1
|
Xu K, Yu M, Sun Q, Zhang L, Qian X, Su D, Gong J, Shang J, Lin Y, Li X. Cost-effectiveness of PD-1 inhibitors combined with chemotherapy for first-line treatment of oesophageal squamous cell carcinoma in China: a comprehensive analysis. Ann Med 2025; 57:2482019. [PMID: 40131366 PMCID: PMC11938309 DOI: 10.1080/07853890.2025.2482019] [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: 10/27/2024] [Revised: 02/20/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Programmed death-1 (PD-1) inhibitors combined with chemotherapy have become a standard first-line treatment for advanced oesophageal squamous cell carcinoma (ESCC). Given the high costs associated with immunotherapy, evaluating the cost-effectiveness of different PD-1 inhibitors in the Chinese healthcare setting is essential for guiding treatment decisions and policy development. METHODS A cost-effectiveness analysis was conducted comparing six PD-1 inhibitors-sintilimab, toripalimab, tislelizumab, camrelizumab, serplulimab, and pembrolizumab-combined with chemotherapy for first-line treatment of advanced ESCC. A partitioned survival model was used to calculate incremental cost-effectiveness ratios (ICERs) from healthcare system perspective, with a willingness-to-pay (WTP) threshold set at $36,598.19 per quality-adjusted life year (QALY). Sensitivity analyses were performed to evaluate the robustness of the results. RESULTS The ICERs for toripalimab, camrelizumab, pembrolizumab, serplulimab, sintilimab, and tislelizumab were $32,356.79/QALY, $48,410.64/QALY, $312,743.54/QALY, $121,200.84/QALY, $29,663.42/QALY, and $35,304.33/QALY, respectively. Sintilimab, toripalimab, and tislelizumab were below the WTP threshold. Among all regimens, the top three in life years (LYs) gained were toripalimab, serplulimab, and tislelizumab. Sensitivity analysis showed that utility values and drug prices were key factors influencing ICERs. Probabilistic analysis indicated that toripalimab, sintilimab, and tislelizumab had the highest probabilities of being cost-effective, at 83.1%, 81.4%, and 70.0%, respectively. CONCLUSION Sintilimab, toripalimab, and tislelizumab are the most cost-effective PD-1 inhibitors when combined with chemotherapy for the first-line treatment of advanced ESCC in China, with ICERs below the WTP threshold. While all six PD-1 inhibitors demonstrated clinical benefits, pembrolizumab and serplulimab were less favourable from a cost-effectiveness standpoint. Sensitivity analysis confirmed that drug prices and utility values are significant determinants of cost-effectiveness.
Collapse
Affiliation(s)
- Kai Xu
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
- Department of Pharmaceutical Regulatory Science and Pharmacoeconomics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Man Yu
- Department of Pharmaceutical Regulatory Science and Pharmacoeconomics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qingli Sun
- Department of Pharmacy, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
| | - Lingli Zhang
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Xiaodan Qian
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Dan Su
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Jinhong Gong
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Jingjing Shang
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
| | - Yingtao Lin
- Department of Pharmaceutical Regulatory Science and Pharmacoeconomics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- Clinical Medical Research Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Xin Li
- Department of Pharmacy, The Second People’s Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, China
- Department of Pharmaceutical Regulatory Science and Pharmacoeconomics, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of Health Policy, School of Health Policy and Management, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
2
|
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
|
3
|
Tai Y, Kong L, Wang Y, Zhao D, Chen X, Wu Q, Hao J, Wang X, Liu X, Chen D, Li J, Hu Y, Zhang W, Yun CH, Zhan Q. Identification and characterization of Bufalin as a novel EGFR degrader. Cancer Lett 2025; 623:217715. [PMID: 40220852 DOI: 10.1016/j.canlet.2025.217715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/17/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) stands out as a common cancer type worldwide, characterized by its notably high rates of occurrence and mortality. The epidermal growth factor receptor (EGFR) is one of the main targets for cancer treatment as it is one of the genes whose expression is often altered by overexpression, amplification, and mutation in a variety of solid tumors. Substantial efforts have been made to develop EGFR-targeted therapeutic agents, including monoclonal antibodies and tyrosine kinase inhibitors (TKIs). However, these agents exhibited limited efficacy due to the emergence of acquired resistance. Therefore, novel treatment strategies targeting EGFR are urgently needed. Recent studies have identified a few natural compounds that can efficiently inhibit EGFR, indicating that natural products may be potential sources for the development of new EGFR inhibitors. Here, using the Drug Affinity Responsive Target Stability (DARTS) assay combined with liquid chromatography/tandem mass spectrometry analysis, co-crystal method, we discovered that Bufalin directly interacts with EGFR and causes EGFR endocytosis and degradation in the lysosome. Moreover, Bufalin exhibits superior anti-tumor activity compared with another EGFR TKIs. Our study identified Bufalin as the first natural small-molecule EGFR degrader, which suppresses EGFR signaling by inducing the degradation of EGFR via the endosome-lysosome pathway.
Collapse
Affiliation(s)
- Yidi Tai
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lulu Kong
- Department of Biophysics, Department of Integration of Chinese and Western Medicine, and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yan Wang
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Dongyu Zhao
- Soochow University Cancer Institute, Suzhou, 215000, China
| | - Xu Chen
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Qingnan Wu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jia Hao
- Department of Biophysics, Department of Integration of Chinese and Western Medicine, and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Xi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xingyang Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Dongshao Chen
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jinting Li
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Yuying Hu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Weimin Zhang
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Department of Oncology, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, China.
| | - Cai-Hong Yun
- Department of Biophysics, Department of Integration of Chinese and Western Medicine, and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Department of Oncology, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, China; Soochow University Cancer Institute, Suzhou, 215000, China.
| |
Collapse
|
4
|
Zhang L, Yuan J, Yao S, Wen G, An J, Jin H, Tuo B. Role of m5C methylation in digestive system tumors (Review). Mol Med Rep 2025; 31:142. [PMID: 40183387 PMCID: PMC11979572 DOI: 10.3892/mmr.2025.13507] [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: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Currently, the incidence of digestive system tumors has been increasing annually, thus becoming a prevalent cause of cancer‑related mortalities. Although significant strides have been made in targeting the molecular mechanisms that underpin the development of these tumors, their treatment and prognosis still pose substantial challenges. This is primarily due to the ambiguity of early diagnostic indicators and the fact that most digestive system tumors are detected at an advanced stage. However, epigenetic modifications are capable of altering the expression of oncogenes and regulating biological processes in cancer. In recent years, the study of methylation in relation to tumor pathogenesis has become a focus of prominent research. Among the various types of methylation, 5‑methylcytosine (m5C) methylation plays a crucial role in the development of digestive system tumors and is anticipated to serve as a novel therapeutic target. However, to date, a comprehensive and systematic review concerning the role of m5C methylation in digestive system tumors is lacking. Consequently, the present study reviewed the role of m5C methylation in digestive system tumors such as esophageal cancer, gastric cancer and hepatocellular carcinoma, with the aim of providing a valuable reference for future research endeavors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianbo Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
5
|
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
|
6
|
Dai X, Huang H, Liu F. Rapid Identification of Esophageal Squamous Cell Carcinoma Biomarkers by MALDI-TOF MS Fingerprinting of Extracellular Vesicles. Anal Chem 2025. [PMID: 40326690 DOI: 10.1021/acs.analchem.4c06273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Esophageal cancer is a major global health challenge, with high incidence and mortality due to the lack of rapid and sensitive diagnostic tools and specific biomarkers. Cancer-cell-derived extracellular vesicles (EVs) carry unique proteins and nucleic acids, making them valuable sources of cancer biomarkers. We report an integrated method that combines an ultrafast exosome isolation system (EXODUS) with matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) to detect EVs and identify protein biomarkers for diagnosing and monitoring esophageal squamous cell carcinoma (ESCC). EVs derived from 20 mL culture medium supernatant of ESCC cells with varying degrees of differentiation serve as analysis models. We use EXODUS to isolate EVs rapidly. We then analyze the intact EVs using MALDI-TOF MS, which provides cell line-specific EV fingerprints in minutes. These protein fingerprints allow the discrimination of ESCC from normal control cells and enable the classification of ESCC based on the degree of cell differentiation. We explore critical EV biomarker peaks for ESCC diagnosis (5555 m/z, 8603 m/z, etc.) and monitoring (2268 m/z, etc.). Potential EV biomarker candidates, including YBX1, DIRAS2, HIST1H2AH, and MYBBP1A, are identified through tandem mass tag (TMT) proteomics. We tentatively assign the protein identities of EV marker peaks by correlation with the TMT proteomics. Applying this method to plasma-derived EVs shows promise for rapid, minimally invasive diagnosis and monitoring of ESCC.
Collapse
Affiliation(s)
- Xiaodan Dai
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Huiying Huang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Fei Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
7
|
Duan Q, Wang M, Cui Z, Ma J. Saikosaponin D suppresses esophageal squamous cell carcinoma via the PI3K-AKT signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6059-6070. [PMID: 39638887 DOI: 10.1007/s00210-024-03676-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
Saikosaponin D is the saikosaponin with the highest biological activity in Bupleurum chinense DC, which has anti-tumor effects on a variety of human tumors. In this study, we aimed to explore the SSD-induced apoptosis mechanism in ESCC cells. We predicted the targets of SSD and ESCC through several databases and analyzed the intersecting targets to identify the connections and possible pathways between proteins. We evaluated the binding activity between proteins and SSD through molecular docking. Based on the network pharmacology results, different concentrations of SSD were used to treat Eca-109 alongside Te-10 cells. The CCK-8, colony formation, wound healing, transwell, apoptosis, and western blot assays were performed to verify the inhibitory SSD impact on Eca-109 and Te-10 cells. Network pharmacology predicted 186 potential targets of SSD, and 500 targets of ESCC, along with 31 common targets, 5 core protein targets, and 94 potential pathways. Depending on molecular docking findings, SSD was closely bound to five core targets. Cellular experiments showed that SSD suppressed the Eca-109 and Te-10 cell proliferation and metastasis and enhanced apoptosis via the PI3K-AKT signaling. This study suggests SSD inhibited Eca-109 and Te-10 cell proliferation and migration by inhibiting the PI3K-AKT pathway and promoting apoptosis.
Collapse
Affiliation(s)
- Qiong Duan
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China
| | - Mingxiao Wang
- Sichuan Integrative Medicine Hospital, Chengdu, 610000, China
| | - Zhenting Cui
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China
| | - Jianxin Ma
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China.
| |
Collapse
|
8
|
Wang C, Zha YL, Wang H, Sun B, Qiang WG, Yuan Y, Shi HB, Hu WW. Carfilzomib promotes Iodine-125 seed radiation-induced apoptosis, paraptosis, and ferroptosis in esophageal squamous cell carcinoma by aggravating endoplasmic reticulum stress. Transl Oncol 2025; 57:102393. [PMID: 40315760 DOI: 10.1016/j.tranon.2025.102393] [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: 09/03/2024] [Revised: 02/09/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025] Open
Abstract
Iodine-125 (125I) seed brachytherapy has been applied to treat various malignant tumors such as esophageal cancer, however, radioresistance can reduce its efficacy. Endoplasmic reticulum stress (ERS) and subsequent unfolded protein response (UPR) is one of the core mechanisms of 125I seed radiation-induced cell death, thus aggravating ERS has been considered a promising sensitization strategy. Herein, we show that combination therapy of an irreversible proteasome inhibitor carfilzomib (CFZ) and 125I seed radiation displayed strong anti-tumor effect on esophageal squamous cell carcinoma (ESCC). Mechanistically, ERS and UPR regulated multiple cell death modalities induced by the combination therapy, including apoptosis, paraptosis, and ferroptosis. 125I seed radiation induced reactive oxygen species (ROS) production, DNA damage, p53 activation, and apoptosis. CFZ promoted ROS production, and augmented 125I seed radiation-induced apoptosis via the mitochondrial pathway, which was mediated by the UPR-C/EBP homologous protein (CHOP) pathway and was independent of the p53 pathway. CFZ enhanced 125I seed radiation-induced intracellular Ca2+ overload, protein ubiquitination, ERS, and UPR, consequently promoting paraptosis. 125I seed radiation induced accumulation of intracellular Fe2+ and lipid peroxides but upregulated the expression of ferroptosis inhibitors, SLC7A11 and glutathione peroxidase 4 (GPX4). The combination therapy promoted ferroptosis by enhancing the accumulation of intracellular Fe2+ and downregulating GPX4 expression. The mouse experiment demonstrated that CFZ can promote the efficacy of 125I seed radiation with good tolerance. Our findings suggest that combination therapy of 125I seed radiation and CFZ is associated with multiple cell death modalities and may serve as a promising therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Chao Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Yin-Lin Zha
- Department of Radiation Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Hao Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Bai Sun
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Wei-Guang Qiang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Ye Yuan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Hong-Bing Shi
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China.
| | - Wen-Wei Hu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China.
| |
Collapse
|
9
|
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
|
10
|
El Maimouni C, Córdova H, Feliz-Ruiz S, Luzko Scheid I, Moreira L, Llach J, Araujo IK, González-Suárez B, Ginés À, Fernández-Esparrach G. Association of esophageal squamous cell carcinoma with head and neck cancer. GASTROENTEROLOGIA Y HEPATOLOGIA 2025; 48:502318. [PMID: 39662764 DOI: 10.1016/j.gastrohep.2024.502318] [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: 08/09/2024] [Revised: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND AND OBJECTIVES Esophageal squamous cell carcinomas (ESCC) are often accompanied by head and neck squamous cell carcinoma (HNSCC) and vice versa. Our study aimed to describe the prevalence of HNSCC in patients with ESCC, the chronology of appearance and the impact on survival. METHODS A retrospective review was carried out through a computerized database of patients diagnosed with ESCC at Hospital Clinic of Barcelona between January 1999 and June 2019. Demographic data, date of ESCC diagnosis, survival time, primary tumor location, diagnosis of HNSCC and chronological relationship were recorded. RESULTS A total of 231 patients with ESCC confirmed histologically were included in the study with a median age of 64 years (IQR, 56.0-72.0), and 178 (77%) were male. The majority of the patients had a history of smoking and alcohol consumption (69.7% and 60.6%, respectively). The predominant location of ESCC was the middle esophagus (n=124, 53.7%). Forty-one patients (17.7%) had HNSCC: 21 (51.2%) were previous, 14 (34.1%) synchronous and 6 (14.6%) metachronous. All the patients were followed and 196 (84.8%) died with a median survival time of 19 months (IQR, 7-66). There were not statistically significant differences among the living patients and the deceased. CONCLUSIONS In our setting, a 17.7% of patients with ESCC have an associated HNSCC with no significant differences in survival between patients with both ESCC and HNSCC compared to those with only ESCC. However, the implementation of a screening program could allow the detection of a second primary tumor at early stages.
