology, dermatology, and oncology. By digging more information and extracting multilevel features from medical data, we found that DL helps doctors assess diseases automatically and monitor patients' physical health. In gliomas, research regarding application prospect of DL was mainly shown through magnetic resonance imaging and then by pathological slides. However, multi-omics data, such as whole exome sequence, RNA sequence, proteomics, and epigenomics, have not been covered thus far. In general, the quality and quantity of DL datasets still need further improvements, and more fruitful multi-omics characteristics will bring more comprehensive and accurate diagnosis in precision medicine and glioma.
Collapse
Affiliation(s)
- Yihao Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research InstituteCentral South UniversityChangshaHunanChina
| | - Minghua Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research InstituteCentral South UniversityChangshaHunanChina
| |
Collapse
|
45
|
Sijben J, Peters Y, Rainey L, Gashi M, Broeders MJ, Siersema PD. Professionals' views on the justification for esophageal adenocarcinoma screening: A systematic literature search and qualitative analysis. Prev Med Rep 2023; 34:102264. [PMID: 37273526 PMCID: PMC10236474 DOI: 10.1016/j.pmedr.2023.102264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/26/2023] [Accepted: 05/22/2023] [Indexed: 06/06/2023] Open
Abstract
Screening for early esophageal adenocarcinoma (EAC), including screening for its precursor Barrett's esophagus (BE), has the potential to reduce EAC-related mortality and morbidity. This literature review aimed to explore professionals' views on the justification for EAC screening. A systematic search of Ovid Medline, EMBASE, and PsycInfo, from January 1, 2000 to September 22, 2022, identified 5 original studies and 63 expert opinion articles reporting professionals' perspectives on EAC screening. Included articles were qualitatively analyzed using the framework method, which was deductively led by modernized screening principles. The analyses showed that many professionals are optimistic about technological advancements in BE detection and treatment. However, views on whether the societal burden of EAC merits screening were contradictory. In addition, knowledge of the long-term benefits and risks of EAC screening is still considered insufficient. There is no consensus on who to screen, how often to screen, which screening test to use, and how to manage non-dysplastic BE. Professionals further point out the need to develop technology that facilitates automated test sample processing and public education strategies that avoid causing disproportionately high cancer worry and social stigma. In conclusion, modernized screening principles are currently insufficiently fulfilled to justify widespread screening for EAC. Results from future clinical screening trials and risk prediction modeling studies may shift professionals' thoughts regarding justification for EAC screening.
Collapse
Affiliation(s)
- Jasmijn Sijben
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yonne Peters
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Linda Rainey
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mejdan Gashi
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mireille J.M. Broeders
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
- Dutch Expert Center for Screening, Nijmegen, The Netherlands
| | - Peter D. Siersema
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
46
|
Zhang H, Zhang J, Luan S, Liu Z, Li X, Liu B, Yuan Y. Unraveling the Complexity of Regulated Cell Death in Esophageal Cancer: from Underlying Mechanisms to Targeted Therapeutics. Int J Biol Sci 2023; 19:3831-3868. [PMID: 37564206 PMCID: PMC10411468 DOI: 10.7150/ijbs.85753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/13/2023] [Indexed: 08/12/2023] Open
Abstract
Esophageal cancer (EC) is the sixth most common and the seventh most deadly malignancy of the digestive tract, representing a major global health challenge. Despite the availability of multimodal therapeutic strategies, the existing EC treatments continue to yield unsatisfactory results due to their limited efficacy and severe side effects. Recently, knowledge of the subroutines and molecular mechanisms of regulated cell death (RCD) has progressed rapidly, enhancing the understanding of key pathways related to the occurrence, progression, and treatment of many types of tumors, including EC. In this context, the use of small-molecule compounds to target such RCD subroutines has emerged as a promising therapeutic strategy for patients with EC. Thus, in this review, we firstly discussed the risk factors and prevention of EC. We then outlined the established treatment regimens for patients with EC. Furthermore, we not only briefly summarized the mechanisms of five best studied subroutines of RCD related to EC, including apoptosis, ferroptosis, pyroptosis, necroptosis and autophagy, but also outlined the recent advances in the development of small-molecule compounds and long non-coding RNA (lncRNA) targeting the abovementioned RCD subroutines, which may serve as a new therapeutic strategy for patients with EC in the future.
