1
|
Duan H, Li Y, Zheng X, Hou J, Tao H, Liu X, Dai M, He S. Paeonol enhances a recombinant EGFR-targeted fusion protein-drug conjugate induced antitumor efficacy in esophageal cancer. Biochem Pharmacol 2025; 236:116856. [PMID: 40054783 DOI: 10.1016/j.bcp.2025.116856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Esophageal cancer (EC) is a gastrointestinal cancer with high morbidity and mortality, along with a low 5-year survival rate, which urgently requires the discovery of new drugs for prevention and treatment. Our previous studies have found a novel EGFR-targeted fusion protein-drug conjugate, Fv-LDP-D3-AE, which exhibits significant inhibitory activity against esophageal cancer. However, the effectiveness of monotherapy still faces major challenges in clinical translation for esophageal cancer treatment. Therefore, there is an urgent need to identify a candidate anti-tumor drug that can be combined with Fv-LDP-D3-AE to enhance therapeutic efficacy. In this study, we report a novel combination treatment regimen of paeonol with Fv-LDP-D3-AE, using human esophageal cancer cells KYSE70 and EC109 for in vitro studies and establishing a BALB/c nude mouse xenograft model for in vivo experiments to investigate the anti-tumor efficacy and potential mechanisms of the combination therapy in esophageal cancer. The results indicated that the combined treatment emerged a synergistic effect, which could effectively inhibit the proliferation, migration, and invasion of esophageal cancer cells, induce more obvious cell apoptosis and DNA damage, and suppress tumor growth in the xenograft mouse model with a tumor inhibition rate of 76%. This may be attributed to the combination therapy simultaneously inhibiting the EGFR/AKT/mTOR signaling pathway and downregulating the expression of nucleolin. Overall, these findings suggest that paeonol could synergize with Fv-LDP-D3-AE to enhance anti-esophageal cancer efficacy, which may be a promising therapeutic strategy for esophageal cancer.
Collapse
Affiliation(s)
- Huaiyu Duan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yuting Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xue Zheng
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Junqi Hou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hongyu Tao
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Min Dai
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Shiming He
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
2
|
Zhao S, Dong G, Guo Y, Sun Y, Li M, Sha B, Huang W, Zhang Y, Du Y, Yan J, Ma Y, Yang R, Shi J, Li P, Hu T, Chen P. NSC632839 suppresses esophageal squamous cell carcinoma cell proliferation in vitro by triggering spindle assembly checkpoint-mediated mitotic arrest and CREB-Noxa-dependent apoptosis. Cancer Cell Int 2025; 25:198. [PMID: 40450316 DOI: 10.1186/s12935-025-03831-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 05/15/2025] [Indexed: 06/03/2025] Open
Abstract
OBJECTIVE Esophageal cancer is one of the most common digestive cancers in the world. Because of the limitation and resistence of the traditional chemotherapy drugs, it is important to explore new therapeutic targets and strategies for this refractory cancer. Recently, targeting deubiquitinases has emerged as a promising avenue for the development of anti-tumor drugs. However, the role and underlying mechanism of NSC632839, a broad-spectrum deubiquitinases inhibitor, in esophageal squamous cell carcinoma in vitro remain elusive. METHODS Cell Counting Kit-8 assay, colony formation assay, EdU proliferation experiment and cell morphology observation were used to detect the effect of NSC632839 on cell growth. Flow cytometry was employed to detect cell apoptosis and cell cycle arrest. Immunoblot and immunofluorescence was used to evaluate the expression level of cell cycle-, apoptosis-, and autophagy-related proteins. RESULTS NSC632839 inhibited the proliferation of Kyse30 and Kyse450 cells. Mechanistically, NSC632839 induced the formation of multipolar spindles, and its concomitant spindle assembly checkpoint-dependent mitotic arrest, followed by CREB-Noxa-mediated apoptosis. Reversine, a classical MPS1 kinase inhibitor known for its ability to inhibit the spindle assembly checkpoint, could rescue NSC632839-induced cell cycle arrest and apoptosis. Additionally, NSC632839 could trigger pro-survival autophagy. Combination of autophagy inhibitor, CQ and BafA1, with NSC632839 could induce stronger cell proliferation inhibition and apoptosis than NSC632839 alone. CONCLUSIONS These findings provided a novel anti-cancer mechanism of NSC632839 and highlighted it as a potential anti-tumor agent for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Shan Zhao
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- XinJiang Hotan College, Xinjiang, 848000, China
| | - Guihong Dong
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yixuan Guo
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yaxin Sun
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Sanquan College of Xinxiang Medical University, Xinxiang, 453003, China
| | - Miaomiao Li
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Beibei Sha
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450014, China
| | - Wenjing Huang
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuan Zhang