Collapse
Affiliation(s)
- Cautar El Maimouni
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Henry Córdova
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Sterling Feliz-Ruiz
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Irina Luzko Scheid
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Leticia Moreira
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Joan Llach
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Isis K Araujo
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Begoña González-Suárez
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Àngels Ginés
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain
| | - Gloria Fernández-Esparrach
- Endoscopy Unit, Gastroenterology Department, Hospital Clínic of Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Spain.
| |
Collapse
|
11
|
Li Y, Wang Y, Ren G, Yu H, Yin Y, Ma L, Yu X, Chen W, Zhang K, Zhao Y, Liu Z. Characterization of novel mouse esophageal squamous cell carcinoma cell lines and their utility as preclinical models. Cancer Lett 2025; 616:217600. [PMID: 40024567 DOI: 10.1016/j.canlet.2025.217600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) has a high incidence, poor treatment response, and high mortality. Subcutaneous transplant tumor models are commonly used to study the immunosuppressive tumor microenvironment and its impact on immunotherapy. In this study, we established two new ESCC mouse cell lines, mEC525M and mEC586F, from 4NQO-induced ESCC mouse models of different sexes. We compared their proliferation, motility, and molecular characteristics with existing lines (HNM007, AKR, mEC25) using in vitro experiments and whole-exome sequencing. Treatment sensitivity analysis of all murine ESCC tumor cell lines revealed that AKR and HNM007 cells were more responsive to chemotherapy, mEC25 cells were more sensitive to radiotherapy, whereas mEC525M and mEC586F cells exhibited greater sensitivity to immunotherapy. Multiplex immunohistochemistry (mIHC) staining analysis revealed differences in immune infiltration among the tumors derived from the five mouse ESCC cell lines, with the highest proportion of T cells in mEC525M tumors, the highest proportion of CD11b+ myeloid cells in mEC586F tumors and the highest proportion of CD19+ B cells in mEC25 tumors. In addition, RNA sequencing results also revealed differences in immune responses exhibited by tumor tissues derived from the five mouse ESCC cell lines after anti-PD1 treatment. Therefore, this study offers a valuable tool for investigating the immune microenvironment in ESCC and supports the selection of mouse models for preclinical ESCC research.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuhao Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guanzhu Ren
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hui Yu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yin Yin
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiao Yu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wangtianjiao Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kai Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yahui Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
12
|
Ding Y, Zhou L, He X, Li L, Wang X. Rhomboid Domain Containing 2 (RHBDD2) is Upregulated and Responds to Cisplatin-Induced DNA Damage in Esophageal Cancer. Biochem Genet 2025:10.1007/s10528-025-11118-y. [PMID: 40295376 DOI: 10.1007/s10528-025-11118-y] [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: 12/06/2024] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
As a pseudoprotease, RHBDD2 regulates tumor survival and progression in several cancers, but its expression and function in most cancers remain unclear. This study aimed to investigate the role of RHBDD2 in esophageal carcinoma (ESCA) via the use of public datasets and performing cytology experiments and RNA-seq analysis in ESCA cells. RHBDD2 was obviously upregulated and contributed to worse relapse-free survival (RFS) in patients with ESCA. Furthermore, chemotherapy increased RHBDD2 expression, and several GEO datasets also demonstrated that RHBDD2 expression was upregulated in both cisplatin-treated and resistant ESCA cells. Additionally, cytology experiments revealed that cisplatin treatment markedly upregulated the expression of RHBDD2 and γH2AX, which is a DNA damage marker, whereas RHBDD2 knockdown or overexpression affected cisplatin-induced γH2AX expression and the cytotoxic effects of cisplatin. The functional enrichment of RNA-seq data from RHBDD2-knockdown EC9706 cells suggested that RHBDD2-induced DEGs were involved in the DNA damage response and repair. Pearson's correlation analysis revealed that RHBDD2 expression was positively correlated with the expression of several genes, such as DMC1, CBX5, RAD1, DDB2, and ERCC1, which are responsible for DNA damage repair. Taken together, our results first revealed that RHBDD2 is upregulated and responds to DNA damage in ESCA, which provides new evidence for the potential role of RHBDD2 in the chemotherapy sensitivity and survival of patients with ESCA.
Collapse
Affiliation(s)
- Youxiang Ding
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Lin Zhou
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xintao He
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Xiaoping Wang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211100, China.
| |
Collapse
|
13
|
Nakai S, Makino T, Momose K, Yamashita K, Tanaka K, Miyata H, Yamamoto S, Motoori M, Kimura Y, Kawabata R, Hirao M, Matsuyama J, Akamaru Y, Morihara H, Ueyama A, Kurokawa Y, Morii E, Wada H, Eguchi H, Doki Y. Exploring predictive biomarkers of efficacy and survival with nivolumab treatment for unresectable/recurrent esophageal squamous cell carcinoma. Esophagus 2025:10.1007/s10388-025-01120-z. [PMID: 40274705 DOI: 10.1007/s10388-025-01120-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/11/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Programmed cell death protein-1 (PD-1) blockade has improved survival for patients with esophageal squamous cell carcinoma (ESCC), but response rates are low. Biomarkers to predict who will benefit from PD-1 blockade are urgently needed. METHODS This multicenter study involved 250 patients with recurrent/unresectable advanced ESCC receiving nivolumab as second- or later-line therapy. We assessed tumor-infiltrating T lymphocytes (TILs) and tertiary lymphoid structure (TLS) density using immunohistochemistry and hematoxylin/eosin staining in surgical specimens and pre-nivolumab endoscopic biopsies. RESULTS In surgical specimens, clinical response (vs. non-response) to nivolumab correlated significantly with CD8+ lymphocyte count (160 vs. 95.2 cells/field, P = 0.0494), CD8/Foxp3 ratio (6.52 vs. 2.72, P = 0.0053), and TLS density (0.21/mm2 vs. 0.10/mm2, P = 0.0005). In terms of overall survival, multivariate analysis identified CD8/Foxp3 ratio (hazard ratio [HR] = 1.83, P = 0.0050) and TLS density (HR = 1.67, P = 0.0171 as independent prognostic parameters in surgical specimens. Similarly, in endoscopic biopsies, clinical response (vs. non-response) to nivolumab correlated significantly with CD8+ counts (254 cells/mm2 vs. 124 cells/mm2, P = 0.0344), CCR8+ lymphocyte count (62.6 cells/mm2 vs. 140 cells/mm2, P = 0.0355), CD8/Foxp3 ratio (2.09 vs. 0.89, P = 0.040), and CD8/CCR8 ratio (2.34 vs. 0.89, P = 0.0020). Multivariate analysis also identified CD8/CCR8 ratio in endoscopic biopsies (HR = 1.66, P = 0.0313) as an independent prognostic parameter. CONCLUSIONS CD8+ and CCR8+ cell counts, CD8/Foxp3 and CD8/CCR8 ratios, and TLS density may be predictive biomarkers of therapeutic efficacy and survival with PD-1 blockade for ESCC.
Collapse
Affiliation(s)
- Shigeto Nakai
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Kota Momose
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Hiroshi Miyata
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Sachiko Yamamoto
- Department of Gastrointestinal Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Masaaki Motoori
- Department of Surgery, Osaka General Medical Center, Osaka, Japan
| | - Yutaka Kimura
- Department of Gastroenterological Surgery, Kindai University Nara Hospital, Nara, Japan
| | - Ryohei Kawabata
- Department of Surgery, Sakai City Medical Center, Osaka, Japan
| | - Motohiro Hirao
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Jin Matsuyama
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Osaka, Japan
| | - Yusuke Akamaru
- Department of Surgery, Osaka Rosai Hospital, Osaka, Japan
| | - Hitomi Morihara
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Azumi Ueyama
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hisashi Wada
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, 2-2-E2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
14
|
Pan Y, Sun Y, Xiao Y, Ding J, Hu G, Lin Z, Chen C. DOCK9 as a predictive biomarker linked to angiogenesis and immune response in esophageal squamous cell carcinoma. Clin Exp Med 2025; 25:126. [PMID: 40272582 PMCID: PMC12021961 DOI: 10.1007/s10238-025-01653-8] [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: 12/14/2024] [Accepted: 03/26/2025] [Indexed: 04/25/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) remains a serious health concern due to its high prevalence and mortality rates. Identifying prognostic biomarkers is essential to improving patient outcomes and treatment strategies. DOCK9, a gene implicated in various cellular functions, may play a significant role in ESCC progression and prognosis. We analyzed RNA microarray datasets and single-cell RNA sequencing data to identify survival-associated genes in ESCC. Using protein expression analysis, we examined DOCK9 in ESCC tissues and assessed its functional impact on human umbilical vein endothelial cells to understand its role in angiogenesis. Additionally, we developed a 21-gene prognostic risk model, focusing on the relevance of DOCK9. Our findings revealed that DOCK9 expression is significantly reduced in ESCC tissues and correlates with poor survival outcomes. Functionally, DOCK9 was found to regulate angiogenesis and modulate the tumor-associated fibroblast environment in ESCC. Furthermore, the DOCK9/CD31 ratio emerged as a potential marker to predict immune therapy response in ESCC. DOCK9 serves as a prognostic biomarker in ESCC, influencing both angiogenesis and immune response, and could guide future therapeutic strategies, particularly in immunotherapy. This study highlights DOCK9's relevance in ESCC prognosis, supporting its potential role in tailored therapies aimed at angiogenesis and immune modulation.
Collapse
Affiliation(s)
- Yaqiang Pan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People's Republic of China
| | - Yangyong Sun
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Ying Xiao
- Department of Emergency, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, 211100, Jiangsu, People's Republic of China
| | - Jifei Ding
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Ge Hu
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Zhiqiang Lin
- Department of Otolaryngology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Suzhou, 215008, Jiangsu, People's Republic of China.
| | - Chang Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People's Republic of China.
