1
|
Li L, Lan Z, Qiao H, Meng X, Shi Z, Zhang W, Wang Y, Sun Z, Cui Q, Wang L, Zhou S, Hu F, Zhang D, Dai Y, Chen H, Geng Y. Design of NanoBiT-Nanobody-based FGL1 biosensors for early assisted diagnosis of esophageal cancer. Biomaterials 2025; 320:123286. [PMID: 40138964 DOI: 10.1016/j.biomaterials.2025.123286] [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: 12/09/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Esophageal cancer (EC) is one of the most common causes of cancer-related mortality due in part to challenges in early diagnosis. Biomarker identification is crucial for improved early screening and treatment strategies for patients. Firstly, we employed serum proteomics techniques to screen for potential biomarkers in 15 early-stage EC patients and 5 healthy individuals. Among the differentially expressed proteins, FGL1 emerged as a promising candidate (AUC = 0.974) for early detection of EC. Subsequently, we developed NanoBiT-conjugated dual nanobodies (NBNB) sensors for robust and quantitative signal detection in fetal bovine serum (FBS) in 30 min or less, with a limit of detection (LoD) of 11.38 pM. In a case-control study recruiting 96 EC patients and 99 control samples, testing serum samples with the developed NBNB sensors revealed significantly elevated serum level of FGL1 in all-stage EC patients (AUC = 0.7880) and early-stage EC patients (AUC = 0.8286). Additionally, the combined diagnostic performance of FGL1 and CEA in EC samples is notably enhanced (AUC = 0.8847). These findings propose FGL1 as a novel and promising target for the early-stage EC diagnosis and treatment selection. Furthermore, we applied the assay to patients across six types of cancer, suggesting FGL1 as a potential pan-cancer marker. This study introduces a rapid, easy-to-use, cost-effective, reliable, universal, and high-throughput alternative to meet the growing demand for cancer biomarker testing in both academic and clinical settings.
Collapse
Affiliation(s)
- Lingyun Li
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhongyun Lan
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huarui Qiao
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiangjing Meng
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Ziyang Shi
- Queen Mary University of London Engineering School, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Wanting Zhang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi'ang Wang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zengchao Sun
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qianqian Cui
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lu Wang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Siyu Zhou
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fangzheng Hu
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Daizhou Zhang
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China.
| | - Yuanyuan Dai
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital of Chinese Academy of Medical Sciences Langfang Campus, Langfang, 065001, China.
| | - Hao Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Yong Geng
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
2
|
Iizuka A, Kanda M, Sato Y, Shimizu D, Umeda S, Tanaka H, Hattori N, Hayashi M, Tanaka C, Kodera Y. Association of NR0B1 with Malignant Phenotypes in Esophageal Squamous Cell Carcinoma Through Modulation of p53-Independent Cell-Cycle Regulation. Ann Surg Oncol 2025; 32:4464-4475. [PMID: 40080367 DOI: 10.1245/s10434-025-17109-y] [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: 10/01/2024] [Accepted: 02/17/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Transcriptome analysis of primary tumor tissues from esophageal squamous cell carcinoma (ESCC) patients with early postoperative distant metastasis identified nuclear receptor subfamily 0, group B, member 1 (NR0B1) as a novel gene associated with the malignant phenotypes of ESCC. This study aimed to elucidate the oncological functions of NR0B1 in ESCC and assess the significance of its tissue expression. METHODS We investigated the effects of NR0B1 knockdown on the proliferation, migration, and adhesion capacities, in vivo tumor growth, and intracellular signaling pathways of ESCC cell lines. The correlation between tissue NR0B1 expression at both the mRNA and protein levels and postoperative prognosis was analyzed by using two independent cohorts. RESULTS Silencing NR0B1 significantly inhibited the proliferation, migration, and adhesion capacities of ESCC cell lines and decreased tumor growth in mouse cell line derived xenograft models. Knockdown of NR0B1 results in the upregulation of cell cycle regulators p21 and p27, alongside the downregulation of TK1, cyclin E1, CDK2 and CDT1, in a manner independent of p53. Although elevated tissue NR0B1 expression did not show a significant association with TNM stage, it was identified as an independent prognostic factor at both the mRNA and protein levels across two distinct patient cohorts. CONCLUSIONS NR0B1 plays a critical role in the malignant phenotype of ESCC by modulating cell cycle regulators. Tissue NR0B1 expression may serve as a valuable biomarker for assessing prognostic risk in ESCC patients.
Collapse
Affiliation(s)
- Akimitsu Iizuka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Yusuke Sato
- Department of Thoracic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Haruyoshi Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
3
|
Crecca E, Di Giuseppe G, Camplone C, Vigiano Benedetti V, Melaiu O, Mezza T, Cencioni C, Spallotta F. The multifaceted role of agents counteracting metabolic syndrome: A new hope for gastrointestinal cancer therapy. Pharmacol Ther 2025; 270:108847. [PMID: 40216262 DOI: 10.1016/j.pharmthera.2025.108847] [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: 10/04/2024] [Revised: 01/27/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
Metabolic syndrome (MetS) is defined by the presence of at least three of five clinical parameters including abdominal obesity, insulin resistance, elevated triglycerides, reduced high-density lipoprotein (HDL) and hypertension. Major features describing MetS have been recognized risk factors for cancer onset, with an alarming impact on gastrointestinal (GI) tumors. Intriguingly, therapeutic administration of drugs to improve glycemic control and dyslipidemia (including metformin, statins) has been shown to have a preventive role in the development and in prognosis improvement of several cancer types. Overall, these observations highlight the key role of altered metabolism prevalently in cancer risk development and unveil anti-MetS agent repurposing potential beyond their conventional pharmacological action. The objective of this review is to summarize the current knowledge about the antitumor activity of anti-diabetic and anti-lipemic agents in GI cancer onset and progression. Here, pre-clinical evidence of their therapeutic potential and of their integration in novel compelling therapeutic strategies will be discussed. Possible clinical outcomes of these novel therapeutic combined protocols specifically dedicated to GI cancer patients will be put under the spotlight. In the future, these novel therapeutic options should be considered to improve conventional chemotherapy response and prognosis of this group of patients.
Collapse
Affiliation(s)
- Elena Crecca
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185 Rome, Italy
| | - Gianfranco Di Giuseppe
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy; Department of Translational Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Claudia Camplone
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | | | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Teresa Mezza
- Department of Translational Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy; Pancreas Unit, CEMAD Digestive Diseases Center, Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Chiara Cencioni
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185 Rome, Italy.
| | - Francesco Spallotta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
4
|
Zhang M, Zhang C, Zhou F, Yang R, Feng Y, Ji Y, Ren H, Ming L. LINC02154 Promotes Esophageal Squamous Cell Carcinoma Progression via the PI3K-AKT-mTOR Signaling Pathway by Interacting With IGF2BP2. Mol Carcinog 2025; 64:985-996. [PMID: 40099590 DOI: 10.1002/mc.23903] [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: 09/30/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
As important types of noncoding RNAs, long noncoding RNAs (lncRNAs) have been found to be involved in the progression of various cancers. Accumulating evidence indicates that LINC02154 plays a critical role in cancer progression, but the underlying mechanisms regulating esophageal squamous cell carcinoma (ESCC) remain unclear. Here, we found that LINC02154 is significantly upregulated in ESCC cell lines and ESCC tissues. LINC02154 knockdown significantly inhibited the proliferation and migration of ESCC cells in vitro and suppressed the progression of ESCC in vivo. Mechanistically, LINC02154 can bind to IGF2BP2 and activate the PI3K-AKT-mTOR signaling pathway. High expression of LINC02154 is positively correlated with poor prognosis in ESCC patients. In conclusion, LINC02154 functions as an oncogenic factor to facilitate ESCC progression through the IG2BP2-PI3K-AKT-mTOR pathway and has the potential to be a promising diagnostic marker and therapeutic target for ESCC patients.
Collapse
Affiliation(s)
- Mingyuan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Cai Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Fuyou Zhou
- Thoracic Department, Anyang Tumor Hospital, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, China
| | - Ruotong Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Yang Feng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Yangyang Ji
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Huijun Ren
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| | - Liang Ming
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Clinical Laboratory of Henan province, Zhengzhou, China
| |
Collapse
|
5
|
Jiang M, Yu H. Ginsenoside 20(S)-Rg3 Hinders Esophageal Squamous Cell Carcinoma Cells Malignant Behaviors by miR-210-3p/B4GALT5 Axis. Cell Biochem Biophys 2025; 83:1555-1563. [PMID: 39422791 DOI: 10.1007/s12013-024-01566-5] [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] [Accepted: 09/17/2024] [Indexed: 10/19/2024]
Abstract
Ginsenoside 20(S)-Rg3 (20(S)-Rg3) belongs to a natural chemical with an anti-tumor function, but its potential function and underlying mechanism in esophageal squamous cell carcinoma (ESCC) are unknown. Several reports have manifested that microRNA (miRNA) miR-210-3p functions as a tumor repressor in tumors, but its biofunction in ESCC remains obscure. Herein, the role and interaction of 20(S)-Rg3 and miR-210-3p in ESCC cells were investigated. We performed a series of functional experiments to validate that 20(S)-Rg3 notably restrained ESCC cell proliferation and migration while promoting cell apoptosis. Besides, miR-210-3p was found to be lowly expressed in ESCC cells. Overexpressing miR-210-3p suppressed the malignant behaviors of ESCC cells. More importantly, 20(S)-Rg3 could upregulate miR-210-3p expression in ESCC cells. MiR-210-3p knockdown offset the inhibitive impacts of 20(S)-Rg3 treatment on ESCC cell growth and migration. Furthermore, through luciferase reporter assay, beta-1,4-galactosyltransferase 5 (B4GALT5) was certified to be targeted by miR-210-3p. B4GALT5 upregulation neutralized the suppressive function of 20(S)-Rg3 on ESCC progression. Overall, 20(S)-Rg3 attenuated malignant behaviors of ESCC cells by modulating miR-210-3p/B4GALT5 axis, indicating 20(S)-Rg3 has therapeutic potential for ESCC.
Collapse
Affiliation(s)
- Min Jiang
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China.
| |
Collapse
|
6
|
Vanan AG, Vesal S, Seraj P, Ghezel MA, Eini P, Talebileili M, Asgari Z, Tahmasebi S, Hashemi M, Taheriazam A. DCLK1 in gastrointestinal cancer: A driver of tumor progression and a promising therapeutic target. Int J Cancer 2025; 156:2068-2086. [PMID: 40056091 DOI: 10.1002/ijc.35365] [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: 10/24/2024] [Revised: 01/12/2025] [Accepted: 01/29/2025] [Indexed: 04/05/2025]
Abstract
Cancers of the gastrointestinal (GI) tract, including colorectal, pancreatic, and hepatocellular carcinomas, represent a significant global health burden due to their high incidence and mortality rates. Doublecortin-like kinase 1 (DCLK1), initially identified for its role in neurogenesis, has emerged as a crucial player in GI cancer progression. This review comprehensively examines the multifaceted roles of DCLK1 in GI cancers, focusing on its structural isoforms, functions in normal and inflammatory states, and contributions to cancer progression and metastasis. DCLK1 is overexpressed in various GI cancers and is associated with poor prognosis, enhanced tumorigenic potential, and increased metastatic capacity. The review discusses the molecular mechanisms through which DCLK1 influences cancer stem cell maintenance, epithelial-mesenchymal transition (EMT), and cell survival pathways, as well as its interactions with key signaling pathways such as Notch, WNT/β-catenin, and NF-κB. The potential of DCLK1 as a therapeutic target is also explored, highlighting preclinical and early clinical efforts to inhibit its function using small molecule inhibitors or monoclonal antibodies. Despite significant advancements, further research is needed to fully elucidate DCLK1's role in GI cancers and to develop effective therapeutic strategies targeting this protein.
Collapse
Affiliation(s)
- Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheil Vesal
- Department of Molecular Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Parmida Seraj
- Department of Medicine, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | | | - Pooya Eini
- Toxicological Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Talebileili
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Zeynab Asgari
- Department of Immunology, School of Medicine Kerman University of Medical Sciences, Kerman, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Faculty of Advanced Science and Technology, Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Faculty of Medicine, Department of Orthopedics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
7
|
Cui Y, Deng Y, Wu Z, Sun X. To verify the biological characteristics of disulfidptosis associated gene ADORA2B in esophageal cancer. BMC Gastroenterol 2025; 25:382. [PMID: 40389870 PMCID: PMC12087064 DOI: 10.1186/s12876-025-03768-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/06/2025] [Indexed: 05/21/2025] Open
Abstract
Esophageal cancer is an aggressive malignant tumor. Statistics show that esophageal cancer has claimed the lives of approximately 300,000 people worldwide. Many patients are diagnosed as stage 3 or 4 when they visit a doctor; however, the prognosis may not be accurate because the disease's early signs are not always evident. The gene ADORA2B may be important in the diagnosis and prognosis of esophageal cancer, as adenosine (ADO) is implicated in the proliferation and spread of many malignancies. Through the use of bioinformatics analysis, this study sought to discover and validate particular genes and putative pathways linked to the course and prognosis of esophageal cancer. Utilizing integrated transcriptomics and single-cell proteomics, the involvement of immune cells in the tumor microenvironment was examined, while bioinformatics was used to investigate the expression, function, and survival data of ADORA2B. Western blot (WB) and qRT-PCR were then used to determine the expression level of ADORA2B in the postoperative tissues of patients with esophageal cancer. Tests using Transwell, Edu, and CCK8 were performed to ascertain its capacity for erosion, migration, invasion, and proliferation. Flow cytometry was used to quantify apoptosis. The results of this investigation validate ADORA2B as a potential therapeutic target and diagnostic biomarker.