Collapse
Affiliation(s)
- Haowen Zhang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, 518000, China
| | - Siyuan Luan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiying Liu
- School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, 518000, China
| | - Xiaokun Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
47
|
Chou CK, Nguyen HT, Wang YK, Chen TH, Wu IC, Huang CW, Wang HC. Preparing Well for Esophageal Endoscopic Detection Using a Hybrid Model and Transfer Learning. Cancers (Basel) 2023; 15:3783. [PMID: 37568599 PMCID: PMC10417640 DOI: 10.3390/cancers15153783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/17/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Early detection of esophageal cancer through endoscopic imaging is pivotal for effective treatment. However, the intricacies of endoscopic diagnosis, contingent on the physician's expertise, pose challenges. Esophageal cancer features often manifest ambiguously, leading to potential confusions with other inflammatory esophageal conditions, thereby complicating diagnostic accuracy. In recent times, computer-aided diagnosis has emerged as a promising solution in medical imaging, particularly within the domain of endoscopy. Nonetheless, contemporary AI-based diagnostic models heavily rely on voluminous data sources, limiting their applicability, especially in scenarios with scarce datasets. To address this limitation, our study introduces novel data training strategies based on transfer learning, tailored to optimize performance with limited data. Additionally, we propose a hybrid model integrating EfficientNet and Vision Transformer networks to enhance prediction accuracy. Conducting rigorous evaluations on a carefully curated dataset comprising 1002 endoscopic images (comprising 650 white-light images and 352 narrow-band images), our model achieved exceptional outcomes. Our combined model achieved an accuracy of 96.32%, precision of 96.44%, recall of 95.70%, and f1-score of 96.04%, surpassing state-of-the-art models and individual components, substantiating its potential for precise medical image classification. The AI-based medical image prediction platform presents several advantageous characteristics, encompassing superior prediction accuracy, a compact model size, and adaptability to low-data scenarios. This research heralds a significant stride in the advancement of computer-aided endoscopic imaging for improved esophageal cancer diagnosis.
Collapse
Affiliation(s)
- Chu-Kuang Chou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan;
- Obesity Center, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
| | - Hong-Thai Nguyen
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi 62102, Taiwan;
| | - Yao-Kuang Wang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City 80756, Taiwan;
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 80756, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 80756, Taiwan
| | - Tsung-Hsien Chen
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan;
| | - I-Chen Wu
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 80756, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 80756, Taiwan
| | - Chien-Wei Huang
- Department of Gastroenterology, Kaohsiung Armed Forces General Hospital, Kaohsiung City 80284, Taiwan
- Department of Nursing, Tajen University, 20, Weixin Rd., Yanpu Township, Pingtung 90741, Taiwan
| | - Hsiang-Chen Wang
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi 62102, Taiwan;
- Hitspectra Intelligent Technology Co., Ltd., Kaohsiung City 80661, Taiwan
| |
Collapse
|
48
|
Han Y, Chen G, Liu S, Zhou G, Xu X, Zhang H, Li Z, Wu C, Liu Y, Fang K, Chen G. MUC13 promotes the development of esophageal cancer by upregulating the expression of O-glycan process-related molecules. Discov Oncol 2023; 14:123. [PMID: 37395858 PMCID: PMC10317945 DOI: 10.1007/s12672-023-00713-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Esophageal cancer is one of the most common malignant tumors in the world, which is characterized by poor prognosis, aggressiveness, and poor survival. Mucin 13 (MUC13) is a member of the membrane-bound mucin and located on chromosome 3q21.2 and consists of α and β subunits. It has been found that MUC13 is overexpressed in a variety of tumor cells and acts a vital role in the invasiveness and malignant progression of several types of tumors. However, the role and regulatory mechanism of MUC13 in the progression of esophageal cancer remain unclear. METHODS The expression level of MUC13 was detected in 15 esophageal cancer tissues and 15 pairs of adjacent nontumor tissues by immunohistochemistry (IHC). In addition, the expression of MUC13 mRNA level in human esophageal cancer cell lines (EC9706 and ECA109 and TE-1) was measured by qRT-PCR. In vitro, after silencing MUC13 with lentiviral interference technology, CCK8 assay, clone formation assay, and flow cytometry were applied to investigate the proliferation activity, clone formation ability and anti-apoptosis ability of EC9706 and ECA109 cells. The tumor xenograft growth assay was used to confirm the influence of MUC13 knockdown on the growth of esophageal tumors in vivo. The qRT-PCR assay and western blot experiments were taken to study the mechanism of MUC13 regulating the proproliferation and antiapoptotic of esophageal cancer. RESULTS The results showed that MUC13 was overexpressed in esophageal cancer tissues and cell lines (EC9706 and ECA109 and TE-1), especially in EC9706 and ECA109 cells, but low expressed in human esophageal epithelial cell line (HEEC). Next, silencing MUC13 inhibits proliferation, blocks cell cycle progression, and promotes cell apoptosis in vitro, and restrains the growth of esophageal cancer tissues in vivo. Finally, MUC13 affects the proproliferation and antiapoptotic by regulating the expression of GLANT14, MUC3A, MUC1, MUC12, and MUC4 that closely related to O-glycan process. CONCLUSIONS This study proved that MUC13 is an important molecule that regulates the O-glycan process and then affects the progress of esophageal cancer. MUC13 may be a novel therapeutic target for patients with esophageal cancer.