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
15
|
Zhang X, Du J, Lin X, Zhang S, Zeng T, Chen M, Huang G, Chen C, Zheng B. Identification of disulfidptosis in esophageal squamous cell carcinoma based on single-cell and bulk RNA-seq data to predict prognosis and treatment response. Front Immunol 2025; 16:1567793. [PMID: 40303412 PMCID: PMC12037556 DOI: 10.3389/fimmu.2025.1567793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Purpose Our study aims to identify the molecular subtypes of genes associated with disulfidptosis in esophageal squamous cell carcinoma(ESCC), develop a prognostic model, and identify potential therapeutic targets. Methods Based on the GSE53625 expression profile data, we identified molecular subtypes with significant survival differences through consensus cluster analysis. Subsequently, univariate Cox, multivariate Cox, and LASSO-Cox regression analysis were used to establish risk stratification models. The transcriptome data of the TCGA-ESCC cohort and the GSE160269 single-cell sequencing dataset were integrated to verify the biological significance of the model, and further analyze the heterogeneity of the tumor immune microenvironment, explore the differences in the intercellular communication network, and screen potential targeted drugs, providing a theoretical basis for subsequent translational research. Results We identified two distinct patterns of disulfidptosis expression with significant differences in overall survival. Then, we constructed the prognostic signature of disulfidptosis, and results showed patients with high score had worse prognosis. Univariate and multivariate Cox analysis demonstrated that the constructed prognostic signature was an independent prognostic factor and was validated in an independent validation set. The two subgroups differed in the proportion of immune cell infiltration and related signaling pathways in ESCC. The exploration of immunotherapy data confirmed our prognostic signature also had certain predictive power for immunotherapy. Drug screening suggested AZD8186 and JQ1 as potential therapies for high-score patients. Conclusion This study provides a new prognostic signature for ESCC, explores new therapeutic targets, and provides new theoretical support for personalized treatment.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Jianting Du
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Xiao Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Shuliang Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Taidui Zeng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Maohui Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Guanglei Huang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Bin Zheng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Key Laboratory of Cardio-Thoracic Surgery(Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| |
Collapse
|
16
|
Tan S, Wang R, Fang J, Yi M, Guo P, Han S, Li X, Gan Y, Liao J, Yu X, Li W. E3 ligase Skp2-mediated stabilization of survivin contributes to radioresistance. Cell Death Discov 2025; 11:151. [PMID: 40195315 PMCID: PMC11977269 DOI: 10.1038/s41420-025-02463-3] [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: 11/14/2024] [Revised: 03/15/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a frequently occurring neck and head malignancy. Therapies for OSCC are improving, but radiotherapy resistance remains a major clinical challenge. Here, we found that the S-phase kinase-associated protein 2 (Skp2) is overexpressed in OSCC cells and tissues. Knockdown of Skp2 significantly increased the radiotherapy sensitivity of OSCC cells. Further potential mechanisms suggest that Skp2-deficient restoration of radiotherapy sensitivity in OSCC cells may induce intrinsic apoptosis through inhibition of the Akt/Wee1/CDK1 axis, which inhibits Survivin phosphorylation and promotes its ubiquitination and degradation by FBXL7. Clinicopathologic histological analysis showed that Skp2 was positively correlated with the expression of p-Akt and Survivin in OSCC tissues. Furthermore, knockdown or inhibition of Skp2 overcame the radiotherapy resistance of OSCC cells. In conclusion, our study demonstrated that targeting the Skp2-Survivin axis could serve as an attractive and promising potential therapeutic target for radiotherapy sensitization in OSCC.
Collapse
Affiliation(s)
- Shiming Tan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
- Department of Haematology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ruirui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jinglin Fang
- School of Stomatology Hunan University of Chinese Medicine, Changsha, China
| | - Ming Yi
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Pengfei Guo
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xinfang Yu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, XiangYa Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
17
|
Rossi T, Valgiusti M, Puccetti M, Miserocchi G, Zanoni M, Angeli D, Arienti C, Pace I, Bassi C, Vannini I, Melloni M, Bandini E, Urbini M, Negrini M, Bonafè M, Ferracin M, Gallerani G. Gastroesophageal circulating tumor cell crosstalk with peripheral immune system guides CTC survival and proliferation. Cell Death Dis 2025; 16:223. [PMID: 40157906 PMCID: PMC11954855 DOI: 10.1038/s41419-025-07530-2] [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: 08/05/2024] [Revised: 02/12/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025]
Abstract
Tumor dissemination is a key event in tumor progression. During this event, a main role is played by circulating tumor cells (CTCs), immune cells, and their interaction. How the immune system supports the survival and proliferation of CTCs is not fully elucidated. In this study we established an in-vitro co-culture system consisting of immune cells and CTCs from the same patient, which increased the success rate in the establishment of CTC-derived long-term cell cultures. In this system, we characterized the immune cells of successful co-cultures and the signals they exchange with cancer cells, including cytokines and extracellular vesicle (EV) content. Using this protocol, we stabilized four CTC-derived cell lines from patients with metastatic gastroesophageal cancer, which were cultured for over a year and characterized from a genetic and molecular point of view. The four cell lines harbor shared chromosomal aberrations including the amplification at 8q24.21 containing MYC and deletion 9p21.3 containing CDKN2A/B and the IFN type I cluster. The transcriptomic profile of CTC cell lines is distinct from primary tumors, and we detected the activation of E2F, G2M and MYC pathways and the downregulation of interferon response pathway. Each cell line shows a degree of invasiveness in zebrafish in-vivo, and the most invasive ones share the same mutation in RAB14 gene. In addition, the four cell lines secrete cell-line specific EVs containing microRNAs that target YAP, BRG1-AKT1, TCF8-HDAC pathways. Overall, we highlight how the immune system plays a key role in the proliferation of CTCs through EV signaling, and how CTC cell line genomic and transcriptomic alterations make these cells less visible from the immune system and likely responsible for the survival advantage in sites distant from the microenvironment of origin.
Collapse
Affiliation(s)
- Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Martina Valgiusti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "DinoAmadori", Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Davide Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Arienti
- Immuno-Gene Therapy Factory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ilaria Pace
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Cristian Bassi
- Department of Translational Medicine, Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA) Centre, University of Ferrara, Ferrara, Italy
| | - Ivan Vannini
- Pathology Unit, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Mattia Melloni
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Erika Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Massimo Negrini
- Department of Translational Medicine, Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA) Centre, University of Ferrara, Ferrara, Italy
| | - Massimiliano Bonafè
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Manuela Ferracin
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giulia Gallerani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
| |
Collapse
|
18
|
Tkachev S, Brosalov V, Kit O, Maksimov A, Goncharova A, Sadyrin E, Dalina A, Popova E, Osipenko A, Voloshin M, Karnaukhov N, Timashev P. Unveiling Another Dimension: Advanced Visualization of Cancer Invasion and Metastasis via Micro-CT Imaging. Cancers (Basel) 2025; 17:1139. [PMID: 40227647 PMCID: PMC11988112 DOI: 10.3390/cancers17071139] [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: 11/04/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Invasion and metastasis are well-known hallmarks of cancer, with metastatic disease accounting for 60% to 90% of cancer-related deaths [...].
Collapse
Affiliation(s)
- Sergey Tkachev
- Institute for Regenerative Medicine, Sechenov University, 119992 Moscow, Russia
| | | | - Oleg Kit
- National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia
| | - Alexey Maksimov
- National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia
| | - Anna Goncharova
- National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia
| | - Evgeniy Sadyrin
- Laboratory of Mechanics of Biocompatible Materials, Don State Technical University, 344003 Rostov-on-Don, Russia
| | - Alexandra Dalina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Elena Popova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, 115682 Moscow, Russia
| | - Anton Osipenko
- Department of Pharmacology, Siberian State Medical University, 634050 Tomsk, Russia
| | - Mark Voloshin
- A.S. Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
| | - Nikolay Karnaukhov
- A.S. Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
- Institute of Clinical Morphology and Digital Pathology, Sechenov University, 119991 Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 119992 Moscow, Russia
| |
Collapse
|
19
|
Zhang J, Yu Q, Zhu W, Sun X. Recent advances in the role of circRNA in cisplatin resistance in tumors. Cancer Gene Ther 2025:10.1038/s41417-025-00899-4. [PMID: 40148680 DOI: 10.1038/s41417-025-00899-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/08/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major threat to human health, with chemotherapy serving as one of the main treatment strategies to alleviate patient suffering. However, prolonged chemotherapy often leads to the development of drug resistance, complicating treatment outcomes. Cisplatin, a commonly utilized chemotherapeutic agent, demonstrates efficacy against a range of cancers but frequently encounters resistance, posing a significant challenge in tumor management and prognosis. Drug resistance not only facilitates tumor progression but also reduces survival rates, highlighting the urgent need for innovative strategies to overcome this issue. In recent years, non-coding RNAs, particularly circular RNAs (circRNAs), have gained attention in cancer therapy due to their stability and specificity. Moreover, an increasing number of studies have reported that circRNAs are involved in cisplatin resistance across various types of cancer. This paper primarily reviews the mechanisms and roles of circRNA in mediating cisplatin resistance over the past 3 years. These findings highlight circRNAs as promising therapeutic targets for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Jiawen Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiwen Yu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Weijin Zhu
- Department of Clinical Laboratory Medicine, Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| | - Xiaochun Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
20
|
Zhang T, Tang X. Untangling immune cell contributions in the progression from GERD to Barrett's esophagus and esophageal cancer: Insights from genetic causal analysis. Int Immunopharmacol 2025; 150:114271. [PMID: 39965389 DOI: 10.1016/j.intimp.2025.114271] [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: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) is a rapidly increasing malignancy with significant morbidity and mortality. The progression from gastroesophageal reflux disease (GERD) to Barrett's esophagus (BE) and ultimately to EAC is thought to be influenced by chronic inflammation and immune cell dynamics. Despite the observed correlations in observational studies, the causal relationships between immune cell phenotypes and this disease continuum remain unclear. METHODS This study utilized a two-sample Mendelian Randomization (MR) approach to investigate the causal roles of 731 distinct immune cell phenotypes in the GERD-BE-EAC continuum. The analysis leveraged genome-wide association study (GWAS) data for immune phenotypes from a Sardinian cohort and data for GERD, BE, and EAC from the FinnGen and Open GWAS databases. A comprehensive set of MR methods, including inverse variance weighted (IVW), MR-Egger, and weighted median estimators, was employed to assess causality. Sensitivity analyses were conducted to evaluate heterogeneity and pleiotropy, ensuring the robustness of the findings. RESULTS The study revealed complex and multifaceted roles of immune cells across the GERD-BE-EAC continuum. In GERD, 34 immune phenotypes were found to be causally associated with either increased or decreased risk. Protective effects were observed in phenotypes such as Unswitched Memory B cells, while others like CD45RA- CD4+ T cells were linked to an elevated risk. In the context of BE, 28 immune phenotypes demonstrated significant causal associations, with the majority being protective, including Unswitched Memory B cells and CD62L on Granulocytes. Conversely, certain phenotypes, such as CD24 on Transitional B cells, were identified as risk factors for BE. For EAC, 34 immune phenotypes were implicated, with various B cell subsets, particularly those expressing BAFF-R and CD24, associated with an increased risk, while Switched Memory B cells and specific myeloid cell phenotypes showed protective effects. CONCLUSIONS This study provides novel insights into the complex role of immune cells in the pathogenesis of EAC, revealing a dynamic interplay where certain immune phenotypes may be protective in early stages but become risk-enhancing in later stages of disease progression. These findings highlight the potential of immune cell phenotypes to serve as biomarkers for early detection and targeted therapeutic interventions across the GERD-BE-EAC continuum. Further research is warranted to validate these findings in diverse populations and to explore the underlying mechanisms driving these immune-mediated effects.
Collapse
Affiliation(s)
- Tai Zhang
- Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University Health Science Center, Beijing 100091, China; Peking University Health Science Center, Beijing 100191, China
| | - Xudong Tang
- Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Peking University Health Science Center, Beijing 100091, China; Institute of Digestive Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
21
|
Yi Z, Li X, Li Y, Wang R, Zhang W, Wang H, Ji Y, Zhao J, Song J. Multi-cohort validation based on a novel prognostic signature of anoikis for predicting prognosis and immunotherapy response of esophageal squamous cell carcinoma. Front Oncol 2025; 15:1530035. [PMID: 40165896 PMCID: PMC11955476 DOI: 10.3389/fonc.2025.1530035] [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: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunotherapy is recognized as an effective and promising treatment modality that offers a new approach to cancer treatment. However, identifying responsive patients remains challenging. Anoikis, a distinct form of programmed cell death, plays a crucial role in cancer progression and metastasis. Thus, we aimed to investigate prognostic biomarkers based on anoikis and their role in guiding immunotherapy decisions for esophageal squamous cell carcinoma (ESCC). By consensus clustering, the GSE53624 cohort of ESCC patients was divided into two subgroups based on prognostic anoikis-related genes (ARGs), with significant differences in survival outcomes between the two subgroups. Subsequently, we constructed an ARGs signature with four genes, and its reliability and accuracy were validated both internally and externally. Additional, different risk groups showed notable variances in terms of immunotherapy response, tumor infiltration, functional enrichment, immune function, and tumor mutation burden. Notably, the effectiveness of the signature in predicting immunotherapy response was confirmed across multiple cohorts, including GSE53624, GSE53625, TCGA-ESCC, and IMvigor210, highlighting its potential utility in predicting immunotherapy response. In conclusion, the ARGs signature has the potential to serve as an innovative and dependable prognostic biomarker for ESCC, facilitating personalized treatment strategies in this field, and may represent a valuable new tool for guiding ESCC immunotherapy decision-making.
Collapse
Affiliation(s)
- Zhongquan Yi
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - Xia Li
- Department of General Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - Yangyang Li
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Rui Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Weisong Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Hao Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Yanan Ji
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - Jing Zhao
- Department of Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, China
| | - JianXiang Song
- Department of Cardiothoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| |
Collapse
|
22
|
Rebelo A, Friedrichs J, Grilli M, Vey J, Klose J, Merling M, Kleeff J, Ronellenfitsch U. Approaches for thoracoabdominal oesophagectomy for oesophageal cancer: a network meta-analysis - study protocol. BMJ Open 2025; 15:e093561. [PMID: 40082002 PMCID: PMC11907055 DOI: 10.1136/bmjopen-2024-093561] [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: 09/10/2024] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
INTRODUCTION Oesophageal cancer is the seventh most frequently diagnosed cancer and the sixth leading cause of cancer-related deaths worldwide. Oesophagectomy remains the main curative treatment option. The effect of different surgical approaches (completely open, hybrid, completely minimally invasive and robot-assisted) on patients undergoing thoracoabdominal oesophagectomy (Ivor-Lewis's procedure) for oesophageal cancer is evaluated, focusing on overall survival, postoperative mortality and morbidity. METHODS AND ANALYSIS A systematic literature search will be conducted in PubMed/Medline, Cochrane Library, Embase, Cumulated Index in Nursing and Allied Health Literature, ClinicalTrials.gov and International Clinical Trials Registry Platform using predefined search terms. A random-effects (network) meta-analysis using the frequentist framework will be performed. ETHICS AND DISSEMINATION As this study is based on previously published data, no ethical approval is required. Findings will be disseminated through peer-reviewed publications and conference presentations to inform clinical decision-makers (eg, surgeons, gastroenterologists). TRAIL REGISTRATION NUMBER CRD42024564915.