Collapse
Affiliation(s)
- Yixiao Cui
- Department of Thoracic and Cardiovascular Surgery, Xinjiang Medical University Affiliated Tumor Hospital, 789 East Suzhou Street, Urumqi, Xinjiang, 830011, People's Republic of China
| | - Yuhan Deng
- Department of Thoracic and Cardiovascular Surgery, Xinjiang Medical University Affiliated Tumor Hospital, 789 East Suzhou Street, Urumqi, Xinjiang, 830011, People's Republic of China
| | - Zhenhua Wu
- Department of Thoracic and Cardiovascular Surgery, Xinjiang Medical University Affiliated Tumor Hospital, 789 East Suzhou Street, Urumqi, Xinjiang, 830011, People's Republic of China
| | - Xiaohong Sun
- Department of Thoracic and Cardiovascular Surgery, Xinjiang Medical University Affiliated Tumor Hospital, 789 East Suzhou Street, Urumqi, Xinjiang, 830011, People's Republic of China.
| |
Collapse
|
8
|
Pan M, Dang A, Huang T, Stover J, Tong MM, Jiang C, Achacoso NS, Bien J, Solorzano AV, Tse P, Chung E, Kanakaveti VP, Felsher D, Fisher GA, Thomas S, Habel L. Association of HER2 amplification or overexpression with overall survival in advanced upper gastrointestinal adenocarcinomas. BJC REPORTS 2025; 3:31. [PMID: 40369309 PMCID: PMC12078702 DOI: 10.1038/s44276-025-00148-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Advanced esophageal (EAC), gastroesophageal junction (GEJAC) and gastric (GAC) adenocarcinomas with HER2 amplification or overexpression (HER2+) are routinely treated with trastuzumab. However, it remains unclear if HER2+ is associated with superior overall survival (OS). METHODS The cohort included recurrent or de novo metastatic GAC, GEJAC and EAC from Kaiser Permanente Northern California. We used Cox regression modelling to examine association between HER2+ and OS, adjusting for demographics, performance status, CCI, receipt of chemotherapy and p53 (mutp53), KRAS (mutKRAS), CDKN2A, PIK3CA co-mutations and MYC amplification. RESULTS Of 875 total eligible patients, 173 had EAC, 276 had GEJAC and 426 had GAC. HER2+ was associated with better OS among the full cohort (HR = 0.74, 95% CI [0.60-0.93]), among EAC (HR = 0.62; [95% CI, 0.40-0.96]) and GEJAC (HR = 0.59; [95% CI, 0.38-0.87]), but not among GAC (HR = 0.89; [95% CI, 0.59-1.35]) patients. GEJAC had better OS than EAC (HR = 0.68, [95% CI, 0.54-0.86]). Trastuzumab treatment was associated with better OS (HR = 0.40, 95% CI [0.21-0.77]). In addition, HER2+ was associated with better OS across the molecular subgroups except that of KRAS mutation (mutKRAS). Our data also show that GEJAC, EAC and GAC were differentially associated with mutp53, mutKRAS and MYC amplification. CONCLUSION HER2+ and treatment with trastuzumab in HER2+ patients were associated with superior OS in upper gastrointestinal adenocarcinomas across molecular subgroups except that of mutKRAS. These results reaffirm the importance of anti-HER2 treatment in HER2+ patients and provide insight on the prognostic and biological divergence among these anatomically linked upper gastrointestinal adenocarcinomas.
Collapse
Affiliation(s)
- Minggui Pan
- Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Research, Kaiser Permanente, Oakland, CA, USA.
| | - Arun Dang
- Internal Medicine Residency Program, Kaiser Permanente, Santa Clara, CA, USA
| | - Tina Huang
- Internal Medicine Residency Program, Kaiser Permanente, Santa Clara, CA, USA
| | - Jack Stover
- Internal Medicine Residency Program, Kaiser Permanente, Santa Clara, CA, USA
| | - Meng M Tong
- Internal Medicine Residency Program, Kaiser Permanente, Santa Clara, CA, USA
| | - Chen Jiang
- Division of Research, Kaiser Permanente, Oakland, CA, USA
| | | | - Jeffrey Bien
- Internal Medicine Residency Program, Kaiser Permanente, Santa Clara, CA, USA
| | | | - Pamela Tse
- Division of Research, Kaiser Permanente, Oakland, CA, USA
| | - Elaine Chung
- Division of Research, Kaiser Permanente, Oakland, CA, USA
| | - Vishnu P Kanakaveti
- Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dean Felsher
- Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - George A Fisher
- Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sachdev Thomas
- Department of Oncology and Hematology, Kaiser Permanente, Vallejo, CA, USA
| | - Laurel Habel
- Division of Research, Kaiser Permanente, Oakland, CA, USA
| |
Collapse
|
9
|
Zhu Y, Fu Z, Duan T, Wang J, Zhang L, Liu G, Guo X, Zhang R. miR-508-5p regulates macrophage polarization via targeting TSGA10 to promote malignant behavior in esophageal cancer cells. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2025:1-15. [PMID: 40349371 DOI: 10.1080/15257770.2025.2491561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/01/2025] [Accepted: 04/05/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Esophageal cancer (EC) is among the deadliest malignancies in humans, with various miRNAs shown to regulate its progression by targeting distinct genes. miR-508-5p was identified as being linked to the malignant behavior of various tumors. Nevertheless, the precise role and mechanism of miR-508-5p in esophageal cancer (EC) remain ambiguous. OBJECTIVE This investigation focuses on the role and mechanism of the miR-508-5p/TSGA10 axis in the progression of EC. METHODS The expression of miR-508-5p and TSGA10 in EC cell lines was evaluated using quantitative Real-Time Polymerase Chain Reaction (qRT-PCR). Cell transfection techniques were used to knock down miR-508-5p and observe its effects on cell proliferation, migration, invasion, and apoptosis. A dual-luciferase reporter gene assay was conducted to verify the targeting relationship of miR-508-5p with TSGA10. Co-culture studies were undertaken to examine the regulatory effect of the miR-508-5p/TSGA10 axis on the polarization state of tumor-associated macrophages (TAMs) and the malignant behavior of EC cells. RESULTS The expression of miR-508-5p was significantly elevated in EC cells. Knocking down miR-508-5p curbed cell proliferation, migration, and invasion while promoting apoptosis. TSGA10 was validated as a primary target gene of miR-508-5p. miR-508-5p knockdown could inhibit the M2 polarization of TAMs by upregulating TSGA10, thereby suppressing the tumorigenic behavior of EC cells. CONCLUSION miR-508-5p promotes the M2 polarization of TAMs and enhances the malignant behavior of EC cells by inhibiting TSGA10.
Collapse
Affiliation(s)
- Yuan Zhu
- Ultrasonic Diagnostic Center, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Zuojun Fu
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Tianjiao Duan
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jing Wang
- Ultrasonic Diagnostic Center, Fugu County People's Hospital, Yulin, China
| | - Lingjuan Zhang
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Guisheng Liu
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xueyan Guo
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Rong Zhang
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
10
|
Zaparte A, Cruz FF, de Souza JB, Morrone FB. P2 receptors signaling in the esophagus: from inflammation to cancer. Purinergic Signal 2025:10.1007/s11302-025-10089-4. [PMID: 40338451 DOI: 10.1007/s11302-025-10089-4] [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: 12/21/2024] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
The signaling mechanisms of nucleotides and nucleosides have been extensively studied over the past decades in various conditions affecting distinct organs and tissues. It is well-established that purinergic receptors are expressed in healthy tissues, with expression levels often increasing under pathological conditions. These receptors play crucial roles in numerous physiological and pathological processes, including inflammation, tissue repair, and cellular signaling. However, the purinergic context in the esophagus and its associated pathologies remains poorly understood, representing a significant gap in current knowledge. In this review, we compiled and analyzed the available data on the involvement of P2 purinergic receptors in esophageal diseases, such as gastroesophageal reflux disease and esophageal carcinoma. Specifically, we discuss the pharmacological modulation, functional characterization, and expression patterns of these receptors in various esophageal cell lines and immune tissue samples, under both healthy and pathological conditions. Understanding the mechanisms of action and signaling pathways involving P2 purinergic receptors in the esophagus can offer valuable insights into their biological roles and emphasize their potential as therapeutic targets for future clinical applications.
Collapse
Affiliation(s)
- Aline Zaparte
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6690, 90619 - 900, Porto Alegre, RS, Brasil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil
| | - Fernanda F Cruz
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil
| | - Julia B de Souza
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6690, 90619 - 900, Porto Alegre, RS, Brasil
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil
| | - Fernanda B Morrone
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6690, 90619 - 900, Porto Alegre, RS, Brasil.
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil.
- Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifical Catholic University of RS, Avenida Ipiranga, 6681, Partenon, 90619 - 900, Porto Alegre, RS, Brasil.
| |
Collapse
|
11
|
Jing T, Tang D. Intratumoral microbiota: a new force in the development and treatment of esophageal cancer. Clin Transl Oncol 2025; 27:1921-1932. [PMID: 39455494 DOI: 10.1007/s12094-024-03757-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Esophageal cancer (EC) ranks among the most prevalent cancers worldwide, with a particularly high incidence in the Asian population. Due to the inconspicuous nature of early symptoms, patients with esophageal cancer are typically diagnosed in the middle to late stages, resulting in suboptimal overall treatment outcomes. Consequently, there is an urgent need to explore and refine therapeutic strategies. Microorganisms have been identified in numerous tumor tissues, including EC, and these microorganisms are referred to as the intratumoral microbiome. Intratumoral microbiota and their metabolic byproducts can influence the progression and treatment of esophageal cancer through various mechanisms, such as modulating tumor cell metabolism and local immune responses. Therefore, the intratumoral microbiota may potentially serve as a target for the treatment of esophageal cancer. This review delineates the composition, origin, and diagnostic significance of intratumoral microbiota in esophageal cancer tissue, and discusses the mechanisms by which intratumoral microbiota contribute to the onset of esophageal cancer. In addition, the impact of intratumoral microbiota on the treatment of esophageal cancer and its intervention measures are also addressed.
Collapse
Affiliation(s)
- Tianyang Jing
- Clinical Medical College, Yangzhou University, Yangzhou, 22500, Jiangsu Province, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
12
|
Wen L, Fu J, Wang Z, Xie R, Tang S, Yu L, Zhou H. Regulatory mechanisms of m6A RNA methylation in esophageal cancer: a comprehensive review. Front Genet 2025; 16:1561799. [PMID: 40330012 PMCID: PMC12053326 DOI: 10.3389/fgene.2025.1561799] [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: 01/22/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Esophageal cancer is an aggressively malignant neoplasm characterized by a high mortality rate. Frequently diagnosed at an advanced stage, it presents challenges for optimal therapeutic intervention due to its non-specific symptoms, resulting in lost opportunities for effective treatment, such as surgery, radiotherapy, chemotherapy and target therapy. The N6-methyladenosine (m6A) modification represents the most critical post-transcriptional modification of eukaryotic messenger RNA (mRNA). The reversible m6A modification is mediated by three regulatory factors: m6A methyltransferases, demethylating enzymes, and m6A recognition proteins. These components identify and bind to specific RNA methylation sites, thereby modulating essential biological functions such as RNA processing, nuclear export, stability, translation and degradation, which significantly influence tumorigenesis, invasion, and metastasis. Given the importance of m6A modification, this paper offers a comprehensive examination of the regulatory mechanisms, biological functions, and future therapeutic implications of m6A RNA methylation in the context of esophageal cancer.
Collapse
Affiliation(s)
- Long Wen
- Department of Thoracic Surgery, Suining Central Hospital, An Affiliated Hospital of Chongqing Medical University, Suining, China
- Graduate School, North Sichuan Medical College, Institute of Surgery, Nanchong, China
| | - Jiang Fu
- Graduate School, Institute of Surgery, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zixu Wang
- Graduate School, Institute of Surgery, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rangping Xie
- Department of Thoracic Surgery, Suining Central Hospital, An Affiliated Hospital of Chongqing Medical University, Suining, China
- Graduate School, Institute of Surgery, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengjie Tang
- Department of Thoracic Surgery, Suining Central Hospital, An Affiliated Hospital of Chongqing Medical University, Suining, China
| | - Li Yu
- Department of Physical Examination, Suining Central Hospital, An Affiliated Hospital of Chongqing Medical University, Suining, China
| | - Haining Zhou
- Department of Thoracic Surgery, Suining Central Hospital, An Affiliated Hospital of Chongqing Medical University, Suining, China
- Graduate School, North Sichuan Medical College, Institute of Surgery, Nanchong, China
- Graduate School, Institute of Surgery, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
13
|
Jiang KC, Zhu YH, Jiang ZL, Liu Y, Hussain W, Luo HY, Sun WH, Ji XY, Li DX. Regulation of PEST-containing nuclear proteins in cancer cells: implications for cancer biology and therapy. Front Oncol 2025; 15:1548886. [PMID: 40330830 PMCID: PMC12052563 DOI: 10.3389/fonc.2025.1548886] [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: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
The PEST-containing nuclear protein (PCNP) is a nuclear protein involved in the regulation of cell cycle progression, protein degradation, and tumorigenesis. PCNP contains a PEST sequence, a polypeptide structural motif rich in proline (P), glutamic acid (E), serine (S), and threonine (T), which serves as a proteolytic recognition signal. The degradation of specific proteins via the PEST sequence plays a crucial role in modulating signaling pathways that control cell growth, differentiation, apoptosis, and stress responses. PCNP is primarily degraded through the ubiquitin-proteasome system (UPS) and the calpain pathway, with phosphorylation of threonine and serine residues further accelerating its degradation. The ubiquitination of PCNP by the ring finger protein NIRF in an E3 ligase-dependent manner is well documented, along with its involvement in the MAPK and PI3K/AKT/mTOR signaling pathways. Additionally, PCNP is implicated in p53-mediated cell cycle arrest and apoptosis, which are essential for inhibiting tumor growth. To explore the role of PCNP in cancer, this review examines its effects on cell growth, differentiation, proliferation, and apoptosis in lung adenocarcinoma, thyroid cancer, ovarian cancer, and other malignancies derived from glandular epithelial cells. By focusing on PCNP and its regulatory mechanisms, this study provides a scientific basis for further research on the biological functions of the PEST sequence in tumor development and cancer progression.