Collapse
Affiliation(s)
- Yi Han
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221002, China
| | - Gang Chen
- Department of Plastic Surgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Shiyu Liu
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221002, China
| | | | - Xinxin Xu
- Xuzhou Medical University, Xuzhou, 221002, China
| | - Haihan Zhang
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221002, China
| | - Zhentao Li
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221002, China
| | - Chuannan Wu
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221002, China
| | - Yulan Liu
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Kai Fang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Guangxia Chen
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
49
|
Sun D, Yi J, Gong L, Wu Y, Liu X. Prognostic analysis and nomogram establishment in patients with early esophageal cancer receiving endoscopic therapy: a population-based study. Therap Adv Gastroenterol 2023; 16:17562848231170470. [PMID: 37163166 PMCID: PMC10164252 DOI: 10.1177/17562848231170470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 04/01/2023] [Indexed: 05/11/2023] Open
Abstract
Background The growing numbers of early esophageal cancer (EEC) have increased the demand for endoscopic therapy. Objectives To clarify the influential factors for the prognosis of patients with EEC receiving endoscopic surgery, and to construct a nomogram to evaluate the prognostic value of endoscopic therapy. Design Prognostic analysis study. Methods Clinical data of EEC patients who received endoscopic therapy between 2004 and 2015 were collected from the Surveillance, Epidemiology, and End Results database and used to construct the nomogram. The prognosis was analyzed by R language; the nomogram was constructed by Cox survival analysis; and the accuracy of the nomogram was verified by C index and the receiver operating characteristic (ROC) and calibration curves. X-Tile software was used to stratify the risk of patients. Results Our study constructed the nomogram of the prognosis of patients with EEC treated by endoscopic surgery, including 1118 patients and 5 independent prognostic factors of esophageal cancer-specific survival. The C index and the area under the ROC curve (AUC) of the training and verification cohorts were all >0.75. The calibration curve also reflected the good consistency of the model in predicting survival. Significant difference in the risk of patients from different stratifications with the same T staging existed, and the model had a better C index than that of the T staging. Conclusion Our study reports potential influential factors affecting the prognosis of EEC patients who received endoscopic therapy and establishes a reliable nomogram to predict the risk and prognosis, which has certain advantages compared with traditional TNM staging system.