Collapse
Affiliation(s)
- Artur Rebelo
- Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University of Halle Wittenberg Faculty of Medicine, Halle (Saale), Germany
| | - Juliane Friedrichs
- Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University of Halle Wittenberg Faculty of Medicine, Halle (Saale), Germany
| | - Maurizio Grilli
- Library of the Medical Faculty, Mannheim, Heidelberg University, Mannheim, Germany, UK
| | - Johannes Vey
- University Hospital Heidelberg, Germany, Institute of Medical Biometry, Heidelberg, Germany, Heidelgerg, Germany
| | - Johannes Klose
- Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University of Halle Wittenberg Faculty of Medicine, Halle (Saale), Germany
| | - Marie Merling
- University Hospital Heidelberg, Germany, Institute of Medical Biometry, Heidelberg, Germany, Heidelgerg, Germany
| | - Joerg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University of Halle Wittenberg Faculty of Medicine, Halle (Saale), Germany
| | - Ulrich Ronellenfitsch
- Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University of Halle Wittenberg Faculty of Medicine, Halle (Saale), Germany
| |
Collapse
|
23
|
Nicholas B, Bailey A, McCann KJ, Walker RC, Johnson P, Elliott T, Underwood TJ, Skipp P. Comparative analysis of protein expression between oesophageal adenocarcinoma and normal adjacent tissue. PLoS One 2025; 20:e0318572. [PMID: 40073002 PMCID: PMC11902292 DOI: 10.1371/journal.pone.0318572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/19/2025] [Indexed: 03/14/2025] Open
Abstract
Oesophageal adenocarcinoma (OAC) is the 7th most common cancer in the United Kingdom (UK) and remains a significant health challenge. This study presents a proteomic analysis of seven OAC donors complementing our previous neoantigen identification study of their human leukocyte antigen (HLA) immunopeptidomes. Our small UK cohort were selected from donors undergoing treatment for OAC. We used label-free mass spectrometry proteomics to compare OAC tumour tissue to matched normal adjacent tissue (NAT) to quantify expression of 3552 proteins. We identified differential expression of a number of proteins previously linked to OAC and other cancers including common markers of tumourigenesis and immunohistological markers, as well as enrichment of processes and pathways relating to RNA processing and the immune system. Our findings also offer insight into the role of the protein stability in the generation of an OAC neoantigen we previously identified. These results provide independent corroboration of existing oesophageal adenocarcinoma biomarker studies that may inform future diagnostic and therapeutic research.
Collapse
Affiliation(s)
- Ben Nicholas
- Centre for Proteomic Research, Biological Sciences and Institute for Life Sciences, Building 85, University of Southampton, Southampton, United Kingdom,
- Centre for Cancer Immunology and Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Alistair Bailey
- Centre for Proteomic Research, Biological Sciences and Institute for Life Sciences, Building 85, University of Southampton, Southampton, United Kingdom,
- Centre for Cancer Immunology and Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Katy J. McCann
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Robert C. Walker
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Peter Johnson
- Cancer Research UK Clinical Centre, University of Southampton, Southampton, United Kingdom,
| | - Tim Elliott
- Centre for Cancer Immunology and Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
- Oxford Cancer Centre for Immuno-Oncology and CAMS-Oxford Institute, Nuffield Department of Medicine, University of Oxford, Southampton, United Kingdom
| | - Tim J. Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Paul Skipp
- Centre for Proteomic Research, Biological Sciences and Institute for Life Sciences, Building 85, University of Southampton, Southampton, United Kingdom,
| |
Collapse
|
24
|
Shen W, Liu C, Hu Y, Lei Y, Wong HS, Wu S, Zhou XM. CSsingle: A Unified Tool for Robust Decomposition of Bulk and Spatial Transcriptomic Data Across Diverse Single-Cell References. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.07.588458. [PMID: 38645128 PMCID: PMC11030304 DOI: 10.1101/2024.04.07.588458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
We introduce CSsingle, a novel method that enhances the decomposition of bulk and spatial transcriptomic (ST) data by addressing key challenges in cellular heterogeneity. CSsingle applies cell size correction using ERCC spike-in controls, enabling it to account for variations in RNA content between cell types and achieve accurate bulk data deconvolution. In addition, it enables fine-scale analysis for ST data, advancing our understanding of tissue architecture and cellular interactions, particularly in complex microenvironments. We provide a unified tool for integrating bulk and ST with scRNA-seq data, advancing the study of complex biological systems and disease processes. The benchmark results demonstrate that CSsingle outperforms existing methods in accuracy and robustness. Validation using more than 700 normal and diseased samples from gastroesophageal tissue reveals the predominant presence of mosaic columnar cells (MCCs), which exhibit a gastric and intestinal mosaic phenotype in Barrett's esophagus and esophageal adenocarcinoma (EAC), in contrast to their very low detectable levels in esophageal squamous cell carcinoma and normal gastroesophageal tissue. We revealed a dynamic relationship between MCCs and squamous cells during immune checkpoint inhibitors (ICI)-based treatment in EAC patients, suggesting MCC expression signatures as predictive and prognostic markers of immunochemotherapy outcomes. Our findings reveal the critical role of MCC in the treatment of EAC and its potential as a biomarker to predict outcomes of immunochemotherapy, providing insight into tumor epithelial plasticity to guide personalized immunotherapeutic strategies.
Collapse
Affiliation(s)
- Wenjun Shen
- Department of Bioinformatics, Shantou University Medical College, Shantou, China
- Chaoshan Branch of State Key Laboratory for Esophageal Cancer Prevention and Treatment, Shantou University Medical College, Shantou, China
| | - Cheng Liu
- Department of Computer Science, Shantou University, Shantou China
| | - Yunfei Hu
- Department of Computer Science, Vanderbilt University, Nashville, USA
| | - Yuanfan Lei
- Department of Bioinformatics, Shantou University Medical College, Shantou, China
| | - Hau-San Wong
- Department of Computer Sciences, City University of Hong Kong, Kowloon, Hong kong
| | - Si Wu
- Department of Computer Science, South China University of Technology, Guangzhou, China
| | - Xin Maizie Zhou
- Department of Computer Science, Vanderbilt University, Nashville, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, USA
| |
Collapse
|
25
|
Wang Z, Wu H, Li Z, Chen Z, Feng A, Chu Y, Fang K, Zhang Z, Zhao Z, Leng Z, Zhang S, Wang X, He L, Chen T, Xu M. PADI4 facilitates stem-like properties and cisplatin resistance through upregulating PRMT2/IDs family in oesophageal squamous cell carcinoma. Clin Transl Med 2025; 15:e70272. [PMID: 40078091 PMCID: PMC11904308 DOI: 10.1002/ctm2.70272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/15/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Oesophageal squamous cell carcinoma (OSCC) is a highly lethal cancer characterized by its aggressive nature and chemotherapy resistance. Peptidylarginine deiminase 4 (PADI4) regulates protein citrullination and is associated with various cancer developments. The role of PADI4 in OSCC progression and chemoresistance remains unexplored. METHODS The protein interactions were conducted by immunoprecipitation assays. Quantitative real-time PCR and western blotting were utilized to quantifyexpression levels in cancer cells. The stem-like properties were assessed through spheroid growth assays and Cancer Stem Cells (CSCs) markers. Additionally, the resistance of cancer cells to cisplatin was evaluated using CCK8 assay. RESULTS This study shows that PADI4 promotes cellular stemness, contributing to the progression and chemoresistance of OSCC. Mechanistically, PADI4 facilitates the citrullination of protein arginine methyltransferase 2 (PRMT2), a process essential for the stabilization of PRMT2 expression and the enhancement of its function in promoting the transcription of IDs family (ID1 and ID2) via histone arginine methylation. This mechanism subsequently increases tumour stemness and contributes to the cisplatin resistance observed in OSCC. Mutations at the R312 site or inhibition by GSK484 can attenuate tumour stemness in OSCC, thereby reducing cisplatin resistance. CONCLUSION PADI4 promotes citrullination and stabilization of PRMT2, enhancing its function in upregulating ID1 and ID2 expression via histone arginine methylation, which increases stemness and contributes to cisplatin resistance in OSCC; this effect can be mitigated by R312 mutations or GSK484 inhibition, reducing stemness and cisplatin resistance. KEY POINTS The role of citrullinization in cisplatin resistance of OSCC. PADI4 citrullinate of PRMT2 and stabilize PRMT2. PADI4 citrullinate of PRMT2 promoting the transcription of IDs family (ID1, ID2 and ID3) via histone arginine methylation. PADI4 citrullinated PRMT2 affected the combination of PRMT2 and USP7. PADI4 citrullinate of PRMT2 at R312 site. PADI4 inhibitor GSK484 can affect the stemness of OSCC and cisplatin resistance.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Wu
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhaoxing Li
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhukai Chen
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anqi Feng
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan Chu
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kang Fang
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zehua Zhang
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziying Zhao
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhuyun Leng
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shihan Zhang
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoyuan Wang
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lingnan He
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Chen
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Meidong Xu
- Department of Gastroenterology, Endoscopy Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
26
|
Cui Y, Hu Z, Zhang C. RNA Methyltransferase NSUN5 Promotes Esophageal Cancer via 5-Methylcytosine Modification of METTL1. Mol Carcinog 2025; 64:399-409. [PMID: 39601515 DOI: 10.1002/mc.23857] [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: 07/18/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Aberrant RNA modifications can drive carcinogenic transformation and tumor progression, with 5-methylcytosine (m5C) emerging as one of the predominant RNA modifications in eukaryotic cells. However, the function and molecular mechanisms of m5C in esophageal cancer (ESCA) remain insufficiently defined. Here we report that the m5C methyltransferase NOP2/Sun domain family member 5 (NSUN5) is significantly upregulated in ESCA tumors and shows promising diagnostic potential. Functionally, knockdown of NSUN5 impairs the proliferation capacity of ESCA cells and arrests cell cycle at the G0/G1 phase, while enforced expression of NSUN5 accelerates ESCA progression. In vivo, deficiency of NSUN5 significantly reduces tumor growth in a cell-based xenograft mouse model. Mechanistically, NSUN5 correlates with the oncogenic methyltransferase like 1 (METTL1), positively regulating its expression; NSUN5 binds directly to the METTL1 transcript, facilitating its m5C modification in ESCA cells. Additionally, overexpression of METTL1 effectively counteracts the tumor-suppressive effects resulting from NSUN5 ablation in both in vitro and in vivo settings. A comprehensive pan-cancer analysis further underscores NSUN5's essential role in digestive system tumors, with downregulation of NSUN5 notably inhibiting gastric and colon cancer cell growth. These findings provide new insights into epigenetic regulation in ESCA and propose the NSUN5/METTL1 axis as a promising therapeutic target for this malignancy.
Collapse
Affiliation(s)
- Yuanbo Cui
- Department of Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- Department of Trauma and Metabolism Institute of Zhengzhou University, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Zhaoyang Hu
- Department of Respiratory and Critical Care Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Chunyan Zhang
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
27
|
Wang H, Zou J, Zhao S, Zhang A. Optimization of three-dimensional esophageal tumor ablation by simultaneous functioning of multiple electrodes. Med Biol Eng Comput 2025; 63:793-806. [PMID: 39496870 DOI: 10.1007/s11517-024-03230-9] [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: 03/02/2024] [Accepted: 10/19/2024] [Indexed: 11/06/2024]
Abstract
Radiofrequency ablation is a widely accepted minimal-invasive and effective local treatment for tumors. However, its current application in esophageal cancer treatment is limited to targeting thin and superficial lesions, such as Barrett's Esophagus. This study proposes an optimization method using multiple electrodes simultaneously to regulate the temperature field and achieve conformal ablation of tumors. A particle swarm optimization algorithm, coupled with a three-dimensional thermal ablation model, was developed to optimize the status of the functioning electrodes, the optimal voltage (Vopt), and treatment duration (ttre) for targeted esophageal tumors. This approach takes into account both the electrical and thermal interactions of the electrodes. The results indicate that for esophageal cancers at various stages, with thickness (c) ranging from 4.5 mm to 10.0 mm, major axis (a) ranging from 7.3 mm to 27.3 mm, and minor axis (b) equaling 7.3 mm or 27.3 mm, as well as non-symmetrical geometries, complete tumor coverage (over 99.5%) close to conformal can be achieved. This method illustrates possible precise conformal ablation of esophageal cancers and it may also be used for conformal treatments of other intraluminal lesions.