Collapse
Affiliation(s)
- Kai-Chun Jiang
- Department of Traditional Chinese Medicine, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
| | - Yong-Hao Zhu
- School of Stomatology, Henan University, Kaifeng, Henan, China
| | - Zhi-Liang Jiang
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Yi Liu
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Wahab Hussain
- School of Stomatology, Henan University, Kaifeng, Henan, China
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
| | - Huang-Yin Luo
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Wei-Hang Sun
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Xin-Ying Ji
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Oncology, Huaxian County Hospital, Anyang, Henan, China
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
| | - Ding-Xi Li
- The Affiliated Cancer Hospital, Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
14
|
Bao J, Tian X, Pan Y, Guo Y, Yang Z, Gan M, Zheng J. SNRPB2: a prognostic biomarker and oncogenic driver in esophageal cancer via β-catenin/c-Myc signaling. Front Oncol 2025; 15:1536473. [PMID: 40303992 PMCID: PMC12037380 DOI: 10.3389/fonc.2025.1536473] [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: 11/29/2024] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Background The SNRPB2 gene encodes Small Nuclear Ribonucleoprotein Polypeptide B2, a crucial component involved in RNA splicing processes. While SNRPB2 dysregulation has been observed in various cancers, its role in esophageal cancer (ESCA) remains unclear. Methods The mRNA level of SNRPB2 in ESCA was evaluated in combination with TCGA, GTEX, and GEO databases. The prognostic value of SNRPB2 was assessed using Kaplan-Meier analysis. Immunohistochemistry (IHC) was employed to confirm the expression of the SNRPB2 protein in tumor tissues from clinical samples. The biological functions of SNRPB2 were assessed in vitro cell assay and in vivo tumor models. The molecular mechanisms were determined by correlation and gene set enrichment analysis. Western blot experiments validated involvement in signaling pathways. Results Our findings unveiled that SNRPB2 was upregulated at both mRNA and protein levels in ESCA, which was associated with the pathological progression of the disease. Additionally, SNRPB2 served as a robust prognostic biomarker, implicated in driving oncogenic functions in ESCA. It facilitated cell proliferation, migration, and invasion, transitioned the cell cycle, and inhibited apoptosis. Mechanistically, SNRPB2 activated genes associated with the β-catenin/c-Myc signaling pathway, such as β-catenin, c-Myc, CCNA2, CCNB1, CDK1, and CDK2. This activation also regulated the epithelial-to-mesenchymal transition (EMT), thereby facilitating the progression of ESCA. Conclusion Our findings demonstrate that SNRPB2 contributes to ESCA progression by regulating the β-catenin/c-Myc axis, suggesting its potential as a prognostic biomarker and therapeutic target for ESCA patients.
Collapse
Affiliation(s)
- Jiaqian Bao
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Xiong Tian
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yixiao Pan
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhenyu Yang
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Meifu Gan
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Pathology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Jingmin Zheng
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
15
|
Kolstad A, Emanuel G, Hjortland GO, Nilssen Y, Ulvestad M, Areffard A, Aahlin EK. Long-term trends in the clinical management and outcomes of patients with gastroesophageal cancer in Norway. Acta Oncol 2025; 64:540-549. [PMID: 40235057 PMCID: PMC12016665 DOI: 10.2340/1651-226x.2025.43167] [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: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND AND PURPOSE Gastroesophageal cancers are highly prevalent internationally, with many patients diagnosed with metastatic disease, leading to challenging treatment and poor survival. This study uses real-world evidence from a population-level database to describe demographics, clinical characteristics, initial treatment patterns, and survival for patients with gastroesophageal cancer in Norway. MATERIAL AND METHODS Individual patient data was sourced from the Cancer Registry of Norway for patients diagnosed with oesophageal squamous cell carcinoma (ESCC), oesophageal adenocarcinoma (EAC), gastroesophageal junction cancer (GEJC), and gastric cancer from 2001 to 2021, with follow-up from diagnosis to death or last follow-up. Treatment patterns were captured from 2010 to 2022, defined as curative or palliative based on surgery, chemotherapy, and radiotherapy. RESULTS AND INTERPRETATION The cohort included 14,334 Norwegian patients with gastroesophageal cancer; predominantly male, mean age 69-73 years, with a median follow-up of 9-11 months across cancer subtypes. Approximately 40% of patients received curative treatment, and multi-modality treatments increased for EAC, GEJC, and ESCC. Median survival ranged from 6 to 11 months for patients treated palliatively, and 17-95 months for those treated with curative intent. Interestingly, median survival was higher for patients with EAC and GEJC treated with neoadjuvant chemotherapy (86.1 and 75.1 months) versus neoadjuvant chemoradiotherapy (49.1 and 42.1 months), which was confirmed by a multivariate Cox regression model adjusted for age, sex, and disease stage. This study demonstrates that multimodal treatment strategies, consisting of chemotherapy and surgery, may be associated with improved survival outcomes for gastroesophageal cancers. Future studies are required to identify optimum treatment strategies for gastroesophageal cancer subtypes.
Collapse
Affiliation(s)
- Alexander Kolstad
- Department of Gastrointestinal Surgery, University Hospital of North Norway, Tromsø, Norway; Institute of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | | | | | - Yngvar Nilssen
- Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | | | | | - Eirik Kjus Aahlin
- Department of Gastrointestinal Surgery, University Hospital of North Norway, Tromsø, Norway; Institute of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.
| |
Collapse
|
16
|
Wang J, Zhang Z, Song L, Liu X, He X. Inhibition of esophageal squamous cell carcinoma progression by MIR210HG and activation of the P53 signaling pathway to promote apoptosis and autophagy. Eur J Med Res 2025; 30:269. [PMID: 40211342 PMCID: PMC11987309 DOI: 10.1186/s40001-025-02512-8] [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: 10/27/2024] [Accepted: 03/26/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) stands among the frequently occurring malignancies. The lack of efficient early detection methods and therapeutic approaches leads to a high mortality rate for ESCC. The long noncoding RNA MIR210HG is strongly related to various malignant tumors. However, its involvement in ESCC remains unexplored. Thus, this investigation aimed to assess the involvement of MIR210HG in ESCC development. METHODS The MIR210HG expression was analyzed in numerous tumor types through pan-cancer analysis of The Cancer Genome Atlas(TCGA) database. This research investigated the MIR210HG role in the survival and prognosis of individuals with ESCC. The biological functions of MIR210HG were examined by enrichment analyses, including GO, GSEA, and KEGG. Moreover, immune cell infiltration, tumor microenvironment (TME) characteristics, and immune checkpoint expression levels associated with MIR210HG were explored. To get more insight into the connection between MIR210HG and ESCC, we assessed related gene and protein expression using Western blotting and qRT-PCR. To evaluate the proliferation, invasion, migration, apoptosis, and autophagy of ESCC cells, various techniques were employed, including EdU proliferation tests, monodansylcadaverine (MDC) staining, wound healing assays, cell colony formation, transwell assays, flow cytometry, and an established xenograft mouse model. RESULTS MIR210HG exhibited low expression levels in ESCC. High expression of MIR210HG correlated with a higher survival rate among patients. The elevated expression of MIR210HG hindered the ESCC cell's ability to proliferate, invade, and migrate, both in vivo and in vitro settings. Furthermore, a positive correlation between MIR210HG and the P53 signaling pathway was observed, which could affect autophagy and apoptosis in ESCC cells. CONCLUSIONS MIR210HG emerges as a pivotal gene in ESCC, influencing both the immunity and prognosis of patients. Moreover, it may affect autophagy and apoptosis via the P53 signaling pathway. Overall, these outcomes present novel ideas for ESCC treatment.
Collapse
Affiliation(s)
- Jianyu Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong Province, China
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Zhenhu Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong Province, China
| | - Liang Song
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong Province, China
| | - Xiangyan Liu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong Province, China
| | - Xiaopeng He
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, Shandong Province, China.
| |
Collapse
|
17
|
Bhatt A, Zaidi HM, Maitra R, Goel S. Infectious Agents and Esophageal Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:1248. [PMID: 40227819 PMCID: PMC11988037 DOI: 10.3390/cancers17071248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Esophageal cancer, primarily comprising the squamous cell carcinoma (ESCC) and adenocarcinoma (EAC) subtypes, is the sixth leading cause of cancer deaths globally. In addition to many well-established endogenous and exogenous risk factors, there is emerging evidence for the etiologic role of infectious agents in esophageal cancer, although these associations are incompletely understood. Here, we review the currently available literature on the relationship between infectious agents and esophageal cancer. By far, human papilloma virus (HPV), particularly HPV 16 and 18, have the strongest etiologic association with ESCC. Less robust is the association of high-risk HPV (hr-HPV) with EAC. Although H. pylori has been implicated in the development of EAC via increased acid reflux, decreased lower esophageal sphincter tone, and the resultant Barrett's metaplasia-dysplasia-adenocarcinoma pathway, some hypothesize based on epidemiological trends that H. pylori may in fact be a protective factor. In rare cases, EBV can cause esophageal lymphoepithelial carcinoma. Several other agents including HSV, polyomaviruses, and Candida are associated with esophageal cancer to varying degrees. In summary, while several studies, including those conflicting with each other, implicate several infectious agents, the evidence is weak, at best. Clearly, further work is needed to help solidify clear etiologies that will help facilitate prevention and treatment.
Collapse
Affiliation(s)
- Ahan Bhatt
- Jacobi Medical Center, Bronx, NY 10461, USA;
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hasan Musanna Zaidi
- Robert Wood Johnson University Hospital, New Brunswick, NJ 08901, USA;
- Department of Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Radhashree Maitra
- Department of Biology, Yeshiva University, Bronx, NY 10461, USA;
- Department of Oncology, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Sanjay Goel
- Robert Wood Johnson University Hospital, New Brunswick, NJ 08901, USA;
- Department of Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Division of Medical Oncology, Rutgers Cancer Institute, New Brunswick, NJ 08901, USA
| |
Collapse
|
18
|
Ma Y, Jiang Z, Wang Y, Pan L, Liu K, Xia R, Yuan L, Cheng X. Tongue coating microbiota-based machine learning for diagnosing digestive system tumours. J Oral Microbiol 2025; 17:2487645. [PMID: 40206097 PMCID: PMC11980229 DOI: 10.1080/20002297.2025.2487645] [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: 12/14/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
Background Digestive system tumours (DSTs) often diagnosed late due to nonspecific symptoms. Non-invasive biomarkers are crucial for early detection and improved outcomes. Patients and Methods We collected tongue coating samples from 710 patients diagnosed with DST and 489 healthy controls (HC) from April 2023, to December 2023. Microbial composition was analyzed using 16S rRNA sequencing, and five machine learning algorithms were applied to assess the diagnostic potential of tongue coating microbiota. Results Alpha diversity analysis showed that the microbial diversity in the tongue coating was significantly increased in DST patients. LEfSe analysis identified DST-enriched genera Alloprevotella and Prevotella, contrasting with HC-dominant taxa Neisseria, Haemophilus, and Porphyromonas (LDA >4). Notably, when comparing each of the four DST subtypes with the HC group, the proportion of Haemophilus in the HC group was significantly higher, and it was identified as an important feature for distinguishing the HC group. Machine learning validation demonstrated superior diagnostic performance of the Extreme Gradient Boosting (XGBoost) model, achieving an AUC of 0.926 (95% CI: 0.893-0.958) in internal validation, outperforming the other four machine learning models. Conclusion Tongue coating microbiota shows promise as a non-invasive biomarker for DST diagnosis, supported by robust machine learning models.
Collapse
Affiliation(s)
- Yubo Ma
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhengchen Jiang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yanan Wang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Libin Pan
- Department of Pharmacy, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Kang Liu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruihong Xia
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yuan
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Department of Integrated Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
19
|
Sun Y, Tang Y, Qi Q, Pang J, Chen Y, Wang H, Liang J, Tang W. 101 Machine Learning Algorithms for Mining Esophageal Squamous Cell Carcinoma Neoantigen Prognostic Models in Single-Cell Data. Int J Mol Sci 2025; 26:3373. [PMID: 40244296 PMCID: PMC11989522 DOI: 10.3390/ijms26073373] [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: 01/24/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive malignant tumors in the digestive tract, characterized by a high recurrence rate and inadequate immunotherapy options. We analyzed mutation data of ESCC from public databases and employed 10 machine learning algorithms to generate 101 algorithm combinations. Based on the optimal range determined by the concordance index, we randomly selected one combination from the best-performing algorithms to construct a prognostic model consisting of five genes (DLX5, MAGEA4, PMEPA1, RCN1, and TIMP1). By validating the correlation between the prognostic model and antigen-presenting cells (APCs), we revealed the antigen-presentation efficacy of the model. Through the analysis of immune infiltration in ESCC, we uncovered the mechanisms of immune evasion associated with the disease. In addition, we examined the potential impact of the five prognostic genes on ESCC progression. Based on these insights, we identified anti-tumor small-molecule compounds targeting these prognostic genes. This study primarily simulates the tumor microenvironment (TME) and antigen presentation processes in ESCC patients, predicting the role of the neoantigen-based prognostic model in ESCC patients and their potential responses to immunotherapy. These results suggest a potential approach for identifying therapeutic targets in ESCC, which may contribute to the development of more effective treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wenru Tang
- Laboratory of Molecular Genetics of Aging & Tumor, Medicine School, Kunming University of Science and Technology, No. 727, Jingming South Road, Kunming 650500, China; (Y.S.); (Y.T.); (Q.Q.); (J.P.); (Y.C.); (H.W.); (J.L.)
| |
Collapse
|
20
|
Liu K, Bie J, Zhang R, Xiong R, Peng L, Luo Y, Yang S, Feng G, Song G. AGTR1 potentiates the chemotherapeutic efficacy of cisplatin in esophageal carcinoma through elevation of intracellular Ca 2+ and induction of apoptosis. Int J Oncol 2025; 66:32. [PMID: 40084687 PMCID: PMC11900935 DOI: 10.3892/ijo.2025.5738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 10/25/2024] [Indexed: 03/16/2025] Open
Abstract
Cisplatin is one of the principal chemotherapeutic agents used for esophageal cancer (EC) treatment; however, EC mortality remains high. It is therefore imperative to find new therapeutic targets and approaches to potentiate the chemotherapeutic efficacy of cisplatin. Angiotensin II receptor type 1 (AGTR1) is a potential therapeutic target in multiple cancer types. In the present study, RNA‑sequencing analysis of EC and normal esophageal tissues was performed and AGTR1 was identified as a differentially expressed gene that is markedly downregulated in recurrent and metastasized EC. AGTR1 upregulation in the esophageal squamous cell carcinoma cell lines, KYSE‑150 and EC109, promoted their chemosensitivity to cisplatin both in vitro and in vivo. Additionally, AGTR1 suppressed the metastasis‑relevant traits of EC cells, as evidenced by the reduced migration, invasion and wound healing of EC cells with higher AGTR1 expression levels. Moreover, AGTR1 overexpression in EC cells upregulated intracellular Ca2+ levels, reduced ATP levels and mitochondrial membrane potentials, which was accompanied by enhanced mitochondrial pathway apoptosis. Notably, either AGTR1 overexpression or treatments with the calcium channel blocker, fendiline, caused Ca2+ influx and promoted mitochondria‑dependent apoptosis in KYSE‑150 cells in vitro. These effects were augmented when both AGTR1 overexpression and fendiline stimulation were imposed in the absence or presence of cisplatin treatments. Furthermore, fendiline administration enhanced the chemosensitivity of cisplatin in an EC xenograft mouse model. Collectively, these findings offer an alternative treatment option and provide mechanistic insights into using fendiline to potentiate the chemotherapy efficacy of cisplatin in treating EC.