Collapse
Affiliation(s)
- Danping Sun
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Yi
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, China
| | - Lingqi Gong
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Yu Wu
- Department of Gastroenterology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, Hunan 410000 China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, China
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, Hunan 410000, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
50
|
Gao Y, Xin L, Lin H, Yao B, Zhang T, Zhou AJ, Huang S, Wang JH, Feng YD, Yao SH, Guo Y, Dang T, Meng XM, Yang ZZ, Jia WQ, Pang HF, Tian XJ, Deng B, Wang JP, Fan WC, Wang J, Shi LH, Yang GY, Sun C, Wang W, Zang JC, Li SY, Shi RH, Li ZS, Wang LW. Machine learning-based automated sponge cytology for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction: a nationwide, multicohort, prospective study. Lancet Gastroenterol Hepatol 2023; 8:432-445. [PMID: 36931287 DOI: 10.1016/s2468-1253(23)00004-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction have a dismal prognosis, and early detection is key to reduce mortality. However, early detection depends on upper gastrointestinal endoscopy, which is not feasible to implement at a population level. We aimed to develop and validate a fully automated machine learning-based prediction tool integrating a minimally invasive sponge cytology test and epidemiological risk factors for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction before endoscopy. METHODS For this multicohort prospective study, we enrolled participants aged 40-75 years undergoing upper gastrointestinal endoscopy screening at 39 tertiary or secondary hospitals in China for model training and testing, and included community-based screening participants for further validation. All participants underwent questionnaire surveys, sponge cytology testing, and endoscopy in a sequential manner. We trained machine learning models to predict a composite outcome of high-grade lesions, defined as histology-confirmed high-grade intraepithelial neoplasia and carcinoma of the oesophagus and oesophagogastric junction. The predictive features included 105 cytological and 15 epidemiological features. Model performance was primarily measured with the area under the receiver operating characteristic curve (AUROC) and average precision. The performance measures for cytologists with AI assistance was also assessed. FINDINGS Between Jan 1, 2021, and June 30, 2022, 17 498 eligible participants were involved in model training and validation. In the testing set, the AUROC of the final model was 0·960 (95% CI 0·937 to 0·977) and the average precision was 0·482 (0·470 to 0·494). The model achieved similar performance to consensus of cytologists with AI assistance (AUROC 0·955 [95% CI 0·933 to 0·975]; p=0·749; difference 0·005, 95% CI, -0·011 to 0·020). If the model-defined moderate-risk and high-risk groups were referred for endoscopy, the sensitivity was 94·5% (95% CI 88·8 to 97·5), specificity was 91·9% (91·2 to 92·5), and the predictive positive value was 18·4% (15·6 to 21·6), and 90·3% of endoscopies could be avoided. Further validation in community-based screening showed that the AUROC of the model was 0·964 (95% CI 0·920 to 0·990), and 92·8% of endoscopies could be avoided after risk stratification. INTERPRETATION We developed a prediction tool with favourable performance for screening of oesophageal squamous cell carcinoma and adenocarcinoma of the oesophagogastric junction. This approach could prevent the need for endoscopy screening in many low-risk individuals and ensure resource optimisation by prioritising high-risk individuals. FUNDING Science and Technology Commission of Shanghai Municipality.
Collapse
Affiliation(s)
- Ye Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Lei Xin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Han Lin
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Bin Yao
- School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Tao Zhang
- Department of Gastroenterology, Nanchong Central Hospital, Nanchong, Sichuan Province, China
| | - Ai-Jun Zhou
- Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Shu Huang
- Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Jian-Hua Wang
- Department of Gastroenterology, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Ya-Dong Feng
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Sheng-Hua Yao
- Department of Gastroenterology, Yangzhong People's Hospital, Zhenjiang, Jiangsu Province, China
| | - Yan Guo
- Department of Gastroenterology, Yangzhong People's Hospital, Zhenjiang, Jiangsu Province, China
| | - Tong Dang
- Department of Digestive Diseases, Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Xian-Mei Meng
- Department of Digestive Diseases, Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Zeng-Zhou Yang
- Digestive Endoscopy Unit, Linzhou People's Hospital, Anyang, Henan Province, China
| | - Wan-Qi Jia
- Gastrointestinal Endoscopy Center, Nanyang Second People's Hospital, Nanyang, Henan Province, China
| | - Hui-Fang Pang
- Department of Gastroenterology, Digestive Endoscopy Unit, Tongliao City Hospital, Tongliao, Inner Mongolia, China
| | - Xiao-Juan Tian
- Department of Gastroenterology, Xixia County People's Hospital, Nanyang, Henan Province, China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Jun-Ping Wang
- Department of Gastroenterology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi Province, China
| | - Wen-Chuan Fan
- Department of Gastroenterology, Digestive Endoscopy Center, The People's Hospital of Yanting City, Mianyang, Sichuan Province, China
| | - Jun Wang
- Department of Gastroenterology, Jinhu County People's Hospital, Huaian, Jiangsu Province, China
| | - Li-Hong Shi
- Department of Gastroenterology, the Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Guan-Yu Yang
- School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu Province, China
| | - Chang Sun
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Wei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Jun-Cai Zang
- Harbor Scientific Instrument, Xiangtan, Hunan, China
| | - Song-Yang Li
- Harbor Scientific Instrument, Xiangtan, Hunan, China
| | - Rui-Hua Shi
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China
| | - Luo-Wei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; National Clinical Research Center for Digestive Diseases (Shanghai), Shanghai, China.
| |
Collapse
|