Collapse
Affiliation(s)
- Hongying Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jincheng Zou
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shiqing Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Aili Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
28
|
Saadh MJ, Ehymayed HM, Alazzawi TS, Fahdil AA, Athab ZH, Yarmukhamedov B, Al-Anbari HHA, Shallal MM, Alsaikhan F, Farhood B. Role of circRNAs in regulating cell death in cancer: a comprehensive review. Cell Biochem Biophys 2025; 83:109-133. [PMID: 39243349 DOI: 10.1007/s12013-024-01492-6] [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] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
Despite multiple diagnostic and therapeutic advances, including surgery, radiation therapy, and chemotherapy, cancer preserved its spot as a global health concern. Prompt cancer diagnosis, treatment, and prognosis depend on the discovery of new biomarkers and therapeutic strategies. Circular RNAs (circRNAs) are considered as a stable, conserved, abundant, and varied group of RNA molecules that perform multiple roles such as gene regulation. There is evidence that circRNAs interact with RNA-binding proteins, especially capturing miRNAs. An extensive amount of research has presented the substantial contribution of circRNAs in various types of cancer. To fully understand the linkage between circRNAs and cancer growth as a consequence of various cell death processes, including autophagy, ferroptosis, and apoptosis, more research is necessary. The expression of circRNAs could be controlled to limit the occurrence and growth of cancer, providing a more encouraging method of cancer treatment. Consequently, it is critical to understand how circRNAs affect various forms of cancer cell death and evaluate whether circRNAs could be used as targets to induce tumor death and increase the efficacy of chemotherapy. The current study aims to review and comprehend the effects that circular RNAs exert on cell apoptosis, autophagy, and ferroptosis in cancer to investigate potential cancer treatment targets.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of dentist, National University of Science and Technology, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical technical college, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Bekhzod Yarmukhamedov
- Department of Surgical Dentistry and Dental Implantology, Tashkent State Dental Institute, Tashkent, Uzbekistan
- Department of Scientific affairs, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
29
|
Wu Y, Lin C, Qian Y, Huang X, Xu Y, Li J, He Y, Xie C, Su H. Identification of immune subtypes associated with CD8+ T cell-related genes providing new treatment strategies of esophageal carcinoma. Front Immunol 2025; 16:1512230. [PMID: 40083549 PMCID: PMC11903738 DOI: 10.3389/fimmu.2025.1512230] [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: 10/16/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Background CD8+ T lymphocytes greatly affect the efficacy of immunotherapy, displaying promising potential in various tumors. Here, we aimed to identify immune subtypes associated with CD8+ T cell-related genes to predict the efficacy of treatment in esophageal cancer (ESCA). Methods We obtained 13 immune cell-related datasets from the Gene Expression Omnibus (GEO) database and removed batch effects. Weighted correlation network analysis (WGCNA) and co-expression analysis were performed to identify highly correlated CD8+ T cell genes. Cox analysis was used to process ESCA clinical information, and the immune clusters (ICs) were constructed through consensus cluster analysis. Furthermore, we constructed an immune risk score model to predict the prognosis of ESCA based on these CD8+ T cell genes. This model was verified using the IMvigor210 dataset, and we functionally validated the immune risk score model in vitro. Results The results revealed significant correlations between CD8+ T cell-related genes and immune-related pathways. Three ICs were identified in ESCA, with IC3 demonstrating the most favorable prognosis. The final 6-gene prognostic risk model exhibited stable predictive performance in datasets across different platforms. Compared with that in normal esophageal epithelial (HEEC cells), CHMP7 in the 6-gene prognostic risk model was upregulated in KYSE150 and TE-1 cells. Si-CHMP7 transfection led to a decrease in tumor cell migration, invasion, and proliferation, accompanied by an accelerated apoptotic process. Conclusions Collectively, we identified the immune subtypes of CD8+ T cell-related genes with different prognostic significance. We designated CHMP7 in the 6-gene prognostic risk model as a potential target to improve tumor cell prognosis. These insights provide a strong basis for improving prognosis and facilitating more personalized and accurate treatment decisions for the immunotherapy of ESCA.
Collapse
Affiliation(s)
- Youyi Wu
- Department Oncology Radiotherapy, The Third Affiliated Hospital of Wenzhou Medical University, Rui’an People Hospital, Ruian, Zhejiang, China
| | - Chen Lin
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuchen Qian
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaowei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yajing Xu
- Department of Radiation Oncology Wenzhou Central Hospital Theorem Hospital Affiliated of Wenzhou Medical University, Wenzhou, China
| | - Jiayi Li
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youdi He
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huafang Su
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
30
|
Bassett JK, Peng Y, MacInnis RJ, Hodge AM, Lynch BM, Room R, Giles GG, Milne RL, Jayasekara H. Alcohol consumption trajectories over the life course and all-cause and disease-specific mortality: the Melbourne Collaborative Cohort Study. Int J Epidemiol 2025; 54:dyaf022. [PMID: 40064166 DOI: 10.1093/ije/dyaf022] [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] [Accepted: 02/21/2025] [Indexed: 04/27/2025] Open
Abstract
BACKGROUND Published studies rarely assess associations between trajectories of drinking and mortality. METHODS We aimed to assess associations between long-term sex-specific drinking trajectories and all-cause and disease-specific mortality for 39 588 participants (23 527 women; 16 061 men) enrolled in the Melbourne Collaborative Cohort Study in 1990-94 aged 40-69 years. Cox regression was used to estimate hazard ratios (HR) and 95% confidence intervals (CI) for all-cause, cardiovascular disease- and cancer-specific mortality in relation to group-based alcohol intake trajectories. RESULTS There were 7664 deaths (1117 cardiovascular; 2251 cancer) in women over 595 456 person-years, and 7132 deaths (1283 cardiovascular; 2340 cancer) in men over 377 314 person-years. We identified three distinct group-based alcohol intake trajectories for women: 'lifetime abstention', 'stable light', and 'increasing moderate'; and six for men: 'lifetime abstention', 'stable light', 'stable moderate', 'increasing heavy', 'early decreasing heavy', and 'late decreasing heavy'. We observed 9%-12% lower all-cause mortality, driven by associations with cardiovascular disease-specific deaths, for 'stable light' (women: HR 0.91; 95% CI 0.87-0.96; men: HR 0.88; 95% CI 0.82-0.94) and 'stable moderate' (HR 0.88; 95% CI 0.81-0.96) drinking, compared with 'lifetime abstention'. In contrast, all-cause mortality was 18%-21% higher for 'early decreasing heavy' (HR 1.18; 95% CI 1.05-1.32) and 'late decreasing heavy' (HR 1.21; 95% CI 1.04-1.40) drinking, and cancer-specific mortality 19%-37% higher for 'increasing moderate' (HR 1.19; 95% CI 1.00-1.43), 'early decreasing heavy' (HR 1.34; 95% CI 1.10-1.64), and 'late decreasing heavy' (HR 1.37; 95% CI 1.06-1.77) drinking. CONCLUSIONS Our findings highlight the importance of avoiding higher levels of alcohol intake during the life course to reduce all-cause and cancer-specific mortality.
Collapse
Affiliation(s)
- Julie K Bassett
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Yang Peng
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC, Australia
- School of Public Health, The University of Queensland, Herston, QLD, Australia
| | - Robert J MacInnis
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Allison M Hodge
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Brigid M Lynch
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Robin Room
- Centre for Alcohol Policy Research, School of Psychology & Public Health, La Trobe University, Bundoora, VIC, Australia
- Department of Public Health Sciences, Centre for Social Research on Alcohol and Drugs, Stockholm University, Stockholm, Sweden
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Harindra Jayasekara
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
- School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
31
|
Yao L, Zhou C, Liu L, He J, Wang Y, Wang A. Cancer-associated fibroblasts promote growth and dissemination of esophageal squamous cell carcinoma cells by secreting WNT family member 5A. Mol Cell Biochem 2025:10.1007/s11010-025-05223-0. [PMID: 39954174 DOI: 10.1007/s11010-025-05223-0] [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: 12/05/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common and aggressive subtype of esophageal cancer. This research investigates the functions of cancer-associated fibroblasts (CAFs) in the malignant phenotype of ESCC and probes the underpinning mechanism. Key CAF-associated proteins in ESCC were identified using bioinformatics analyses. ESCC cell lines were co-cultured with CAFs, followed by the addition of neutralizing antibodies against WNT family member 5A (WNT5A) (Anti-WNT5A; AW) and frizzled class receptor 5 (FZD5) (Anti-FZD5; AF), or a human recombinant protein of WNT5A (rWNT5A; rW). The effects of CAF stimulation and the neutralizing or recombinant proteins on the growth and dissemination of ESCC cells were investigated. In addition, ESCC cells were transplanted into nude mice for in vivo assessment of tumor growth and metastasis. WNT5A was identified as a CAF-associated protein linked to poor prognosis in ESCC. Co-culturing with CAFs augmented proliferation, mobility, and apoptosis resistance of ESCC cells. These effects were negated by the AW or AF but restored by rW. WNT5A interacted with FZD5 to activate the WNT signaling in ESCC cells. The rW treatment also enhanced tumorigenesis and metastasis of xenograft tumors in nude mice, with these effects diminished by AW or AF treatment. This study suggests that CAFs promote growth and dissemination of ESCC cell primarily through the secretion of WNT5A.
Collapse
Affiliation(s)
- Lishuai Yao
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510620, Guangdong, China
| | - Changshuai Zhou
- Department of Cardiothoracic Surgery, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
| | - Libao Liu
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510620, Guangdong, China
| | - Jinyuan He
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510620, Guangdong, China
| | - Youbo Wang
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - An Wang
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
32
|
Ürün YY, Beypinar İ. Inflammatory Biomarkers from Blood Counts as Prognostic Tools in Metastatic Esophageal Cancer. Med Sci Monit 2025; 31:e947202. [PMID: 39923126 PMCID: PMC11823267 DOI: 10.12659/msm.947202] [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: 01/03/2025] [Accepted: 12/19/2024] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Globally, esophageal cancer ranks as the sixth leading cause of cancer-related mortality. This retrospective study from a single center in Turkey aimed to evaluate hematological inflammatory biomarkers in complete blood count (CBC) data and outcomes in 113 patients with advanced esophageal carcinomas. MATERIAL AND METHODS We conducted a retrospective analysis of 113 patients with metastatic esophageal cancer composed of squamous (92), adenocarcinoma (18), and small cell (3) histology. We investigated neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), platelet-to-lymphocyte ratio (PLR), neutrophil-to-platelet lymphocyte ratio (NLPR), neutrophile-to-monocyte ratio (NMR), systemic inflammation index (SII), systemic inflammatory response index (SIRI), and aggregate index of systemic inflammation (AISI) in terms of prognosis. RESULTS The initial treatment for 25.7% of patients consisted of a carboplatin-paclitaxel combination. In response to the initial round of chemotherapy, 52.2% of patients showed improvement (15% complete, 37.2% partial), while 18.6% experienced disease progression. Neutropenia was observed as the most prevalent severe (grades 3-4) adverse reaction, affecting 19.8% of patients. Higher NLR, PLR, SII, NLPR, SIRI, and AISI values were associated with worse survival (P=0.016, P=0.008, P=0.011, P=0.028, P=0.014, P=0.001, respectively), whereas higher LMR was correlated with better survival (P=0.001). The NMR analysis showed no significant association (P=0.46). Multivariate analysis identified independent prognostic factors except histology, PLR, and NLPR. CONCLUSIONS Research indicates that inflammatory indicators obtained from complete blood count analyses possess prognostic significance for individuals with metastatic esophageal cancer. These biomarkers demonstrate diverse capacities in forecasting the course of the disease. These simple and inexpensive markers need further confirmation to guide individualized treatment planning.
Collapse
Affiliation(s)
- Yonca Yılmaz Ürün
- Department of Gastroenterology, Van Yüzüncüyıl University, Van, Türkiye
| | - İsmail Beypinar
- Department of Medical Oncology, Alanya Alaaddin Keykubat University, Alanya, Türkiye
| |
Collapse
|
33
|
Wang T, Wang X, Wang K, Yu M, Bai R, Zhang Y, Zhang Z, Liu F, Wang R, Shi X, Jia L, Liu K, Li X, Jin G, Zhao S, Dong Z. Chronic stress-induced cholesterol metabolism abnormalities promote ESCC tumorigenesis and predict neoadjuvant therapy response. Proc Natl Acad Sci U S A 2025; 122:e2415042122. [PMID: 39869796 PMCID: PMC11804521 DOI: 10.1073/pnas.2415042122] [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: 07/25/2024] [Accepted: 12/10/2024] [Indexed: 01/29/2025] Open
Abstract
Recent studies have demonstrated that chronic stress can enhance the development of multiple human diseases, including cancer. However, the role of chronic stress in esophageal carcinogenesis and its underlying molecular mechanisms remain unclear. This study uncovered that dysregulated cholesterol metabolism significantly promotes esophageal carcinogenesis under chronic stress conditions. Our findings indicate that the persistent elevation of glucocorticoids induced by chronic stress stimulates cholesterol uptake, contributing to esophageal carcinogenesis. The activated glucocorticoid receptor (GCR) enrichment at the promoter region of High Mobility Group Box 2 (HMGB2) facilitates its transcription. As a transcription coactivator, HMGB2 enhances Sterol Regulatory Element Binding Transcription Factor 1 (SREBF1) transcription and regulates cholesterol metabolism through LDL particle uptake into cells via Low Density Lipoprotein Receptor (LDLR). These results emphasize the significant impact of chronic stress on esophageal carcinogenesis and establish cholesterol metabolism disorder as a crucial link between chronic stress and the development of ESCC. The implications suggest that effectively managing chronic stress may serve as a viable strategy for preventing and treating ESCC.