Collapse
Affiliation(s)
- Kang Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Jun Bie
- Department of Oncology, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Ruolan Zhang
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Rong Xiong
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Lihong Peng
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yi Luo
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Siyun Yang
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Gang Feng
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guiqin Song
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
- School of Basic Medicine and Forensic Sciences, North Sichuan Medical College, Nanchong, Sichuan 637100, P.R. China
| |
Collapse
|
21
|
Baumeister T, Proaño-Vasco A, Metwaly A, Kleigrewe K, Kuznetsov A, Schömig LR, Borgmann M, Khiat M, Anand A, Strangmann J, Böttcher K, Haller D, Dunkel A, Somoza V, Reiter S, Meng C, Thimme R, Schmid RM, Patil DT, Burgermeister E, Huang Y, Sun Y, Wang HH, Wang TC, Abrams JA, Quante M. Loss of FXR or Bile Acid-dependent Inhibition Accelerate Carcinogenesis of Gastroesophageal Adenocarcinoma. Cell Mol Gastroenterol Hepatol 2025; 19:101505. [PMID: 40139565 DOI: 10.1016/j.jcmgh.2025.101505] [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: 06/17/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND & AIMS The incidence of Barrett esophagus (BE) and gastroesophageal adenocarcinoma (GEAC) correlates with obesity and a diet rich in fat. Bile acids (BAs) support fat digestion and undergo microbial metabolism in the gut. The farnesoid X receptor (FXR) is an important modulator of the BA homeostasis. When activated, FXR can inhibit cancer-related processes, and thus, it is an appealing therapeutic target. Here, we assess the effect of diet on the microbiota-BA axis and evaluate the role of FXR in disease progression. METHODS L2-IL1B mice (mouse model of BE and GEAC) under different diets, and L2-IL1B-FXR KO-mice were characterized. L2-IL1B-derived organoids were exposed to different BAs and to the FXR agonist obeticholic acid (OCA). The BA profile in serum and stool of healthy controls and patients with BE and GEAC was assessed. RESULTS Here we show that a high-fat diet accelerated tumorigenesis in L2-IL1B mice while increasing BA levels and altering the composition of the gut microbiota. Although upregulated in BE, expression of FXR was downregulated in GEAC in mice and humans. In L2-IL1B mice, FXR knockout enhanced the dysplastic phenotype and increased Lgr5 progenitor cell numbers. Treatment of murine BE organoids and L2-IL1B mice with OCA notably ameliorated the phenotype. CONCLUSION GEAC carcinogenesis appears to be partially driven via loss or inhibition of FXR on progenitor cells at the gastroesophageal junction. Considering that the resulting aggravation in the phenotype could be reversed with OCA treatment, we suggest that FXR agonists have great potential as a preventive strategy against GEAC progression.
Collapse
Affiliation(s)
- Theresa Baumeister
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany; Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany
| | - Andrea Proaño-Vasco
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Amira Metwaly
- Department of Nutrition and Immunology, Technical University of Munich, Munich, Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Alexander Kuznetsov
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Linus R Schömig
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Martin Borgmann
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Mohammed Khiat
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Akanksha Anand
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany
| | - Julia Strangmann
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Katrin Böttcher
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany
| | - Dirk Haller
- Department of Nutrition and Immunology, Technical University of Munich, Munich, Germany
| | - Andreas Dunkel
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, Munich, Germany; Department of Food Chemistry and Molecular Sensory Science, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Veronika Somoza
- Leibniz-Institute for Food Systems Biology, Technical University of Munich, Munich, Germany
| | - Sinah Reiter
- Department of Food Chemistry and Molecular Sensory Science, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Robert Thimme
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Roland M Schmid
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany
| | - Deepa T Patil
- Department of Pathology, School of Medicine, Digestive Health Research Institute, Case Western Reserve University; Cleveland, Ohio
| | - Elke Burgermeister
- Department of Internal Medicine II, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Yiming Huang
- Systems and Synthetic Biology, Columbia University Medical Center, New York, New York
| | - Yiwei Sun
- Systems and Synthetic Biology, Columbia University Medical Center, New York, New York
| | - Harris H Wang
- Systems and Synthetic Biology, Columbia University Medical Center, New York, New York
| | - Timothy C Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Michael Quante
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
22
|
Yuan J, Cui Y, Zhang J, Cai Y, Xu X. CircMMP11 is a Potential Recurrence Biomarker and Facilitates Progression of Esophageal Squamous Cell Carcinoma. Biochem Genet 2025:10.1007/s10528-025-11080-9. [PMID: 40085314 DOI: 10.1007/s10528-025-11080-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Esophageal cancer is a deadly tumor with a high mortality rate and unsatisfactory treatment effect. Circular RNAs (circRNAs), as a new kind of noncoding RNA molecules, were found to play a key role in variety of tumors. This study aimed to explore the participation of hsa_circ_0062558 (circMMP11) in the recurrence of ESCC and its role in ESCC progression. The expression of circMMP11 in tissue specimens and cells was measured using the RT-qPCR method. RNase R treatment assay and Actinomycin D treatment assay verified the stability of circMMP11. Receiver Operating Characteristic (ROC) curve was conducted to evaluate the clinical significance of circMMP11 in predicting postoperative recurrence. The capacities of circMMP11 on cellular behaviors were measured using cell counting kit (CCK-8) and Transwell assays. The circMMP11 expression was raised in ESCC tissues. The circMMP11 in tumor tissues of the recurrence/metastasis group was higher than that in the non-recurrence/metastasis group. ROC curve showed that circMMP11 in tumor tissues could detect the postoperative recurrence/metastasis of the patients with an area under the ROC curve (AUC) of 0.838. Silencing circMMP11 led to a reduction in the proliferative, migratory, and invasive capacities of ESCC cells, and miR-671-5p inhibitor partially diminished the inhibitory effects of si-circMMP11. The high circMMP11 expression in postoperative tissues of patients with ESCC is correlated with recurrence and metastasis, and it has potential predictive value for postoperative recurrence and metastasis of patients. Inhibition of circMMP11 repressed ESCC cell behaviors by regulating miR-671-5p, which may be a potential target for early diagnosis of recurrence and treatment of ESCC.
Collapse
Affiliation(s)
- Jingnan Yuan
- College of Life Sciences, University of Chinese Academy of Sciences, No. 19Jia, Yuquan Road, Shijingshan District, Beijing, 100049, China
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, BGI Research, No.1, Banshan East Road, Gongshu District, Hangzhou, 310000, China
| | - Ying Cui
- Department II of Radiation Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, 266042, China
| | - JiWen Zhang
- Department of Gastroenterology, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Yan Cai
- Department of Gastroenterology, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Xun Xu
- College of Life Sciences, University of Chinese Academy of Sciences, No. 19Jia, Yuquan Road, Shijingshan District, Beijing, 100049, China.
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, BGI Research, No.1, Banshan East Road, Gongshu District, Hangzhou, 310000, China.
| |
Collapse
|
23
|
Norollahi SE, Morovat S, Keymoradzadeh A, Hamzei A, Modaeinama M, Soleimanmanesh N, Soleimanmanesh Y, Najafizadeh A, Bakhshalipour E, Alijani B, Samadani AA. Transforming agents: The power of structural modifications in glioblastoma multiforme therapy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:41-56. [PMID: 39701498 DOI: 10.1016/j.pbiomolbio.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024]
Abstract
Glioblastoma (GBM) is a very deadly type of brain tumor with a poor prognosis and a short survival rate. Recent advancements in understanding GBM's molecular and genetic characteristics have led to the development of various therapeutic and diagnostic strategies. Key elements such as microRNAs, lncRNAs, exosomes, angiogenesis, and chromatin modifications are highlighted, alongside significant epigenetic alterations that impact therapy and diagnosis. Despite these advancements, molecular classifications have not improved patient outcomes due to intratumoral diversity complicating targeted therapies. In this article, it is tried to emphasize the potential of investigating the epigenetic landscape of GBM, particularly identifying patients with diffuse hypermethylation at gene promoters associated with better outcomes. Integrating epigenetic and genetic data has enhanced the identification of glioma subtypes with high diagnostic precision. The reversibility of epigenetic changes offers promising therapeutic prospects, as recent insights into the "epigenetic orchestra" suggest new avenues for innovative treatment modalities for this challenging cancer. In this review article, we focus on the roles of translational elements and their alterations in the context of GBM diagnosis and therapy.
Collapse
Affiliation(s)
- Seyedeh Elham Norollahi
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran; Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran
| | - Saman Morovat
- Department of Medical Genetics and Molecular Biology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Arman Keymoradzadeh
- Department of Neurosurgery, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Hamzei
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Morteza Modaeinama
- Department of Neurosurgery, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | - Ali Najafizadeh
- School of Paramedicine Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Elahe Bakhshalipour
- School of Paramedicine Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Babak Alijani
- Department of Neurosurgery, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran; Neuroscience Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
24
|
Wang Z, Li K, Zhang X, Jiang F, Xu L. LINC00942 Accelerates Esophageal Cancer Progression by Raising PRKDC Through Interaction With PTBP1. J Biochem Mol Toxicol 2025; 39:e70220. [PMID: 40103322 DOI: 10.1002/jbt.70220] [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: 08/07/2024] [Revised: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Aberrantly expressed LINC00942 is participated in the progression of several cancers. However, the function of LINC00942 in esophageal cancer (ESCA) is unclear. The objective of this study was to explore the effect of LINC00942 on ESCA and its possible molecular mechanisms. First, differentially expressed lncRNAs in ESCA were analyzed using GSE192662 microarray. catRAPID omics v2.1 was applied to predict the proteins that might interact with LINC00942. SDS-PAGE silver staining assay, RNA pull down, and RIP assay were utilized to validate proteins interacting with LINC00942. Then, RNA seq was applied to detect the downstream targets of PTBP1, and KEGG enrichment analysis was used to analyze the genes involved in proliferation and migration-related signaling pathways. In addition, CCK-8, EdU and transwell were used to detect the impact of LINC00942 on ESCA cell function. Bioinformatics revealed that LINC00942 was significantly overexpressed in ESCA. Patients in low-expression of LINC00942 had an obviously better prognosis. After LINC00942 knockdown, the proliferation and migration of TE-1 and OE19 were dramatically reduced. Subsequently, PTBP1 was found to interact with LINC00942, and PRKDC was a downstream target of PTBP1. Functional analysis showed that TE-1 and OE19 cell proliferation and migration were markedly elevated after LINC00942 overexpression, and knockdown of PRKDC significantly reversed this effect. Mechanistically, LINC00942 promoted PRKDC expression by interacting with PTBP1. In summary, LINC00942 facilitated the proliferation and migration of ESCA cells via binding to PTBP1 to promote PRKDC expression.
Collapse
Affiliation(s)
- Zhongqiu Wang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Kang Li
- Department of Radiology, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xing Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Feng Jiang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
25
|
Veziant J, Routier B, Piessen G. [The role of surgery in preventing esophageal cancer]. Bull Cancer 2025; 112:277-285. [PMID: 40049796 DOI: 10.1016/j.bulcan.2024.06.012] [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: 06/10/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 05/13/2025]
Abstract
The prognosis for esophageal cancer remains poor because it is often diagnosed late and patients often have unfavourable backgrounds. This is reflected in standardised 5-year net survival rates of no more than 20%, regardless of gender. Unlike gastric cancer (CDH1 gene mutation), there is currently no genetic predisposition to esophageal cancer that would justify prophylactic esophagectomy. Primary prevention by identifying and managing modifiable risk factors is therefore the best strategy for preventing esophageal cancer. The role of surgery in the prevention of esophageal cancer is discussed in this review. Although recommended, the value of antireflux surgery (fundoplication) for Barrett's esophagus with the sole aim of reducing the risk of esophageal adenocarcinoma remains controversial. With regard to bariatric surgery, national cohort studies report an equivalent or reduced incidence of esophageal adenocarcinoma in operated patients compared with non-operated obese patients. However, given the significant increase in the number of bariatric procedures, further studies with longer follow-up are needed. In addition, although surgical myotomy is a truly effective therapeutic option for the treatment of achalasia in the first-line setting, its impact on the risk of esophageal cancer remains uncertain and poorly studied.
Collapse
Affiliation(s)
- Julie Veziant
- Hôpital Claude-Huriez, université de Lille, CHU de Lille, service de chirurgie digestive et oncologique, 59000 Lille, France; University Lille, CHU de Lille, CNRS, UMR9020-U1277-CANTHER-Cancer, Inserm, 59000 Lille, France.
| | - Baptiste Routier
- Hôpital Claude-Huriez, université de Lille, CHU de Lille, service de chirurgie digestive et oncologique, 59000 Lille, France
| | - Guillaume Piessen
- Hôpital Claude-Huriez, université de Lille, CHU de Lille, service de chirurgie digestive et oncologique, 59000 Lille, France; University Lille, CHU de Lille, CNRS, UMR9020-U1277-CANTHER-Cancer, Inserm, 59000 Lille, France
| |
Collapse
|
26
|
Akhtar S, Al-Shammari A, Al-Huraiti M, Al-Anjery F. Age-period-cohort modeling of oesophageal carcinoma risk in a middle eastern country: 1980-2019. J Public Health (Oxf) 2025; 47:e59-e66. [PMID: 39674677 DOI: 10.1093/pubmed/fdae311] [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: 08/05/2024] [Accepted: 11/29/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Understanding of the factors influencing oesophageal cancer trends is crucial. Therefore, this cross-sectional cohort study sought to disentangle the age, period and cohort effects on the trends of oesophageal cancer in Kuwait. METHODS The data on incident oesophageal carcinoma cases diagnosed between January 1, 1980, through December 31, 2019, and reference population were obtained. Age-period-cohort (APC) analysis was conducted using a loglinear Poisson regression model. RESULTS A total of 496 oesophageal carcinoma cases in 12.8 million person-years (i.e. squamous-cell carcinoma, 269, 54.23%), adenocarcinoma,147, 29.64% and unspecified cases, 80,16.13%) were diagnosed. The overall age-standardized incidence rate (per 105 person-years) of oesophageal carcinoma during the study period was 10.51 (95% CI: 6.62-14.41). The APC analysis results showed that the age and birth cohort effects were the significant determinants of declining, and subsequently steadying the oesophageal carcinoma incidence rates. CONCLUSIONS A substantial decline in oesophageal carcinoma incidence rates was recorded, which significantly varied in all three temporal dimensions. The observed birth cohort patterns suggest changing lifestyle and dietary patterns seem to be responsible for decreasing oesophageal carcinoma risk in Kuwait. Future studies may look for the component causes maintaining the endemicity of oesophageal carcinoma risk in this and similar countries in the region.
Collapse
Affiliation(s)
- Saeed Akhtar
- Department of Community Medicine and Behavioural Sciences, College of Medicine, Kuwait University, PO Box 24923, Safat 13110, Kuwait
| | - Ahmad Al-Shammari
- Department of Surgery, Faculty of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | | | - Fouzan Al-Anjery
- Ministry of Health, Jamal Abdel Nasser Street, Sulaibkhat 13001, Kuwait
| |
Collapse
|
27
|
Fan M, Zhu Y, Qian L, Hu C, Ding H. Association between preoperative inflammatory status via CALLY index and postoperative pneumonia occurrence in resectable esophageal squamous cell carcinoma patients: a retrospective cohort study. Front Oncol 2025; 15:1486983. [PMID: 40034601 PMCID: PMC11872739 DOI: 10.3389/fonc.2025.1486983] [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: 08/27/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Background Postoperative pneumonia significantly affects recovery and prognosis in patients with esophageal squamous cell carcinoma. The CALLY index, derived from preoperative hematological parameters, may serve as a predictive marker for such complications. Objectives To assess the association between preoperative inflammatory status via the CALLY index and the occurrence of postoperative pneumonia in patients with resectable ESCC. Methods A retrospective cohort study was conducted from January 2020 to December 2022 at The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University. A total of 215 patients who met inclusion criteria were analyzed. Clinical data, including CALLY indices calculated preoperatively, were collected. Propensity score matching was applied to minimize confounding biases. The predictive value of the CALLY index was assessed using receiver operating characteristic analysis, and logistic regression was used to identify factors associated with postoperative pneumonia. Results ROC curve analysis demonstrated the CALLY index had an area under the curve of 0.764 for predicting postoperative pneumonia, with a cutoff value of 1.97 achieving 67.69% sensitivity and 84.67% specificity. In multivariate analysis, a lower CALLY index was significantly associated with increased pneumonia risk, independent of other factors (adjusted OR = 0.66, p < 0.001). High CALLY index scores correlated with a decreased likelihood of postoperative pneumonia, reinforcing its utility as a non-invasive prognostic marker. Conclusions The CALLY index is a robust, independent predictor of postoperative pneumonia in patients with resectable ESCC. Preoperative assessment of this index could enhance risk stratification and guide proactive management strategies to improve postoperative outcomes.
Collapse
Affiliation(s)
| | | | | | - Chuanxian Hu
- Department of Cardiothoracic Surgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| | - Hui Ding
- Department of Cardiothoracic Surgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huaian, China
| |
Collapse
|
28
|
Huang X, Huang Y, Li P, Xu K. CT-Based Deep Learning Predicts Prognosis in Esophageal Squamous Cell Cancer Patients Receiving Immunotherapy Combined with Chemotherapy. Acad Radiol 2025:S1076-6332(25)00101-1. [PMID: 39956748 DOI: 10.1016/j.acra.2025.01.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/18/2025]
Abstract
RATIONALE AND OBJECTIVES Immunotherapy combined with chemotherapy has improved outcomes for some esophageal squamous cell carcinoma (ESCC) patients, but accurate pre-treatment risk stratification remains a critical gap. This study constructed a deep learning (DL) model to predict survival outcomes in ESCC patients receiving immunotherapy combined with chemotherapy. MATERIALS AND METHODS A DL model was developed to predict survival outcomes in ESCC patients receiving immunotherapy and chemotherapy. Retrospective data from 482 patients across three institutions were split into training (N=322), internal test (N=79), and external test (N=81) sets. Unenhanced computed tomography (CT) scans were processed to analyze tumor and peritumoral regions. The model evaluated multiple input configurations: original tumor regions of interest (ROIs), ROI subregions, and ROIs expanded by 1 and 3 pixels. Performance was assessed using Harrell's C-index and receiver operating characteristic (ROC) curves. A multimodal model combined DL-derived risk scores with five key clinical and laboratory features. The Shapley Additive Explanations (SHAP) method elucidated the contribution of individual features to model predictions. RESULTS The DL model with 1-pixel peritumoral expansion achieved the best accuracy, yielding a C-index of 0.75 for the internal test set and 0.60 for the external test set. Hazard ratios for high-risk patients were 1.82 (95% CI: 1.19-2.46; P=0.02) in internal test set. The multimodal model achieved C-indices of 0.74 and 0.61 for internal and external test sets, respectively. Kaplan-Meier analysis revealed significant survival differences between high- and low-risk groups (P<0.05). SHAP analysis identified tumor response, risk score, and age as critical contributors to predictions. CONCLUSION This DL model demonstrates efficacy in stratifying ESCC patients by survival risk, particularly when integrating peritumoral imaging and clinical features. The model could serve as a valuable pre-treatment tool to facilitate the implementation of personalized treatment strategies for ESCC patients undergoing immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Xiaoyu Huang
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (X.H., P.L.)
| | - Yong Huang
- Department of Medical Oncology, The Second People's Hospital of Hefei, Hefei, China (Y.H.)
| | - Ping Li
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (X.H., P.L.); Graduate School of Anhui University of Traditional Chinese Medicine, Hefei, China (P.L.)
| | - Kai Xu
- Scholl of Internet, Anhui University, Hefei, China (K.X.).
| |
Collapse
|
29
|
Yin R, Zhou G, Liu G, Hou X, Yang H, Ge J, Ying J. Pan-cancer and multi-omics analysis: NDUFA1 is a potential therapeutic target and prognostic marker for esophageal cancer. Cell Biol Toxicol 2025; 41:43. [PMID: 39937347 PMCID: PMC11821742 DOI: 10.1007/s10565-025-09993-7] [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: 08/09/2024] [Accepted: 01/28/2025] [Indexed: 02/13/2025]
Abstract
OBJECTIVES The study examined the NDUFA1 expression and its prognostic value in pan-cancer, especially in esophageal cancer (ESCA). Its carcinogenic effect on ESCA was further elucidated. MATERIALS AND METHODS TCGA database was used to examine NDUFA1 expression and its prognostic value in 33 cancer types. GO and KEGG were performed for function and pathway enrichment of NDUFA1-related genes. The carcinogenic effect on ESCA was verified in both KYSE-30 cell and Xenograft mouse model. RESULTS Abnormal high expression of NDUFA1 was detected in pan-cancer, and related to immune cell infiltration. TCGA database indicated an elevated value of NDUFA1 in ESCA tumor tissues, which was linked to patients' poor prognosis. NDUFA1-related genes were mainly enriched in oxidative stress and immune response in ESCA. NDUFA1 knockdown significantly suppressed ESCA cell proliferation, migration and invasion. Similarly, tumor growth of ESCA xenograft mice was inhibited by NDUFA1 knockdown. Activated PI3K-Akt signaling was detected in both ESCA cell lines and tumor tissues, which was reversed by NDUFA1 knockdown. CONCLUSION Multi-omics analysis showed that NDUFA1 might be adopted as a potential therapeutic goal and prognostic indicator for a variety of cancers, especially for ESCA. NDUFA1 knockdown inhibited ESCA tumor growth, which may have the participation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Rui Yin
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China
| | - Gai Zhou
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China
| | - Guanqi Liu
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China
| | - Xiaoting Hou
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China
| | - Haifeng Yang
- Department of Infectious Disease, Economic Development Zone, People's Hospital of Xuyi, No. 28, Hongwu Avenue, Xuyi, 211700, China.
| | - Jianxin Ge
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China.
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China.
| | - Jie Ying
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China.
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China.
| |
Collapse
|
30
|
Guo X, Huang A, Qi Y, Chen J, Yang M, Jin M. METTL3/IGF2BP2 Promotes the Malignant Progression of Esophageal Cancer by Activating the PIK3CA/AKT Pathway. Thorac Cancer 2025; 16:e70022. [PMID: 39980152 PMCID: PMC11842509 DOI: 10.1111/1759-7714.70022] [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: 12/02/2024] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Esophageal cancer (EC) is a leading cause of cancer-related mortality worldwide. Methyltransferase-like 3 (METTL3), a key enzyme involved in m6A methylation, has been implicated in the development and progression of various cancers, including EC. However, its potential mechanism of action in EC progression remains unclear. METTL3 expression was found to be upregulated in EC tissues and cells. Knockdown of METTL3 suppressed EC cell proliferation, invasion, migration, and angiogenesis, while promoting apoptosis. Mechanistically, METTL3 maintained PIK3CA mRNA expression and stability in an m6A-dependent and IGF2BP2-dependent manner, respectively. METTL3 silencing inactivated the AKT pathway by regulating PIK3CA expression. Furthermore, overexpression of PIK3CA mitigated the effects of METTL3 silencing on the malignant growth of KYSE180 and TE1 cells in vivo and in vitro. METTL3/IGF2BP2 promoted the malignant progression of EC by activating the PIK3CA/AKT pathway. Targeting the METTL3-PIK3CA axis may offer a novel therapeutic approach for EC treatment.
Collapse
Affiliation(s)
- Xinmeng Guo
- Department of PathologyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Anqi Huang
- Department of PathologyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Ya'nan Qi
- Department of PathologyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Jiaqi Chen
- Department of PathologyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Meng Yang
- Department of PathologyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Mulan Jin
- Department of PathologyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
31
|
Li J, Wang Y, Wei S, Xu S, Dai S, Zhang L, Tian Z, Zhao L, Lv H. NEK2 Promotes ESCC Malignant Progression by Inhibiting Cellular Senescence via the FOXM1/c-Myc/p27 Signaling Pathway. Mol Carcinog 2025; 64:244-259. [PMID: 39503194 DOI: 10.1002/mc.23839] [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: 06/21/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 01/15/2025]
Abstract
Never in mitosis gene A (NIMA)-related kinase 2 (NEK2) is a crucial serine-threonine kinase involved in the process of cell mitosis. However, the precise relationship between NEK2 and esophageal squamous cell carcinoma (ESCC) remains inadequately understood. NEK2 expression in ESCC tissues was assessed through bioinformatics analysis, reverse transcription-quantitative PCR (RT-qPCR) and immunohistochemistry, revealing a correlation with ESCC patient prognosis. Cultured ESCC cells and human normal esophageal epithelial cells (HEEC) were used to investigate the effects of NEK2 knockdown on the development and progression of ESCC by integrated confluence algorithm, colony formation, wound-healing, transwell, and ESCC xenograft tumor model, in vitro and in vivo. In ESCC tissues, NEK2 was found to be significantly upregulated, and its expression correlated with poor prognosis in ESCC patients. NEK2 may facilitate ESCC development by regulating cell proliferation, migration, and invasion. Additionally, results from in vivo experiments suggested that NEK2 knockdown can inhibit tumor growth. Moreover, forkhead box M1 (FOXM1) was identified as a potential downstream target of NEK2 in the regulation of ESCC, with its overexpression reversing the effects of NEK2 knockdown on ESCC. Mechanistic studies also indicated that NEK2 may promote the malignant progression of ESCC by inhibiting cellular senescence through the activation of the FOXM1/c-Myc/p27 signaling pathways, which may provide a novel perspective for the management of ESCC.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yaojie Wang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sisi Wei
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shi Xu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Suli Dai
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li Zhang
- Department of Geriatric, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huilai Lv
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
32
|
Wang M, Dong W, Wu G, Zhang B, Lai T, Liu A, Sun Q. Efficacy and safety of neoadjuvant immunotherapy combined with chemotherapy for stage II-IVa esophageal cancer: a network meta-analysis. Syst Rev 2025; 14:26. [PMID: 39871293 PMCID: PMC11773777 DOI: 10.1186/s13643-025-02765-8] [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: 05/12/2024] [Accepted: 01/12/2025] [Indexed: 01/29/2025] Open
Abstract
OBJECTIVE The objective of this study was to evaluate the clinical efficacy and safety of neoadjuvant immunochemotherapy in the treatment of locally advanced, resectable esophageal cancer. METHODS Literature published before November 2023 on the clinical efficacy and safety of neoadjuvant immunotherapy in resectable esophageal squamous cell carcinoma was searched in CNKI, VIP, Wanfang, Chinese Biomedical Literature, PubMed, Embase, Cochrane, and the Web of Science. A meta-analysis was conducted using Stata 17.0. RESULTS The cumulative ranked probability results indicated that Camrelizumab + TN had the highest probability of achieving pCR, Camrelizumab + TP of achieving MPR, and Sintilimab + TP of achieving DCR and ORR. Camrelizumab + TP also had the highest probability of achieving an R0 resection rate. In terms of adverse events and postoperative complications, Pembrolizumab + TN had the highest likelihood of inducing myelosuppression and rash. Toripalimab + TP had the highest probability of inducing vomiting, while traditional chemotherapy alone had the highest likelihood of inducing postoperative cardiac adverse events. CONCLUSION Neoadjuvant immunotherapy combined with chemotherapy has demonstrated superior clinical efficacy and safety compared to chemotherapy alone. The regimen of Camrelizumab + TP showed significant advantages in pCR, MPR, DCR, and R0 resection rates, particularly excelling in MPR and R0 resection rates. However, it was associated with a higher incidence of rash compared to chemotherapy alone and the Toripalimab + TP regimen. Neoadjuvant immunotherapy, when combined with chemotherapy, has been shown to reduce the occurrence of postoperative cardiac adverse events. Among the various treatment options, Sintilimab + TP exhibited the most favorable outcomes. SYSTEMATIC REVIEW REGISTRATION PROSPERO Protocol Number: CRD42024623160.