Collapse
Affiliation(s)
- Ting Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Xiangyu Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Keke Wang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Mengyuan Yu
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan450000, China
| | - Ruihua Bai
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan450000, China
| | - Yiru Zhang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan450000, China
| | - Zihan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Feifei Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Rui Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Xiaodan Shi
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Ludan Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan450000, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan450000, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| | - Guoguo Jin
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
- Henan Key Laboratory of Chronic Disease Management, Fuwai Central China Car-Diovascular Hospital, Zhengzhou, Henan450000, China
| | - Simin Zhao
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan450000, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Bio-medical Sciences, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan450000, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Cancer Prevention and Treatment Lab, Zhengzhou University, Zhengzhou, Henan450000, China
| |
Collapse
|
34
|
Yang Y, Han C, Xing X, Qin Z, Wang Q, Lan L, Zhu H. Effects of Postoperative Complications on Overall Survival Following Esophagectomy: A Meta-Analysis Using the Restricted Mean Survival Time Analysis. Thorac Cancer 2025; 16:e70011. [PMID: 39924333 PMCID: PMC11807705 DOI: 10.1111/1759-7714.70011] [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: 12/19/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/11/2025] Open
Abstract
OBJECTIVE This study aims to conduct a comprehensive meta-analysis of the effects of postoperative complications (PCs) on survival following esophagectomy using the restricted mean survival time (RMST) analysis. METHODS A systematic literature search was performed in PubMed, Embase, Web of Science, Cochrane, and Medline, including articles published up to July 2024. Data were reconstructed from Kaplan-Meier curves, and the difference in RMST (RMSTD) and the RMST/restricted mean time loss (RMTL) ratios were calculated to examine the effects of PCs on overall survival. RESULTS A total of 12 articles, including 7925 patients, met the inclusion criteria. RMSTD estimates indicate that patients with overall PCs survived an average of 0.04 years shorter (RMSTD = -0.04, 95% CI: -0.06, -0.03) than those without PCs at the 1-year follow-up and 0.39 years shorter (RMSTD = -0.39, 95% CI: -0.55, -0.22) at the 5-year follow-up. Patients with anastomotic leaks survived an average of 0.34 years shorter (RMSTD = -0.34, 95% CI: -0.49, -0.19), and patients with pulmonary complications survived an average of 0.63 years shorter (RMSTD = -0.63, 95% CI: -0.81, -0.45) at the 5-year follow-up. Additionally, RMTL ratios were estimated to be 1.21 (95% CI: 1.12, 1.31) for overall PCs, 1.19 (95% CI: 1.11, 1.28) for anastomotic leaks, and 1.53 (95% CI: 1.36, 1.73) for pulmonary complications at the 5-year follow-up, respectively. CONCLUSIONS Our findings quantified the annual negative impact of PCs of esophageal cancer on overall patient survival following esophagectomy. Increased efforts are needed to enhance prevention, early screening, and timely treatment for complications, particularly for patients with pulmonary complications.
Collapse
Affiliation(s)
- Yongbo Yang
- First Department of Thoracic SurgeryPeking University Cancer Hospital and InstituteBeijingChina
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of EducationPeking University Cancer Hospital and InstituteBeijingChina
| | - Chunyang Han
- The First Clinical SchoolHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Xing Xing
- School of Public HealthPeking UniversityBeijingChina
| | - Zhen Qin
- School of Public HealthPeking UniversityBeijingChina
| | - Qianning Wang
- School of Public HealthPeking UniversityBeijingChina
| | - Lu Lan
- School of Public HealthPeking UniversityBeijingChina
| | - He Zhu
- School of Public HealthPeking UniversityBeijingChina
| |
Collapse
|
35
|
Fournier V, Fontesse S, Christophe V, Ramdane N, Anota A, Gauchet A, Lelorain S, Baudry AS, Duprez C, Devaux S, Bergeat D, D'Journo XB, Glehen O, Piessen G, Grynberg D. Postoperative complications and symptoms of anxiety and depression in patients with gastric and esophageal cancer: a retrospective cohort study. PSYCHOL HEALTH MED 2025; 30:282-296. [PMID: 39454564 DOI: 10.1080/13548506.2024.2417113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
CONTEXT Gastric and oesophageal cancers are common. They are also expected to increase in incidence in the next few years and are characterized by poor prognosis. Surprisingly, whereas the incidence of severe anxiety and depression is high in patients with gastric and oesophageal cancers, the influence of symptoms of depression and anxiety on postoperative complications has barely been explored. METHODS In a retrospective study based on a prospectively collected database, 629 cancer patients were enrolled. Symptoms of depression and anxiety (Hospital Anxiety and Depression Scale scores) and sociodemographic and medical information were collected immediately after diagnosis and before any treatment. The surgical approach (i.e. gastrectomy or oesophagectomy) and postoperative complications according to the Clavien-Dindo classification were collected after surgery. RESULTS After controlling for known medical predictors (i.e. surgical strategy, alcohol and tobacco consumption, American Society of Anaesthesiologists classification physical status score) of postoperative complications, no effect of symptoms of depression or anxiety was detected. DISCUSSION The observed results are surprising given the literature. However, several potential arguments can be put forwards regarding methods and measures, controlling variables, and conceptual distinctions. Despite the absence of significant results, this topic should be more deeply investigated by applying methodological and conceptual adjustments.
Collapse
Affiliation(s)
- Valentyn Fournier
- University of Lille, CNRS, UMR 9193 - SCALab - Sciences Cognitives et Sciences Affectives, Lille, France
| | - Sullivan Fontesse
- University of Lille, CNRS, UMR 9193 - SCALab - Sciences Cognitives et Sciences Affectives, Lille, France
| | - Véronique Christophe
- Human and Social Sciences Department, Centre Léon Bérard, Lyon, France
- Université Claude Bernard Lyon 1, Centre de Recherche en Cancérologie de Lyon - UMR Inserm 1052 - CNRS 5286 - UCBL - CLB, Lyon, France
| | | | - Amélie Anota
- Human and Social Sciences Department, Centre Léon Bérard, Lyon, France
- Direction of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Alice Gauchet
- Centre de recherche, Centre hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Sophie Lelorain
- Laboratoire de Psychologie de la santé, du sport et du vieillissement, Université de Lausanne, Institut de Psychologie, Lausanne, Suisse
| | - Anne-Sophie Baudry
- University of Lille, CNRS, UMR 9193 - SCALab - Sciences Cognitives et Sciences Affectives, Lille, France
- Pôle cancérologie et spécialités médicales, Centre Hospitalier de Valenciennes, Valenciennes, France
| | - Christelle Duprez
- University of Lille, CNRS, UMR 9193 - SCALab - Sciences Cognitives et Sciences Affectives, Lille, France
| | - Stephanie Devaux
- Department of Digestive and Oncological Surgery, CHU Lille, Lille, France
| | - Damien Bergeat
- Department of Digestive Surgery, CHU Rennes, Rennes, France
| | - Xavier Benoit D'Journo
- Department of Thoracic Surgery, North Hospital, Aix-Marseille University, Marseille, France
| | - Olivier Glehen
- Department of Surgical Oncology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, CHU Lille, Lille, France
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Delphine Grynberg
- University of Lille, CNRS, UMR 9193 - SCALab - Sciences Cognitives et Sciences Affectives, Lille, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
36
|
Lv H, Zhou J, Qiu L, Tang X, Huang C. AURKB and circAURKB_288aa enhance Esophageal cancer drug resistance through inducing abnormal centrosome separation. Biochem Pharmacol 2025; 232:116691. [PMID: 39638069 DOI: 10.1016/j.bcp.2024.116691] [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: 06/22/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Esophageal cancer (EC) is one of the most fatal malignancies worldwide, with a dramatic increase in incidence in the western world occurring over the past few decades. Chromosome instability (CIN) is a major contributor to EC progression, drug resistance, relapse, and the development of intratumoral heterogeneity. This study revealed a striking elevation of AURKB expression in EC patients, with a strong correlation to poor clinical outcomes. AURKB overexpression promoted cellular proliferation and induced drug resistance in both cell culture and animal models. Conversely, genetic targeting of AURKB abrogated these effects. Mechanistically, enforced AURKB expression triggered CIN, a key driver of poor EC outcomes, primarily through CEP250 phosphorylation. Interestingly, we identified a novel circular form of AURKB (circAURKB_288aa) harboring the AURKB kinase domain and encoding a 288-amino acid protein. Elevated levels of circAURKB_288aa in EC peripheral blood samples mirrored poor patient outcomes and synergistically enhanced CIN alongside AURKB. Furthermore, EC cells were capable of secreting circAURKB_288aa, influencing tumor microenvironmental cells similarly to full-length AURKB protein. Notably, AURKB siRNA targeting the shared kinase domain of both AURKB and circAURKB_288aa significantly inhibited EC malignancy. Collectively, these findings establish AURKB and circAURKB_288aa as promising targets for EC prognosis and therapy.
Collapse
Affiliation(s)
- Hongzhen Lv
- School of Basic Medical Sciences, Jiangsu Medical College, Yancheng, China
| | - Jing Zhou
- General Medicine Department, Yancheng Third People's Hospital, Yancheng, China
| | - Limin Qiu
- Thoracic Surgery Department, Yancheng NO.1 People's Hospital, Yancheng, China
| | - Xiaozhu Tang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Cheng Huang
- School of Traditional Chinese Medicine, Jiangsu Medical College, Yancheng, China.
| |
Collapse
|
37
|
Wei H, Zhao D, Zhi Y, Wu Q, Ma J, Xu J, Liu T, Zhang J, Wang P, Hu Y, He X, Guo F, Jiang M, Zhang D, Nie W, Yang R, Zhao T, Dong Z, Liu K. RTN4IP1 Contributes to ESCC via Regulation of Amino Acid Transporters. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406220. [PMID: 39757767 PMCID: PMC11848606 DOI: 10.1002/advs.202406220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/19/2024] [Indexed: 01/07/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) accounts for about 90% of esophageal cancer cases. The lack of effective therapeutic targets makes it difficult to improve the overall survival of patients with ESCC. Reticulon 4 Interacting Protein 1 (RTN4IP1) is a novel mitochondrial oxidoreductase. Here, a notable upregulation of RTN4IP1 is demonstrated, which is associated with poor survival in patients with ESCC. RTN4IP1 depletion impairs cell proliferation and induces apoptosis of ESCC cells. Furthermore, c-Myc regulates RTN4IP1 expression via iron regulatory protein 2 (IRP2) at the post-transcriptional level. Mechanistically, RTN4IP1 mRNA harbors functional iron-responsive elements (IREs) in the 3' UTR, which can be targeted by IRP2, resulting in increased mRNA stability. Finally, RTN4IP1 depletion abrogates amino acid uptake and induces amino acid starvation via downregulation of the amino acid transporters SLC1A5, SLC3A2, and SLC7A5, indicating a possible pathway through which RTN4IP1 contributes to ESCC carcinogenesis and progression. In vivo studies using cell-derived xenograft and patient-derived xenograft mouse models as well as a 4-nitroquinoline 1-oxide-induced ESCC model in esophageal-specific Rtn4ip1 knockout mice demonstrate the essential role of RTN4IP1 in ESCC development. Thus, RTN4IP1 emerges as a key cancer-promoting protein in ESCC, suggesting therapeutic RTN4IP1 suppression as a promising strategy for ESCC treatment.