Collapse
Affiliation(s)
- Mingxing Wang
- Department of Medical Oncology, Lu'an Hospital of Traditional Chinese Medicine Affiliated to Anhui University of Chinese Medicine, No. 76 Renmin Road, Zhongshi Street, Jin'an District, Lu'an, 237000, China
| | - Wanhui Dong
- Department of Medical Oncology, Lu'an Hospital of Traditional Chinese Medicine Affiliated to Anhui University of Chinese Medicine, No. 76 Renmin Road, Zhongshi Street, Jin'an District, Lu'an, 237000, China.
| | - Gongyi Wu
- Department of Medical Oncology, Lu'an Hospital of Traditional Chinese Medicine Affiliated to Anhui University of Chinese Medicine, No. 76 Renmin Road, Zhongshi Street, Jin'an District, Lu'an, 237000, China
| | - Baorui Zhang
- Department of Medical Oncology, Lu'an Hospital of Traditional Chinese Medicine Affiliated to Anhui University of Chinese Medicine, No. 76 Renmin Road, Zhongshi Street, Jin'an District, Lu'an, 237000, China
| | - Tong Lai
- Department of Medical Oncology, Lu'an Hospital of Traditional Chinese Medicine Affiliated to Anhui University of Chinese Medicine, No. 76 Renmin Road, Zhongshi Street, Jin'an District, Lu'an, 237000, China
| | - Aixin Liu
- Department of Medical Oncology, Lu'an Hospital of Traditional Chinese Medicine Affiliated to Anhui University of Chinese Medicine, No. 76 Renmin Road, Zhongshi Street, Jin'an District, Lu'an, 237000, China
| | - Qingming Sun
- Department of Medical Oncology, Lu'an Hospital of Traditional Chinese Medicine Affiliated to Anhui University of Chinese Medicine, No. 76 Renmin Road, Zhongshi Street, Jin'an District, Lu'an, 237000, China
| |
Collapse
|
33
|
Chen J, Zheng Y, Wang Z, Gao Q, Hao K, Chen X, Ke N, Lv X, Weng J, Zhong Y, Huang Z, Fu M, Zhao L, Lin F, Mi H, Tang H, Yu C, Huang Y. Development a glycosylated extracellular vesicle-derived miRNA Signature for early detection of esophageal squamous cell carcinoma. BMC Med 2025; 23:39. [PMID: 39849483 PMCID: PMC11755925 DOI: 10.1186/s12916-025-03871-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is often diagnosed at an advanced stage due to the lack of non-invasive early detection tools, which significantly impacts patient prognosis. Given that glycosylation alterations especially high sialylation and fucosylation, frequently occur during cellular malignant transformation, but their roles are not elucidated. We examined alterations in disease-specific glycosylated extracellular vesicles (EVs)-derived miRNAs in the serum of ESCC patients, evaluating their utility as diagnostic biomarkers. METHODS A total of 371 ESCC and 303 healthy controls (HCs) were recruited in this multi-stage, multicentre case-control study. Fucosylated (Fuc-) and sialylated (Sia-) EVs were isolated utilizing Lentil lectin (LCA) and wheat germ lectin (WGA)-coated magnetic beads, respectively. The glycosylated EVs-derived miRNAs-based signature (RiskscoreFuc-&Sia-) was established through logistic regression in a training cohort and subsequently validated in an internal and an external multicentre cohort. RESULTS The RiskscoreFuc-&Sia- effectively identified ESCC across all stages, demonstrating high AUC values in training (0.980), internal validation (0.957), and external multicentre validation (0.973) cohorts, markedly higher than carcinoembryonic antigen (CEA) (AUC = 0.769, training cohort; AUC = 0.749, internal validation cohort; AUC = 0.765, external validation cohort). Notably, this score exhibited robust accuracy in detecting CEA (-) ESCC cases (CEA < 5 ng/ml) (AUC = 0.974, training & internal cohort; AUC = 0.973, external multicentre validation cohort). Additionally, it displayed strong efficacy in differentiating early-stage ESCC patients (AUC = 0.982, training cohort; AUC = 0.977, external multicentre validation cohort). CONCLUSIONS Our study illustrates the effectiveness of glycosylated EVs capture strategy for isolating tumour-specific EVs. The unique glycosylated EVs-derived miRNAs-based signature shows the optimal potential as a biomarker for early detection of ESCC.
Collapse
Affiliation(s)
- Jianlin Chen
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yue Zheng
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Zhen Wang
- Department of Clinical Laboratory, Shishi Hospital, Fujian, 362700, Shishi, China
| | - Qi Gao
- Research and development center, Beijing Youngen Technology Co. Ltd, Beijing, 102600, People's Republic of China
| | - Kun Hao
- Research and development center, Beijing Hotgen Biotech Co., Ltd, Beijing, 102600, People's Republic of China
| | - Xiongfeng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Scientific Research, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Nantian Ke
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Xiang Lv
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Jiamiao Weng
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yuhong Zhong
- Department of Clinical Laboratory, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, 310009, China
| | - Zhixin Huang
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
- Integrated Chinese and Western Medicine College, Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou, 350108, China
| | - Miao Fu
- Department of Clinical Laboratory, Jinhua Municipal Central Hospital, Zhejiang, 321000, Jinhua, China
| | - Lilan Zhao
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Fan Lin
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
- Fujian Provincial Centre for Geriatrics, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Hui Mi
- Departments of Clinical Laboratory, Changzhi People's Hospital, Shanxi, Changzhi, 046000, China
| | - Haijun Tang
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
| | - Chundong Yu
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Centre for Cell Signalling Network, School of Life Sciences, Xiamen University, Fujian, Xiamen, 361102, China.
| | - Yi Huang
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- Central Laboratory, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian, Fuzhou, 350001, China.
| |
Collapse
|
34
|
Wang Z, Xiao Z, Zhang T, Lu M, Li H, Cao J, Zheng J, Zhou Y, Dai J, Wang C, Chen L, Xu J. Development and validation of a novel artificial intelligence algorithm for precise prediction the postoperative prognosis of esophageal squamous cell carcinoma. BMC Cancer 2025; 25:134. [PMID: 39849452 PMCID: PMC11756118 DOI: 10.1186/s12885-025-13520-6] [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: 04/28/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a highly aggressive malignancy, and current postoperative prognostic assessment methods remain unsatisfactory, underlining the urgent to develop a reliable approach for precision medicine. Given the similarities with gametogenesis, cancer/testis genes (CTGs) are acknowledged for regulation unrestrained multiplication and immune microenvironment during oncogenic processes. These processes are associated with advanced disease and poorer prognosis, indicating that CTGs could serve as ideal prognostic biomarkers in ESCC. The purpose of this study is to develop a novel clinically prognostic prediction system to facilitate the individualized postoperative care. METHODS We conducted LASSO regression analysis of protein-coding CTGs and clinical characteristics from 119 pathologically confirmed ESCC patients to recognize powerful predictive variables. We employed nine supervised machine learning classifiers and integrated best predictive machine learning classifiers by weighted voting method to construct an ensemble model called PPMESCC. Additionally, functional assay was conducted to examine the potential effect of top-ranking CTG HENMT1 in ESCC. RESULTS LASSO regression identified five CTGs and TNM stage as optimized prognostic features. Six machine learning classifiers were integrated to construct an ensemble model, PPMESCC, which exhibited outstanding performance in ESCC prediction. The AUC for PPMESCC was 0.9828 (95% confidence interval: 0.9608 to 0.9926), with an accuracy of 98.32% (95% CI: 96.64-99.16%) in the discovery cohort and 0.9057 (95% CI: 0.8897 to 0.9583) of AUC with an accuracy of 90% (95% CI: 89.08-93.28%) in validation cohort. In addition, the top-ranking CTG HENMT1 encodes 2'-O-methyltransferase of piRNAs that was confirmed positively correlated with the proliferation capacity of ESCC cells. Then we systematically screen piRNAs associated with esophageal carcinoma based on GWAS, eQTL-piRNA, and i2OM databases, and successfully discovered 8 piRNAs potentially regulated by HENMT1. CONCLUSION The study highlights the clinical utility of PPMESCC algorithm in prognostic prediction that may facilitate to establish the personalized screening and management strategies for postoperative ESCC patients.
Collapse
Affiliation(s)
- Zichen Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Zhihan Xiao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Tongyu Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Meiyou Lu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Hai Li
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jing Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jianan Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yichan Zhou
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, No. 101, Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Cheng Wang
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, No. 101, Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jing Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
35
|
Wang Y, Sun H, Li G, Xu J, Wang S, Zhang S, Zhou T, Han T, Sun C, Ma J, Wang X, Yin H. Impact of maternal and offspring smoking and breastfeeding on oesophageal cancer in adult offspring. Nat Commun 2025; 16:938. [PMID: 39843420 PMCID: PMC11754477 DOI: 10.1038/s41467-025-56252-8] [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: 05/06/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Numerous risk factors for oesophageal cancer are linked to lifestyle habits, but the role of early-life factors in its incidence and mortality is unclear. Using UK Biobank data, we explore the association among breastfeeding, maternal smoking, smoking in offspring, and oesophageal cancer risk in adult offspring via multivariable Cox regression. Here, we show that being breastfed, compared with not being breastfed, is associated with a lower risk of oesophageal cancer incidence (HR: 0.83, 95% CI: 0.70-0.98) and mortality (HR: 0.74, 95% CI: 0.61-0.89) in adult offspring. Additionally, it is associated with a reduced impact of smoking in offspring on oesophageal cancer incidence (HR: 0.79, 95% CI: 0.64-0.96) and mortality (HR: 0.73, 95% CI: 0.59-0.91). We subsequently construct a polygenic risk score for oesophageal cancer to explore the influence of genetic factors. Our findings emphasize the importance of breastfeeding, and smoking cessation to prevent oesophageal cancer.
Collapse
Affiliation(s)
- Yixue Wang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hongru Sun
- Department of Epidemiology, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Gen Li
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Jingxue Xu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Siyu Wang
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Shijie Zhang
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Tianle Zhou
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Tianshu Han
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jianqun Ma
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Xiaoyuan Wang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Hang Yin
- Department of Radiation Therapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
36
|
Cao W, Jin D, Min W, Li H, Wang R, Zhang J, Gou Y. Prognostic values of intracellular cell-related genes in esophageal cancer and their regulatory mechanisms. BMC Cancer 2025; 25:105. [PMID: 39833728 PMCID: PMC11744837 DOI: 10.1186/s12885-025-13483-8] [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: 05/22/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Esophageal cancer is a grave malignant condition. While radiotherapy, often in conjunction with chemotherapy, serves as a cornerstone in the management of locally advanced or metastatic cases, patient tolerance and treatment resistance frequently hinder its efficacy. Cell-in-cell structures, prevalent in various tumors, have been linked to prognosis. Hence, investigating the prognostic significance and regulatory mechanisms of genes related to these intracellular structures in esophageal cancer is imperative. The Cancer Genome Atlas (TCGA) Esophageal Cancer (ESCA) dataset served as the training set for the analysis. Differentially expressed genes (DEGs) in ESCA samples were identified, with those related to intercellular structures designated cell-in-cell-related differential expression genes (CIC-related DEGs). Cox regression analysis was employed to identify prognostic genes, categorizing samples into high- and low-risk groups based on median risk scores. Validation was conducted using the GSE53624 risk model. Established methodologies included morphological mapping, enrichment analysis, immune infiltration analysis, prognostic gene expression validation, molecular docking, and Reverse Transcription Polymerase Chain Reaction (RT-PCR) validation. Thirty-eight intersecting genes were identified between the disease and normal groups in ESCA samples. Stepwise multivariate Cox analysis pinpointed three prognostic genes: androgen receptor (AR), C-X-C motif chemokine ligand 8 (CXCL8), and epidermal growth factor receptor (EGFR). The risk model's applicability was confirmed in the GSE53624 dataset, revealing eight significantly different immune-related gene sets. Prognostic gene expression validation demonstrated significant differences between the disease and normal groups in both datasets. The proteins corresponding to the three prognostic genes interacted with gefitinib and osimertinib. RT-PCR results corroborated the differential expression of prognostic genes in esophageal cancer tissues. This study identified AR, CXCL8, and EGFR as prognostic genes and demonstrated their molecular interactions with gefitinib and osimertinib, providing a foundation for ESCA diagnosis and treatment.
Collapse
Affiliation(s)
- Wei Cao
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Dacheng Jin
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Weirun Min
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Haochi Li
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Rong Wang
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jinlong Zhang
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Yunjiu Gou
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China.
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China.
| |
Collapse
|
37
|
Ardalan Moghadam Al F, Forghanifard MM, Zarrinpour V. PYGO2 promotes resistance to chemotherapy via reducing apoptosis and G2/M cell cycle arrest in esophageal carcinoma cells. Med Oncol 2025; 42:45. [PMID: 39808374 DOI: 10.1007/s12032-024-02590-4] [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: 10/17/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
5-FU is a widely used chemotherapy drug for esophageal carcinomas, but therapy failure has been observed in 5-FU-resistant patients. Overcoming this resistance is a significant challenge in cancer treatment, requiring identifying and targeting important resistance mechanisms. PYGO2 expression is crucial in developing resistance to various chemotherapy drugs. In this study, we aimed to investigate the impact of PYGO2 overexpression on the sensitivity of YM-1 and KYSE-30 esophageal carcinoma cells against 5-FU. To do this, we compared cell viability, cell cycle arrest, apoptosis rate, and mRNA expressions of various apoptosis-related genes between pcDNA3-PYGO2 transfected and untransfected KYSE-30 and YM-1 esophageal carcinoma cells following treatment with 5-FU. We showed that PYGO2 expression reduces 5-FU sensitivity in YM-1 and KYSE-30 cells. PYGO2-overexpressing cells treated with 5-FU have exhibited a noteworthy reduction in both early and late apoptotic cells compared to controls. Furthermore, a significant decrease in the Bax/Bcl2 ratio and P53 gene expression was observed. 5-FU induces G2/M cell cycle arrest in YM-1 and KYSE-30 cells. However, PYGO2 overexpression impeded G2/M cell cycle arrest in 5-FU-treated cells, thereby suppressing the toxicity of 5-FU. PYGO2 may mediate its apoptotic effect by regulating cell cycle regulatory proteins, specifically cyclin D1 and p21. These results highlight PYGO2's capacity to alter how esophageal cancer cells respond to 5-FU therapy, emphasizing its importance as a potential focal point for treatment strategies.
Collapse
Affiliation(s)
| | | | - Vajiheh Zarrinpour
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
38
|
Strzelec B, Chmielewski PP, Taboła R. Induction Radiochemotherapy for Esophageal Cancer: Long-Term Outcomes from a Single-Center Study. J Clin Med 2025; 14:394. [PMID: 39860400 PMCID: PMC11766012 DOI: 10.3390/jcm14020394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The management of esophageal cancer (EC) remains a significant clinical challenge, particularly in optimizing therapeutic strategies for different stages and subgroups. This study assessed the impact of preoperative radiochemotherapy (CRT) on clinical staging and identified subgroups for whom definitive CRT (dCRT) may provide a favorable alternative to surgery. Methods: Sixty-one patients with esophageal adenocarcinoma or squamous cell carcinoma were enrolled. Pre-treatment staging included computed tomography, gastroscopy with biopsy, and comprehensive laboratory evaluations. Patients received preoperative CRT following the CROSS or dCRT protocols based on tumor stage. Surgical approaches included staged esophagectomy or single-stage Ivor Lewis procedures. Four patients declined surgery and were treated with dCRT. Postoperative outcomes were evaluated using pTNM classification. Follow-up included imaging and endoscopic surveillance. Statistical analyses assessed changes in staging and factors influencing treatment outcomes. Results: CRT significantly reduced T stage across the entire cohort (p = 0.0002), with complete pathological response (pT0N0M0) observed in 54.5% of patients following induction CRT (p = 0.0001). Male patients demonstrated a significant reduction in T stage (p = 0.0008), while a similar trend in females was not significant (p = 0.068). Among patients declining surgery, dCRT demonstrated acceptable oncologic control over a mean follow-up of 4 ± 0.79 years. Conclusions: Preoperative CRT effectively downstages EC and achieves high rates of response, especially in male patients. Therefore, dCRT may be a viable alternative in selected patients, emphasizing the need for individualized treatment strategies to optimize outcomes. These findings underscore the importance of refining multimodal approaches in EC care.
Collapse
Affiliation(s)
- Bartłomiej Strzelec
- 2nd Department of General Surgery and Surgical Oncology, Medical University Hospital, 50-556 Wroclaw, Poland; (B.S.)
| | - Piotr Paweł Chmielewski
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 6a Chalubinskiego Street, 50-368 Wroclaw, Poland
| | - Renata Taboła
- 2nd Department of General Surgery and Surgical Oncology, Medical University Hospital, 50-556 Wroclaw, Poland; (B.S.)
| |
Collapse
|
39
|
Qian C, Zhang X, Tian YS, Yuan L, Wei Q, Yang Y, Xu M, Wang X, Sun M. Coptisine inhibits esophageal carcinoma growth by modulating pyroptosis via inhibition of HGF/c-Met signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03765-6. [PMID: 39792166 DOI: 10.1007/s00210-024-03765-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Esophageal carcinoma is a highly prevalent malignancy worldwide. The present study aimed to investigate the mechanism by which the natural compound coptisine affects pyroptosis in esophageal squamous cell carcinoma (ESCC). The expression of c-Met in ESCC patients was assessed by immunohistochemical analysis of tissue microarrays. Natural drugs that bind to c-Met were identified by screening and molecular docking. The effect of coptisine on the proliferation of ESCC cells was detected by CCK-8 and colony formation assays. Cell cycle progression and cell apoptosis were detected by flow cytometry. The levels of mRNAs related to pyroptosis and miR-21 after coptisine treatment were assessed via real-time quantitative PCR. The effect of pyroptosis was evaluated by reactive oxygen species level detection and transmission electron microscopy (TEM) analysis. The expression of proteins related to pyroptosis and the HGF/c-Met pathway was detected by western blotting. A xenograft tumor model was established, and the inhibitory effect of coptisine was evaluated by observing tumor growth. The results showed that the highly expressed protein c-Met in esophageal cancer could bind with coptisine. Coptisine inhibited c-Met phosphorylation and proliferation in ESCC cells. Furthermore, coptisine inhibited the expression of downstream proteins of the HGF/c-Met signaling pathway and induced ROS generation. Tumor xenograft experiments demonstrated that coptisine effectively inhibited tumor growth by reducing the levels of pyroptosis-associated proteins. In conclusion, these findings indicate that inhibition of the HGF/c-Met signaling pathway suppresses pyroptosis to enhance the antitumor effect of coptisine in ESCC and support the potential use of coptisine for EC treatment.
Collapse
Affiliation(s)
- Chunmei Qian
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xing Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu-Shi Tian
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Lin Yuan
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qiao Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yifu Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Institute of Pathology, Fudan University, Shanghai, 200032, China.
| | - Xiaoyu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Menghong Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
40
|
Wang A, Wang Y, Ma Q, Chen X. The carcinogenesis of esophageal squamous cell cancer is positively regulated by USP13 through WISP1 deubiquitination. Biofactors 2025; 51:e2139. [PMID: 39468941 DOI: 10.1002/biof.2139] [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: 07/23/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024]
Abstract
The objective was to determine whether USP13 stabilizes WISP1 protein and contributes to tumorigenicity and metastasis in ESCC through the Wnt/CTNNB1 signaling pathway. ESCC cell lines (KYSE150 and TE10) were treated with the proteasome inhibitor MG-132, followed by siRNA screening of deubiquitinases (DUBs) to identify regulators of WISP1. Mass spectrometry, immunoprecipitation, and in vitro functional assays were conducted to explore the interaction between USP13 and WISP1 and to assess the effects of USP13 downregulation on cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), and apoptosis. Additionally, in vivo experiments using mouse models were performed to evaluate the impact of USP13 knockdown on tumor growth and metastasis. USP13 was identified as a key regulator of WISP1, stabilizing its protein levels through deubiquitination. Downregulation of USP13 resulted in reduced WISP1 protein stability, decreased cell proliferation, migration, and EMT, and increased apoptosis in vitro. In vivo, USP13 knockdown significantly inhibited tumor growth and lung metastasis. WISP1 overexpression in USP13-knockdown cells partially rescued these phenotypes, confirming the functional role of the USP13/WISP1 axis. Furthermore, knockdown of USP13 or WISP1 impaired the activation of the Wnt/CTNNB1 signaling pathway and reduced immune checkpoint marker expression, indicating a mechanism by which USP13 promotes immune evasion in ESCC. USP13 stabilizes WISP1 through deubiquitination, enhancing ESCC progression by activating the Wnt/CTNNB1 pathway and promoting immune evasion, making USP13 a potential therapeutic target in ESCC.
Collapse
Affiliation(s)
- An Wang
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Youbo Wang
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Qinyun Ma
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaofeng Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Liu Z, Kim MO. Evolving Therapeutic Strategies in Esophageal Squamous Cell Carcinoma: Advances and Perspectives. J Cancer Prev 2024; 29:99-104. [PMID: 39790226 PMCID: PMC11706721 DOI: 10.15430/jcp.24.026] [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: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is among the most prevalent forms of esophageal cancer globally, with a particularly high incidence in developing countries. Notably, Asia accounts for approximately 80% of global esophageal cancer cases, with China alone contributing to 54% of this burden. The primary treatment modality for ESCC remains esophagectomy, primarily employed for locally advanced disease, often in combination with chemotherapy and radiotherapy for advanced-stage cases. Despite significant advancements in surgical techniques and the advent of precision medicine, which has facilitated the development of targeted and immune-based therapies, critical challenges persist, including suboptimal therapeutic efficacy and the emergence of drug resistance. A comprehensive understanding of the current treatment landscape for ESCC is essential to overcoming these barriers and improving patient outcomes.
Collapse
Affiliation(s)
- Zhibin Liu
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| |
Collapse
|
42
|
Yu Y, Xia L, Wang Z, Zhu T, Zhao L, Fan S. A cross-cohort study identifies potential oral microbial markers for esophageal squamous cell carcinoma. iScience 2024; 27:111453. [PMID: 39758985 PMCID: PMC11699290 DOI: 10.1016/j.isci.2024.111453] [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: 06/04/2024] [Revised: 08/13/2024] [Accepted: 11/19/2024] [Indexed: 01/07/2025] Open
Abstract
Current screening methods for esophageal squamous cell carcinoma (ESCC) face challenges such as low patient compliance and high costs. This study aimed to develop a model based on oral microbiome data for identifying ESCC. By analyzing 249 oral flora samples, we identified microbial markers associated with ESCC and constructed random forest classifiers that distinguished patients with ESCC from controls, achieving an area under the ROC curve (AUC) of 0.87. Key ESCC-associated microbial markers included Neisseria perflava and Haemophilus parainfluenzae. The classifier was validated within the cohort, attaining an AUC of 0.93. For comparison, traditional tumor markers carcinoembryonic antigen (CEA) and squamous cell carcinoma antigen (SCC-Ag) yielded AUCs of 0.84. Functional analysis identified pathways linked to ESCC, such as glycerol degradation and nitrate reduction. This study suggests a potential noninvasive method for detecting ESCC, offering a more accessible and accurate alternative to current screening methods.
Collapse
Affiliation(s)
- Yanxiang Yu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Lei Xia
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 401336, China
| | - Zhouxuan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Tong Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Lujun Zhao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
43
|
Akbari A, Adabi M, Masoodi M, Namazi A, Mansouri F, Tabaeian SP, Shokati Eshkiki Z. Artificial intelligence: clinical applications and future advancement in gastrointestinal cancers. Front Artif Intell 2024; 7:1446693. [PMID: 39764458 PMCID: PMC11701808 DOI: 10.3389/frai.2024.1446693] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 12/02/2024] [Indexed: 04/01/2025] Open
Abstract
One of the foremost causes of global healthcare burden is cancer of the gastrointestinal tract. The medical records, lab results, radiographs, endoscopic images, tissue samples, and medical histories of patients with gastrointestinal malignancies provide an enormous amount of medical data. There are encouraging signs that the advent of artificial intelligence could enhance the treatment of gastrointestinal issues with this data. Deep learning algorithms can swiftly and effectively analyze unstructured, high-dimensional data, including texts, images, and waveforms, while advanced machine learning approaches could reveal new insights into disease risk factors and phenotypes. In summary, artificial intelligence has the potential to revolutionize various features of gastrointestinal cancer care, such as early detection, diagnosis, therapy, and prognosis. This paper highlights some of the many potential applications of artificial intelligence in this domain. Additionally, we discuss the present state of the discipline and its potential future developments.
Collapse
Affiliation(s)
- Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Adabi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohsen Masoodi
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Namazi
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mansouri
- Department of Microbiology, Faculty of Sciences, Qom Branch, Islamic Azad University, Qom, Iran
| | - Seidamir Pasha Tabaeian
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Shokati Eshkiki
- Alimentary Tract Research Center, Clinical Sciences Research Institute, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
44
|
Hua YY, Kuo MC, Chen YH, Lu HI, Lo CM, Chen Y, Wang YM, Lin YH, Li SH, Huang SY. Hypoalbuminemia is Associated with Higher 90-Day Mortality and Poor Prognosis in Patients with Esophageal Squamous Cell Carcinoma and Liver Cirrhosis Receiving Radiotherapy-Based Therapy. Cancer Manag Res 2024; 16:1693-1704. [PMID: 39649944 PMCID: PMC11624664 DOI: 10.2147/cmar.s488998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/11/2024] Open
Abstract
Background Liver cirrhosis (LC) is common among patients with esophageal squamous cell carcinoma (ESCC) due to shared etiologic factor, alcohol. For non-metastatic ESCC (nmESCC) patients with cirrhosis, surgery is often contraindicated or associated with high morbidity, therefore radiotherapy-based therapy was commonly applied. This study aims to investigate prognosticators for overall survival (OS) in nmESCC and cirrhotic patients receiving radiotherapy-based therapy. Furthermore, we will also evaluate the predictors for the 90-day mortality rate to reduce avoidable treatment-related toxic effects, and prevent medical waste. Methods Between January 2001 and December 2021, we retrospectively reviewed medical records of 1298 ESCC patients. Total 78 patients with nmESCC and liver cirrhosis identified based on abdominal ultrasonography, computerized tomography, or liver biopsy were enrolled. Clinicopathologic parameters were collected and correlated with OS and 90-day mortality. Results Univariate analysis revealed that Child-Pugh classification B/C (P<0.001, versus A), radiotherapy alone (P=0.03, versus chemoradiotherapy), prothrombin time (PT) prolonged≧2 seconds (P=0.024), albumin≦3.5g/dl (P<0.001), controlled/refractory ascites (P=0.01, versus none of ascites), and total bilirubin≧1.5mg/dl (P=0.004) were significantly associated with inferior OS. In multivariate analyses, albumin≦3.5g/dl (P=0.001, odds ratio (OR): 2.500) and total bilirubin≧1.5mg/dl (P=0.019, OR: 2.012) were independent adverse prognosticators. The 90-day mortality in these 78 patients receiving radiotherapy-based therapy was 10.3% (n=8), including ESCC progression in 4 patients, liver failure in 1 patient, and others in 3. Clinical 8th American Joint Committee on Cancer (AJCC) stage IVA (P=0.02), clinical T classification T3/4 (P=0.049), PT prolonged≧4 seconds (P=0.009), and albumin≦3.5g/dl (P=0.027) were significantly correlated with higher 90-day mortality. Logistic model showed albumin≦3.5g/dl (P=0.015, OR: 16.129) and clinical 8th AJCC stage IVA (P=0.012, OR: 17.544) were independently correlated with higher 90-day mortality. Conclusion Hypoalbuminemia is associated with higher 90-day mortality and poor prognosis in nmESCC and liver cirrhosis patients receiving radiotherapy-based therapy.