Collapse
Affiliation(s)
- Huifang Wei
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Dengyun Zhao
- Department of PathophysiologySchool of Basic Medical Sciences, Zhengzhou UniversityChina‐US (Henan) Hormel Cancer InstituteChest Hospital of Zhengzhou UniversityZhengzhou450000China
| | - Yafei Zhi
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Qiong Wu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Jing Ma
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhou450000China
| | - Jialuo Xu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhou450000China
| | - Tingting Liu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Jing Zhang
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Penglei Wang
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Yamei Hu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Xinyu He
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Fangqin Guo
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Ming Jiang
- China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Dandan Zhang
- China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Wenna Nie
- China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Ran Yang
- China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Tongjin Zhao
- Department of PathophysiologySchool of Basic Medical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450000China
- State Key Laboratory of Genetic EngineeringShanghai Key Laboratory of Metabolic Remodeling and HealthInstitute of Metabolism and Integrative BiologyZhongshan HospitalShanghai Qi Zhi InstituteFudan UniversityShanghai200438China
| | - Zigang Dong
- Department of PathophysiologySchool of Basic Medical SciencesThe Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, State Key Laboratory of EsophagealCancer Prevention and TreatmentProvincial Cooperative Innovation Center for Cancer ChemopreventionChina‐US (Henan) Hormel Cancer Institute, Tianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450000China
| | - Kangdong Liu
- Department of PathophysiologySchool of Basic Medical SciencesThe Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, State Key Laboratory of EsophagealCancer Prevention and TreatmentProvincial Cooperative Innovation Center for Cancer ChemopreventionChina‐US (Henan) Hormel Cancer Institute, Tianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450000China
| |
Collapse
|
38
|
Yao L, Chen X, Fang Y, Huang Y, Wu K, Huang X, Xu J, Zhang R. Multi-Omics Analysis of Aberrances and Functional Implications of IRF5 in Digestive Tract Tumours. J Cell Mol Med 2025; 29:e70433. [PMID: 39993973 PMCID: PMC11850095 DOI: 10.1111/jcmm.70433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Oesophageal cancer (EC) is a common gastrointestinal malignancy and includes oesophageal squamous cell carcinoma (ESCC) and oesophageal adenocarcinoma (EAC) sub-types. Gene signatures predicting patient outcomes are not routinely used in clinical practice, particularly owing to batch effects and data standardisation. Here, we sought to establish and validate a reliable signature of senescence-related genes (SRGs) that would aid in predicting prognosis in patients with EC. We downloaded transcriptomics data, and a novel pairwise comparison algorithm selected valid SRG pairs (SRGPs) to construct a prognostic SRGP signature. The SRGPs were verified using Kaplan-Meier survival and receiver operating characteristic curve analyses. Additionally, the relationships between the SRGP signatures and prognosis, immune cell infiltration and chemotherapeutic drug responsiveness were evaluated. The random forest algorithm identified the most clinically significant genes, followed by experimental validation. 19 and 26 SRGP signatures were created for ESCC (n = 81) and EAC (n = 79), respectively. Patients with EC were divided into two groups based on the median risk score. The Kaplan-Meier analysis demonstrated significant differences in overall survival between the ESCC and EAC groups (p < 0.001). The sub-types exhibited different immune signatures. IRF5 was the most clinically significant gene for ESCC. It was highly expressed in ESCC cells, and IRF5 knockdown inhibited cell migration and proliferation, while promoting apoptosis and senescence. The SRGP signature may predict prognosis and immunotherapeutic responses, and IRF5 is a potential target gene for ESCC.
Collapse
Affiliation(s)
- Long Yao
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Xiu Chen
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Yanxin Fang
- Department of Cardiothoracic SurgeryAnhui No. 2 Provincial People's HospitalHefeiAnhuiChina
| | - Yunlong Huang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Kaiming Wu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Xin Huang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Junrui Xu
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Renquan Zhang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
39
|
Qu H, Yu Q, Ye L, Zheng J. SLC39A14 promotes the development of esophageal squamous cell carcinoma through PI3K/Akt/mTOR signaling pathway. Int Immunopharmacol 2025; 146:113831. [PMID: 39700956 DOI: 10.1016/j.intimp.2024.113831] [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: 10/12/2024] [Revised: 11/25/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVES This study aims to investigate the expression of solute carrier family 39 member 14 (SLC39A14) in esophageal squamous cell carcinoma (ESCC) tissues and its prognosis, as well as the impact of SLC39A14 expression on the biological behavior of ESCC cells and associated mechanisms. METHODS Bioinformatics analysis was utilized to compare the differential expression of SLC39A14 mRNA between esophageal cancer tissues and adjacent non-cancerous tissues. Immunohistochemistry was employed to evaluate SLC39A14 protein expression in human ESCC tissues and normal esophageal tissues, followed by an analysis of its association with clinicopathological parameters in esophageal cancer patients. Through cell proliferation, migration, invasion, and Western blot assays, we deeply evaluated the specific effects of SLC39A14 gene knockdown (or overexpression) on ESCC cells and explored its potential biological functions in ESCC. Subsequently, we validated the role of SLC39A14 in ESCC in a xenograft model. Furthermore, LY294002 drug intervention was used to verify the regulatory effect of SLC39A14 on PI3K/Akt/mTOR signaling pathway. RESULTS Both mRNA and protein levels of SLC39A14 were significantly elevated in tumor tissues from ESCC patients compared to adjacent normal tissues. Notably, higher levels of SLC39A14 expression positively correlated with ESCC tumor size (p = 0.010) and clinical T stage (p = 0.025), while exhibiting a negative correlation with overall patient survival rates (p = 0.023). In vitro experiments demonstrated that knocking down SLC39A14 significantly inhibited cell proliferation, migration and invasion. In vivo study showed that SLC39A14 facilitated progression within murine models bearing ESCC tumors. Mechanistic analyses suggested that pro-carcinogenic effects exerted by SLC39A14 are mediated through activation of the PI3K/Akt/mTOR signaling pathway. CONCLUSIONS Our findings suggest that SLC39A14 may serve as a potential biomarker for ESCC due to its pro-oncogenic role during ESCC progression.
Collapse
Affiliation(s)
- Hangshuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Qingxin Yu
- Department of pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province, China
| | - Luxia Ye
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Jingmin Zheng
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China.
| |
Collapse
|
40
|
Sun L, Zhao K, Liu X, Meng X. Global, regional, and national burden of esophageal cancer using the 2019 global burden of disease study. Sci Rep 2025; 15:3284. [PMID: 39865149 PMCID: PMC11770103 DOI: 10.1038/s41598-025-86244-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/09/2025] [Indexed: 01/28/2025] Open
Abstract
Esophageal cancer, with its aggressive nature and high mortality, poses diverse epidemiological challenges worldwide. Over the past three decades, esophageal cancer has exhibited a substantial global burden, marked by a significant increase in absolute numbers, contrasting with a decline in age-standardized metrics. Prevalence nearly doubled, reaching 0.961 million in 2019, while the age-standardized rate (ASR) decreased to 11.6 per 100,000 cases. New incidence cases surged by 67.07%, yet the age-standardized incidence rate reduced to 6.5 per 100,000 cases. Deaths increased to 0.498 million, with a decline in age-standardized mortality to 6.1 per 100,000 cases. Disability-Adjusted Life Years (DALYs) rose to 11.67 million, but the ASR decreased to 139.8 per 100,000 cases. Gender-specific analysis revealed consistently higher rates in males, with increasing gaps over time. Correlations with SDI indicated a negative association, and frontier analysis underscored the impact of socio-economic progress on disease control. Projections suggest a continued rise in prevalence, incidence, deaths, and DALYs, with gender-specific variations. The research underscores the importance of continued efforts in public health and medical research to adapt to and manage the changing landscape of esophageal cancer globally.
Collapse
Affiliation(s)
- Liangchao Sun
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Kaikai Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoli Liu
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China.
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
41
|
Lu Z, Geng M, Han Y, Cao J, Wang J, Liu T, Yuan X, Meng X, Zhang Y, Zhao R, Wan L, Li E, Wang W, Li Z, Shi D, Qian J, Shi S, Dong F, Shen L. Retrospective analysis of disease characteristics and treatment patterns among patients with esophageal cancer across 14 surgically represented centers. Cancer Biol Med 2025; 21:j.issn.2095-3941.2024.0336. [PMID: 39831767 PMCID: PMC11745092 DOI: 10.20892/j.issn.2095-3941.2024.0336] [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: 08/13/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Esophageal cancer (EC) ranks eighth among cancers in cancer-related deaths globally, and ~44% of new cases occur in China. We sought to describe the clinical characteristics and treatment landscape of EC in China before the approval of immunotherapy in 2020. METHODS CHANNEL was a large, retrospective study using patient-level data from 14 hospitals/cancer centers across China, including adults initiating therapy for newly diagnosed EC (January to December 2018). Demographics, clinicopathologic characteristics, and treatment patterns over 6 months were descriptively summarized. RESULTS Of 3,493 patients, 75.7% were men, the mean age was 64.1 years, and 75.0% had no family history of cancer. Most (92.8%) had squamous cell carcinoma, with a primary lesion in the middle esophagus (56.4%). Among patients with resectable EC, 92.9% received initial surgery, and 7.1% received neoadjuvant therapy, primarily chemotherapy (85.5% platinum-taxane). Among patients with unresectable early or locally advanced EC, 50.8% and 49.2% received palliative and radical therapy, respectively, as the initial treatment, primarily chemotherapy (66.5% platinum-taxane) and chemoradiotherapy (50.8% platinum-taxane), respectively. Adjuvant therapy was administered to 22.9% of patients undergoing initial surgery, and 2.4% receiving neoadjuvant therapy and surgery. Among patients with advanced EC, 84.6% received systemic therapy as an initial treatment, primarily chemotherapy (61.5% platinum-taxane). CONCLUSIONS Before the approval of immunotherapy in China, most patients with resectable early or locally advanced EC underwent radical surgery without preoperative treatment, whereas most patients with advanced EC received platinum-taxane chemotherapy. These findings highlight the need for novel EC treatments before immunotherapy was introduced, and provide a baseline for evaluating the benefits of immunotherapy, now that this treatment is widely used in this setting.
Collapse
Affiliation(s)
- Zhihao Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mingfei Geng
- Department of Thoracic Surgery, Anyang Cancer Hospital, Anyang 455001, China
| | - Yongtao Han
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Chengdu 610042, China
| | - Jianzhong Cao
- Department of Radiotherapy, Shanxi Provincial Cancer Hospital, Taiyuan 030013, China
| | - Jun Wang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xianglin Yuan
- Department of Medical Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Meng
- Department of Radiotherapy, Shandong Cancer Hospital, Jinan 250117, China
| | - Yanqiao Zhang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Rong Zhao
- Department of Pharmacy, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Lixin Wan
- Department of Medical Oncology, Nanyang Central Hospital, Nanyang 473005, China
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Wenran Wang
- Department of the Cancer Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Zhijie Li
- Department of Thoracic Surgery, Weifang Second People’s Hospital, Weifang 261041, China
| | - Danfeng Shi
- Value & Implementation, Global Medical & Scientific Affairs, MSD China, Shanghai 200233, China
| | - Jing Qian
- Value & Implementation, Global Medical & Scientific Affairs, MSD China, Shanghai 200233, China
| | - Si Shi
- Value & Implementation, Global Medical & Scientific Affairs, MSD China, Shanghai 200233, China
| | - Fengshi Dong
- Value & Implementation, Global Medical & Scientific Affairs, MSD China, Shanghai 200233, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
42
|
Yu SS, Zheng X, Li XS, Xu QJ, Zhang W, Liao ZL, Lei HK. Development of a nomogram for overall survival in patients with esophageal carcinoma: A prospective cohort study in China. World J Gastrointest Oncol 2025; 17:96686. [PMID: 39817137 PMCID: PMC11664612 DOI: 10.4251/wjgo.v17.i1.96686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Esophageal carcinoma (EC) presents a significant public health issue in China, with its prognosis impacted by myriad factors. The creation of a reliable prognostic model for the overall survival (OS) of EC patients promises to greatly advance the customization of treatment approaches. AIM To create a more systematic and practical model that incorporates clinically significant indicators to support decision-making in clinical settings. METHODS This study utilized data from a prospective longitudinal cohort of 3127 EC patients treated at Chongqing University Cancer Hospital between January 1, 2018, and December 12, 2020. Utilizing the least absolute shrinkage and selection operator regression alongside multivariate Cox regression analyses helped pinpoint pertinent variables for constructing the model. Its efficacy was assessed by concordance index (C-index), area under the receiver operating characteristic curve (AUC), calibration curves, and decision curve analysis (DCA). RESULTS Nine variables were determined to be significant predictors of OS in EC patients: Body mass index (BMI), Karnofsky performance status, TNM stage, surgery, radiotherapy, chemotherapy, immunotherapy, platelet-to-lymphocyte ratio, and albumin-to-globulin ratio (ALB/GLB). The model demonstrated a C-index of 0.715 (95%CI: 0.701-0.729) in the training cohort and 0.711 (95%CI: 0.689-0.732) in the validation cohort. In the training cohort, AUCs for 1-year, 3-year, and 5-year OS predictions were 0.773, 0.787, and 0.750, respectively; in the validation cohort, they were 0.772, 0.768, and 0.723, respectively, illustrating the model's precision. Calibration curves and DCA verified the model's predictive accuracy and net benefit. CONCLUSION A novel prognostic model for determining the OS of EC patients was successfully developed and validated to help clinicians in devising individualized treatment schemes for EC patients.
Collapse
Affiliation(s)
- Shi-Shi Yu
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xi Zheng
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xiao-Sheng Li
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Qian-Jie Xu
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Wei Zhang
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Zhong-Li Liao
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Hai-Ke Lei
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
43
|
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
|
44
|
Zhou W, Yi Y, Cao W, Zhong X, Chen L. Functions of METTL1/WDR4 and QKI as m7G modification - related enzymes in digestive diseases. Front Pharmacol 2025; 15:1491763. [PMID: 39850560 PMCID: PMC11754259 DOI: 10.3389/fphar.2024.1491763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
N7-methylguanosine (m7G) modification is one of the most prevalent forms of chemical modification in RNA molecules, which plays an important role in biological processes such as RNA stability, translation regulation and ribosome recognition. Methyl-transferation of m7G modification is catalyzed by the enzyme complex of methyltransferase-like 1 (METTL1) and WD repeat domain 4 (WDR4), and Quaking (QKI) recognizes internal m7G methylated mRNA and regulates mRNA translation and stabilization. Recent studies have found that m7G modification - related enzymes are associated with the onset and progression of digestive cancer, such as colorectal cancer, liver cancer, and other digestive diseases such as ulcerative colitis. This review will focus on the latest research progress on the roles of m7G methyltransferase METTL1/WDR4 and recognized enzyme QKI in digestive diseases.
Collapse
Affiliation(s)
- Wenyan Zhou
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yan Yi
- Institute Center of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Wenyu Cao
- Clinical Anatomy and Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiaolin Zhong
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Ling Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
45
|
Cai Y, Ding J, Cai X, Su W, Weng G, Zheng X, Chen S, Chen L, Lin Y, Yao Q, Yang C. Constructing individualized follow-up strategies for locally advanced esophageal squamous cell carcinoma patients based on dynamic recurrence risk changes. Sci Rep 2025; 15:175. [PMID: 39747490 PMCID: PMC11695730 DOI: 10.1038/s41598-024-84099-4] [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: 07/16/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
The aim of this study was to explore the high-risk factors for recurrence in patients with locally advanced esophageal squamous cell carcinoma (ESCC) undergoing definitive chemoradiotherapy or radiotherapy (dCRT or dRT). Conditional survival (CS) was used to evaluate the dynamic survival and recurrence risk of patients after treatment, and individualized monitoring strategies were developed for patients. Logistic regression analysis was performed to determine independent recurrence risk factors. Calibration curves and receiver operating characteristic (ROC) curve were used to evaluate nomogram models. Kaplan-Meier curves were used to compare survival rates in different groups and to calculate CS rate. A total of 677 patients were included. Multivariate logistic analyses demonstrated that chemotherapy cycles, tumor length, body mass index (BMI), platelet-lymphocyte ratio (PLR), and lymphocyte-monocyte ratio (LMR) were independent recurrence risk factors (p < 0.05). Subsequently, we constructed nomogram models to predict recurrence and risk stratification. Kaplan-Meier curves showed that conditional locoregional recurrence-free survival and distant metastasis-free survival of patients in different risk groups and clinical stages progressively increased with survival time, whereas local recurrence and distant metastasis annual recurrence rates decreased yearly with increasing survival time. Finally, we developed an individualized follow-up strategy based on CS at different frequencies. Individualized follow-up strategies developed on the basis of CS can better monitor the changes in patients' conditions and contribute to timely salvage treatment and rational allocation of healthcare resources.
Collapse
Affiliation(s)
- Yibin Cai
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| | - Jianming Ding
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - XiaoJun Cai
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Weikun Su
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Guibin Weng
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xinlong Zheng
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Shijie Chen
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lin Chen
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - YiJin Lin
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Qiwei Yao
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| | - Chunkang Yang
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| |
Collapse
|
46
|
Hirase Y, Sasaki K, Tsuruda Y, Shimonosono M, Uchikado Y, Matsushita D, Arigami T, Tada N, Baba K, Kawasaki Y, Ohtsuka T. Prognostic impact of preoperative osteosarcopenia on esophageal cancer surgery outcomes: a retrospective analysis. Esophagus 2025; 22:77-84. [PMID: 39604759 DOI: 10.1007/s10388-024-01101-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Osteosarcopenia, recognized as a consequence of aging, has garnered attention as a prognostic marker in recent years; however, its clinical significance in esophageal cancer remains uncertain. This study aimed to investigate the impact of osteosarcopenia on esophageal cancer surgery outcomes. METHODS This retrospective study included patients with advanced esophageal cancer who underwent surgical resection between 2018 and 2021. Skeletal muscle index at the L3 vertebral level and bone density at the Th11 vertebral level were measured on preoperative computed tomography scans. Based on the findings, we divided patients into sarcopenia, osteopenia, and osteosarcopenia groups, and analyzed the relationship between osteosarcopenia and clinicopathological factors, including prognosis. RESULTS Of the 124 patients included, 59 (48%) were diagnosed with osteosarcopenia. Among all, 46 (37%) patients experienced postoperative recurrence, and a significant correlation was observed between osteosarcopenia and recurrence (p < 0.05). Overall survival and relapse-free survival were significantly shorter in the osteosarcopenia group than in the non-osteosarcopenia group (p < 0.05 for both). In a subgroup analysis, overall survival and relapse-free survival were significantly shorter in the osteosarcopenia group than in the non-osteosarcopenia group, or in the sarcopenia and osteopenia alone groups (all p < 0.05). CONCLUSIONS The presence of preoperative osteosarcopenia was found to affect the prognosis following esophageal cancer surgery.
Collapse
Affiliation(s)
- Yuki Hirase
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Ken Sasaki
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan.
| | - Yusuke Tsuruda
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Masataka Shimonosono
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Yasuto Uchikado
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Daisuke Matsushita
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Takaaki Arigami
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Nobuhiro Tada
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Kenji Baba
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Yota Kawasaki
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| | - Takao Ohtsuka
- Department of Digestive Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima City, Kagoshima, 890-8520, Japan
| |
Collapse
|
47
|
Hu Y, Zhang Y, Wang S, Wang R, Yuan Q, Zhu L, Xia F, Xue M, Wang Y, Li Y, Yuan C. LINC00667: A Novel Vital Oncogenic LincRNA. Curr Med Chem 2025; 32:678-687. [PMID: 37855347 DOI: 10.2174/0109298673248494231010044348] [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: 02/24/2023] [Revised: 06/14/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023]
Abstract
Long intergenic noncoding RNAs (lincRNAs) have a variety of properties that differ from those of messenger RNAs (mRNAs) encoding proteins. Long intergenic nonprotein coding RNA 667 (LINC00667) is a non-coding transcript located on chromosome 18p11.31. Recently, many studies have found that LINC00667 can enhance the progression of various cancers and play a key part in a lot of diseases, such as tumorigenesis. Therefore, LINC00667 can be recognized as a potential biomarker and therapeutic target. So, we reviewed the biological functions, relevant mechanisms, as well as clinical significance of LINC00667 in several human cancers in detail.
Collapse
Affiliation(s)
- Yaqi Hu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yifan Zhang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Shuwen Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Rui Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Qi Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Leiqi Zhu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Fangqi Xia
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Mengzhen Xue
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yaqi Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yuanyang Li
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Chengfu Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| |
Collapse
|
48
|
Zhang J, Song S, Li Y, Gong A. Predicting Outcomes in Esophageal Squamous Cell Carcinoma Using scRNA-Seq and Bulk RNA-Seq: A Model Development and Validation Study. Cancer Med 2025; 14:e70617. [PMID: 39840762 PMCID: PMC11751878 DOI: 10.1002/cam4.70617] [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: 06/18/2024] [Revised: 11/25/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Altered glucose metabolism is a critical characteristic from the beginning stage of esophageal squamous cell carcinoma (ESCC), and the phenomenon is presented as a pink-color sign under endoscopy after iodine staining. Therefore, calculating the metabolic score based on the glucose metabolic gene sets may bring some novel insights, enabling the prediction of prognosis and the identification of treatment choices for ESCC. METHODS A total of 8, 99, and 140 individuals from The Gene Expression Omnibus database, The Cancer Genome Atlas database, and the Memorial Sloan Kettering Cancer Center, respectively, were encompassed in the investigation. Patients diagnosed with ESCC after surgery were enrolled for further validation. RESULTS A total of 13 kinds of cell clusters were screened, and the squamous epithelium was identified with the highest score. And 558 differential genes were selected from the single-cell RNA sequencing (scRNA-seq) dataset. Four glucose metabolism-related genes, namely, SERP1, CTSC, RAP2B, and SSR4, were identified as hub genes to develop a risk prognostic model. The model was validated in another external cohort. According to the risk score (RS) determined by the model, the patients were categorized into low- and high-risk groups (LRG and HRG). Compared with LRG, HRG indicated poor survival and decreased drug sensitivity. Additionally, the immune microenvironment and pathway enrichment were different between the two groups. Immunohistochemical staining revealed that hub genes were expressed differently in ESCC tissues, high- and low-grade intraepithelial neoplasia, and adjacent normal tissues. CONCLUSION Four hub genes (SERP1, CTSC, RAP2B, and SSR4) screened based on glucose metabolism developed a predictive model in ESCC patients. The RS was established as an independent risk factor for predicting prognosis. These findings may enhance understanding of ESCC's molecular profile and serve as a new prognostic tool for better patient stratification and treatment planning in clinical practice.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Digestive EndoscopyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
| | - Shunzhe Song
- Department of Digestive EndoscopyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
| | - Yuqing Li
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
| | - Aixia Gong
- Department of Digestive EndoscopyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
- Department of GastroenterologyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
| |
Collapse
|
49
|
Hu J, Meng F, Lv L, Hong F, He Q, Zhu Q, Tian T, Chang N, Zhang S, Yi Q, Qian L. GPR37-enhanced ubiquitination of ATP1A1 inhibits tumor progression and radiation resistance in esophageal squamous cell carcinoma. Cell Death Dis 2024; 15:933. [PMID: 39730361 PMCID: PMC11681203 DOI: 10.1038/s41419-024-07240-1] [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: 07/18/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 12/29/2024]
Abstract
Radiotherapy resistance is one of the main reasons for the dismal clinical outcome of patients with esophageal squamous cell carcinoma (ESCC). Therefore, clarifying the targets and molecular mechanisms of radiotherapy resistance in ESCC is of great theoretical and clinical significance to enhance the efficacy of radiotherapy. In this study, GPR37 was identified as a key factor facilitating ESCC radiosensitization. We found that GPR37 is lowly expressed in ESCC, especially in radioresistant ESCC tumors. And its insufficiency is related to the malignant characteristics and unfavorable prognosis in ESCC. Further investigation revealed that GPR37 level is inversely regulated by promoter methylation but positively regulated by ZNF750. Functionally, GPR37 could not only overcome radioresistance of ESCC, but also inhibit proliferation, migration, and invasion. Mechanistically, GPR37 interacts with the ATP1A1 protein, effectively promoting its ubiquitination-induced degradation, thereby limiting the activation of the AKT/mTOR signaling pathway. Additionally, GPR37 can be transported to recipient cells via exosomes and inhibit the malignant behavior of recipient cells. Overall, these findings suggest that GPR37-ATP1A1 axis holds potential as a therapeutic target for the management of ESCC, especially for overcoming radiation resistance.
Collapse
Affiliation(s)
- Jiaru Hu
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Fang Meng
- Department of Oncology & Hematology, Xishan People's Hospital of Wuxi City, Wuxi, 214105, China
| | - Lei Lv
- Department of Cancer Epigenetics Program, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, 230031, China
| | - Fu Hong
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, 230031, China
| | - Qing He
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Qi Zhu
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Tian Tian
- Department of Respiratory Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, 230001, China
| | - Na Chang
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, 230031, China
| | - Shiqiang Zhang
- Department of Oncology & Hematology, Xishan People's Hospital of Wuxi City, Wuxi, 214105, China
| | - Qiyi Yi
- Institute of Radiation Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Liting Qian
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
50
|
Ohsawa M, Nishi H, Emi M, Yoshikawa T, Hamai Y, Ibuki Y, Kurokawa T, Hirohata R, Kitasaki N, Kawada-Matsuo M, Komatsuzawa H, Kawaguchi H, Okada M. Impact of Fusobacterium nucleatum in the treatment of cancer, including radiotherapy and its future potential in esophageal cancer. JOURNAL OF RADIATION RESEARCH 2024; 65:i126-i134. [PMID: 39679879 DOI: 10.1093/jrr/rrae061] [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: 12/02/2023] [Revised: 05/09/2024] [Indexed: 12/17/2024]
Abstract
Despite advances in multimodality therapy, including surgery, chemotherapy, radiation therapy and chemoradiation, the fatality rate for esophageal cancer remains high. Specifically, Fusobacterium nucleatum, due to its aggregation capacity, has shown a tendency to form biofilms. The biofilm-forming capabilities of microbial communities are of utmost importance in the context of cancer treatment, as they have been shown to drive significant losses in the efficaciousness of various cancer treatments. Therefore, elucidating the dynamics of F. nucleatum will be important for the development of effective treatments for esophageal cancer. Therefore, this review summarizes the current knowledge of F. nucleatum, its involvement in cancer and its impact on chemotherapy and radiation therapy. In conclusion, further research on the role of F. nucleatum is essential for the continued advancement of the treatment of esophageal cancer and patient care.
Collapse
Affiliation(s)
- Manato Ohsawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hiromi Nishi
- Department of General Dentistry, Hiroshima University Hospital, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Manabu Emi
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Toru Yoshikawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Yoichi Hamai
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Yuta Ibuki
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Tomoaki Kurokawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Ryosuke Hirohata
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Nao Kitasaki
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hiroyuki Kawaguchi
- Department of General Dentistry, Hiroshima University Hospital, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| |
Collapse
|