Collapse
Affiliation(s)
- Yu-Yang Hua
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ming-Chun Kuo
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Hao Chen
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-I Lu
- Department of Thoracic & Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Ming Lo
- Department of Thoracic & Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu Chen
- Department of Thoracic & Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Ming Wang
- Department of Radiation Oncology & Proton and Radiation Therapy Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yun-Hsuan Lin
- Department of Radiation Oncology & Proton and Radiation Therapy Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shau-Hsuan Li
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Yu Huang
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
45
|
Fredimoses M, Li P, Zhang Y, Jia H, Liu S, Tian J, Nie W, Liu K, Song M, Dong Z. Design, synthesis, and antiproliferative activity evaluation of novel α-mangostin derivatives by ROS/MAPK signaling pathway. Bioorg Chem 2024; 153:107968. [PMID: 39566271 DOI: 10.1016/j.bioorg.2024.107968] [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: 08/26/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
Novel hydroxamic acid and 3,6-amide modified α-mangostin derivatives were synthesized and evaluated their antiproliferative activities against KYSE 30 (esophageal cancer), HCT 116 (colon cancer), and HGC 27 (gastric cancer) cell lines. Most of the new derivatives displayed stronger anti-proliferative activities compared to α-mangostin. Among all the derivatives, compound 4a exhibited the most potent activity, with IC50 values of 0.57 ± 0.29 μM, 3.27 ± 0.16 μM, and 2.28 ± 1.02 μM against KYSE 30, HCT 116, and HGC 27 cells, respectively. Subsequent mechanism studies revealed that compound 4a inhibited cancer cells proliferation and colonies formation in a concentration-dependent manner. Additionally, compound 4a caused cell cycle arrest in a p53 dependent manner and induced apoptosis in p53 independent way. Meanwhile, 4a suppressed cell cycle related proteins (Cyclin D1 and cyclin B1) expression, increased pro-apoptotic proteins (cleaved PARP, cleaved caspase-7, and cleaved caspase-9) and decreased anti-apoptotic proteins (Bcl-2) expression. Moreover, 4a increased reactive oxygen species (ROS) levels in KYSE 30 cells and upregulated the expression of proteins related to the ROS related MAPK signaling pathway (p-ERK, p-p38, and p-JNK). These findings suggest that compound 4a holds promising potential as an antiproliferative agent by targeting MAPK signaling pathway to inhibit cell cycle progress, induce apoptosis and produce ROS in cancers.
Collapse
Affiliation(s)
- Mangaladoss Fredimoses
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China
| | - Pan Li
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China
| | - Yunqing Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Huajie Jia
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shihui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jie Tian
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Wenna Nie
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China; Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou 450000, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengqiu Song
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China; Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China; The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou 450000, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
46
|
Ling D, Jiang T, Sun J, Wang Y, Wang Y, Wang L. An Ensemble Learning System Based on Stacking Strategy for Survival Risk Prediction of Patients with Esophageal Cancer. Ing Rech Biomed 2024; 45:100860. [DOI: 10.1016/j.irbm.2024.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
47
|
Huang G, Xu J, Li Y, Song L, Wen C, Ruan Q, Wen Z, Qi J, Deng J, Liu Y. Corynoxine exerts the anti-tumor effect on esophageal squamous cell carcinoma principally via the EZH2-DUSP5-ERK1/2-mediated cell growth inhibition. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156103. [PMID: 39383633 DOI: 10.1016/j.phymed.2024.156103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Esophageal cancer is one of the most prevalent malignant tumors and the sixth largest cause of tumor-associated death worldwide. Squamous cell carcinoma (ESCC) accounts for 85 % of all esophageal cancer cases. ESCC treatment remains to be significantly difficult. Corynoxine (Cory) is a tetracyclic hydroxyindole alkaloid isolated from Uncaria macrophylla. It is unclear whether Cory has an anti-tumor effect on ESCC. PURPOSE To determine the anti-tumor activity of Cory and the associated mechanisms in ESCC. STUDY DESIGN Cory's effects on proliferation, apoptosis, migration, and invasion, as well as the underlying molecular causes were assessed using two ESCC cell lines, KYSE150 and TE-1. A xenograft mouse model was then applied to evaluate the anti-tumor activity of Cory in vivo. METHODS Western blot, assays including CCK-8, colony formation, EdU staining, TUNEL staining, cell scratch and Transwell, and a xenograft mouse model were used in this study. RESULTS Cory suppressed cell growth, provoked cell apoptosis, and hindered cell migration and invasion of ESCC cells. DUSP5 knockdown reduced the Cory-induced cell death and restored cell migration and invasion through ERK1/2 activation. Further analyses showed that Cory promoted DUSP5 expression via inhibiting EZH2 expression, leading to inactivation of ERK1/2 signaling and the subsequent cell growth inhibition of ESCC. In vivo experiments disclosed that Cory suppressed tumor growth of ESCC through upregulating DUSP5 expression. CONCLUSIONS Cory plays an anti-tumor role in ESCC by regulating EZH2-DUSP5-ERK1/2 signaling pathway. Cory may be promising to be a novel therapy for treating ESCC.
Collapse
Affiliation(s)
- Gang Huang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; The People's Hospital of Beilun District, Ningbo 315000, China
| | - Jiale Xu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yingchao Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Liangtao Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chunmei Wen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Qingqing Ruan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Zhikai Wen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035 China
| | - Jinxia Qi
- Biobank, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jie Deng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Yu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
48
|
Gao J, Li M, Wang Y, Wang Z, Chen X, Li H. Prognostic Effect of the PNI and LSR in Patients with Esophageal Squamous Cell Carcinoma Patients Receiving Radiotherapy. J Gastrointest Cancer 2024; 56:26. [PMID: 39601941 DOI: 10.1007/s12029-024-01148-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] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
PURPOSE The prognostic nutritional index (PNI) has been used to assess the immunonutritional status of cancer patients and can predict the prognosis of various solid cancers, and the serum alanine transaminase (ALT)/aspartate transaminase (AST) ratio (LSR) is considered a good predictor of liver injury. A retrospective cohort analysis was conducted to investigate the relationship between the prognosis of esophageal squamous cell carcinoma (ESCC) patients and LSR or PNI, as well as to combine these two indicators (LSR-PNI) for further prognostic analysis in ESCC patients undergoing radiotherapy (RT). METHODS In this study, 134 patients with esophageal cancer were retrospectively analyzed. The Chi-square test was utilized to compare count data, and univariate and multivariate Cox proportional hazards models were employed to identify independent risk and prognostic factors. Additionally, the combination of LSR and PNI (LSR-PNI) was analyzed. RESULTS This study included a cohort of 134 patients, comprising 105 males with a mean age of 70.7 years and 29 females with a mean age of 76.3 years. Pathological examination categorized 41 cases as stage I-II and 93 cases as stage III-IV. The predominant treatment modality administered was intensity-modulated radiotherapy (IMRT) for esophageal cancer. Of these patients, 96 received radiation doses ≤ 54 Gy, while 38 were administered doses > 54 Gy. Radiation-induced adverse effects were observed in 67 patients, with the remaining 67 showing no such effects. Kaplan-Meier survival analysis revealed that elevated levels of the lymphocyte-to-serum ratio (LSR) and prognostic nutritional index (PNI) were significantly correlated with improved progression-free survival (PFS) and overall survival (OS). The high-LSR group demonstrated longer PFS (14.4 vs. 9.3 months, p = 0.0469) and OS (19.9 vs. 13.7 months, p = 0.0315) compared to the low-LSR group, with respective 3-year survival rates of 18.4% vs. 12.7%. Similarly, patients in the high-PNI group exhibited superior PFS (13.9 vs. 8.9 months, p = 0.0071) and OS (19.0 vs. 13.5 months, p = 0.0002) compared to the low-PNI group, with 3-year survival rates of 19.6% vs. 11.3%. Stratification based on combined LSR and PNI levels categorized patients into low-, intermediate-, and high-risk groups. The low-risk group demonstrated significantly better PFS (17.8 vs. 10.1 vs. 8.2 months) and OS (24.1 vs. 14.3 vs. 12.9 months, p < 0.0001) compared to the intermediate- and high-risk groups, with 3-year survival rates of 24%, 14%, and 10.3%, respectively. CONCLUSION Pretreatment LSR and PNI can serve as independent prognostic predictors for patients, with higher values of both being associated with improved progression-free survival and overall survival. Additionally, the combined LSR-PNI score effectively stratifies patients into distinct risk groups, offering a robust tool for predicting outcomes in clinical practice.
Collapse
Affiliation(s)
- Junfeng Gao
- Anhui Medical University, Hefei, 230032, China
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230041, China
| | - Meimei Li
- Anhui Medical University, Hefei, 230032, China
| | - Yi Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230041, China
| | - Ziming Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230041, China
| | - Xue Chen
- Bengbu Medical College, Bengbu, 233030, China
| | - Hongxia Li
- Anhui Medical University, Hefei, 230032, China.
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230041, China.
| |
Collapse
|
49
|
Gan L, Zhou L, Chu AL, Sun C, Wang Y, Yang M, Liu Z. GPD1L may inhibit the development of esophageal squamous cell carcinoma through the PI3K/AKT signaling pathway: bioinformatics analysis and experimental exploration. Mol Biol Rep 2024; 51:1149. [PMID: 39535578 DOI: 10.1007/s11033-024-10070-1] [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: 08/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Esophageal squamous carcinoma (ESCC) is the most prevalent pathological subtype of esophageal cancer (EC). It has the characteristics of significant local invasion, quick disease progression, high recurrence rates, and a dismal prognosis for survival. Phosphatidylinositol 3-kinase/serine-threonine kinase (PI3K/AKT) is a signaling system whose aberrant activation regulates downstream factors, leading to the promotion of cancer development. This study looks at a protein called Glycerol-3-phosphate dehydrogenase 1-like (GPD1L), which strongly affects the development of several cancers. However, its association with ESCC development and its underlying mechanisms are not clear. METHODS In this paper, we analyzed six ESCC transcriptome data obtained from the GEO database. We utilized bioinformatics technology and immunohistochemistry to differentially analyze GPD1L levels of mRNA and protein expression in ESCC and normal adjacent tissues. Furthermore, we conducted survival, co-expression, enrichment, immune infiltration and drug sensitivity analysis. Moreover, we further investigated the role and mechanism of GPD1L by Western Blot (WB), Cell Counting Kit-8 (CCK8), wound healing assay, Transwell assay, and flow cytometry. Finally, the addition of IGF-1, the activator of PI3K/AKT, could rescue the inhibitory effect of GPD1L on ESCC. RESULTS The findings manifest that the expression of GPD1L was low in ESCC, and functional experiments showed that GPD1L promoted apoptosis in vitro while blocking cell migration, invasion, and proliferation. Based on mechanism research, GPD1L's impact on ESCC could be explained by its suppression of the PI3K/AKT signaling pathway's activation. CONCLUSION To sum up, our findings imply that GPD1L may impede the initiation and advancement of ESCC via modulating the PI3K/AKT signaling pathway. GPD1L is considered to be a promising therapeutic target and biomarker to diagnose and treat ESCC.
Collapse
Affiliation(s)
- LanLan Gan
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
- Tumor Radiotherapy Department, The Second Clinical Medical School of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - Lu Zhou
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
- Tumor Radiotherapy Department, The Second Clinical Medical School of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - ALan Chu
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - Chen Sun
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - YongTai Wang
- Tumor Radiotherapy Department, The Second Clinical Medical School of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - MengLin Yang
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China
| | - ZongWen Liu
- Tumor Radiotherapy Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, P.R. China.
| |
Collapse
|
50
|
Dou L, Liu Y, Zha B, Zhu J, Zhang Y, He S, Wang G. Retrospective study on endoscopic treatment of recurrent esophageal cancer patients after radiotherapy. Surg Endosc 2024; 38:6637-6642. [PMID: 39294315 DOI: 10.1007/s00464-024-11259-y] [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: 07/10/2024] [Accepted: 08/31/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Esophageal cancer poses a significant health burden globally. Endoscopic treatment has emerged as a viable option for patient ineligible for surgery or experiencing disease recurrence post-radiotherapy. METHODS Patients visiting the Department of Endoscopy at the Cancer Hospital of China Academy of Medical Sciences between March 2009 and March 2024 were retrospectively analyzed. Inclusion criteria encompassed patients with histologically confirmed esophageal cancer who had not undergone surgery, but received radiotherapy or CRT, and subsequently opted for endoscopic treatment. Data on demographics, treatment modalities, recurrence patterns, histopathological characteristics, and outcomes were collected. Statistical analysis was conducted using SPSS 27.0, employing Kolmogorov-Smirnov tests for data normality assessment. RESULTS Out of 25 included patients, the mean age was 60.29 years, with a predominance of males (88%). Most patients (64%) received chemoradiotherapy (CRT), while the rest underwent radiotherapy alone. The median follow-up duration was 50.92 months, with a median recurrence time of 38.92 months. Majority (56%) presented with a solitary lesion and 76% had negative margins. Histopathological analysis revealed various stages of cancer, with the most common being high-grade squamous epithelial neoplasia (64%). Survival analysis indicated a 72% overall survival rate, with 16% surviving beyond 5-year post-treatment. Approximately, 20% succumbed during the study, primarily due to non-esophageal causes (16%). CONCLUSION Endoscopic treatment shows promise as a therapeutic option for selected esophageal cancer patients, offering favorable outcomes in terms of survival and disease control. Further prospective studies are warranted to validate these findings and optimize patient selection criteria for endoscopic interventions in esophageal cancer management.
Collapse
Affiliation(s)
- Lizhou Dou
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Yong Liu
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Bowen Zha
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Jiqing Zhu
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Yueming Zhang
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Shun He
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China.
| | - Guiqi Wang
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China.
| |
Collapse
|