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yue Du
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jie Yan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
| | - Yangcheng Ma
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
| | - Ruiyi Yang
- The Nursing College of Zhengzhou University, Zhengzhou University, Zhengzhou, 450052, China
| | - Jianxiang Shi
- Precision Medicine Center, Henan Institute of Medical and Pharmaceutical Sciences & BGI College, Zhengzhou University, Zhengzhou, 450052, China
| | - Pei Li
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Tao Hu
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Ping Chen
- Academy of Medical Sciences, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
3
|
Pan Y, Yang H, Zhang J, Zhang R, Liu Y, Bie J, Chen Q, Qiao Y, Liu K, Song G. CircSLC22A3 inhibits the invasion and metastasis of ESCC via the miR-19b-3p/TRAK2 axis and by reducing the stability of m 6A-modified ACSBG1 mRNA. BMC Cancer 2025; 25:971. [PMID: 40448098 DOI: 10.1186/s12885-025-14390-8] [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: 03/26/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a major contributor to cancer-related deaths, driven by its invasive and metastatic nature. Circular RNAs (circRNAs) are increasingly recognized as regulators of cancer progression, primarily through miRNA sponging and interactions with RNA-binding proteins. Their dysregulation has been linked to the development of in various cancers. The present study aimed to investigate the potential involvement of circSLC22A3 in the pathogenesis of ESCC. METHODS CircSLC22A3 expression in ESCC tissues and cells was analyzed using transcriptome sequencing and RT-qPCR. Its circular structure was validated through Sanger sequencing, agarose gel electrophoresis, RNase R digestion, and random priming assays. Subcellular localization was determined by nucleoplasmic separation and fluorescence in situ hybridization (FISH). Clinical correlations were assessed via tissue microarrays. Functional roles of circSLC22A3 in ESCC progression were investigated through in vitro and in vivo assays. Downstream miR-19b-3p and target gene TRAK2 were screened by bioinformatics analysis and RT-qPCR, with binding confirmed via luciferase reporter assays. RNA pulldown combined with RNA immunoprecipitation (RIP) identified IGF2BP1 as a circSLC22A3-interacting protein. RNA-seq and RT-qPCR revealed ACSBG1 as a key downstream effector. IGF2BP1-mediated m6A modification of ACSBG1 was mapped by MeRIP-seq and RIP, with mRNA stability assessed via Actinomycin D assay. ACSBG1 expression and biological function in ESCC were confirmed by immunohistochemistry, RT-qPCR, and functional assays. RESULTS Significant downregulation of circSLC22A3 was observed in both ESCC tissues and cell lines. Overexpression of circSLC22A3 significantly reduced ESCC cells' migration and invasion capabilities. Mechanistic investigation revealed that circSLC22A3 played a pivotal role in the invasion and metastasis of esophageal cancer through distinct pathways. On one hand, circSLC22A3 functioned as a miR-19b-3p sponge to augment trafficking kinesin protein 2 (TRAK2) expression, while, on the other hand, circSLC22A3 formed a protein-RNA complex with IGF2BP1, resulting in the degradation of acyl-CoA synthetase bubblegum family member 1 (ACSBG1) mRNA through the recognition of m6A modification, thereby suppressing invasion and metastasis of ESCC. CONCLUSIONS The present study identified circSLC22A3 as a new tumor suppressor that inhibited ESCC progression through both the circSLC22A3/ miR-19b-3p/ TRAK2 and circSLC22A3/ IGF2BP1/ ACSBG1 axes.
Collapse
Affiliation(s)
- Yingjie Pan
- Institute of Tissue Engineering and Stem Cell Research, The Second Clinical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, P.R. China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, P.R. China
| | - Hang Yang
- Institute of Tissue Engineering and Stem Cell Research, The Second Clinical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, P.R. China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, P.R. China
| | - Jiayi Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, P.R. China
| | - Ruolan Zhang
- Institute of Tissue Engineering and Stem Cell Research, The Second Clinical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, P.R. China
| | - Yun Liu
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, P.R. China
| | - Jun Bie
- Institute of Tissue Engineering and Stem Cell Research, The Second Clinical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, P.R. China
| | - Qiaoling Chen
- Institute of Tissue Engineering and Stem Cell Research, The Second Clinical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, P.R. China
| | - Yan Qiao
- Institute of Tissue Engineering and Stem Cell Research, The Second Clinical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, P.R. China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cell Research, The Second Clinical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, P.R. China.
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, P.R. China.
| | - Guiqin Song
- Institute of Tissue Engineering and Stem Cell Research, The Second Clinical College of North Sichuan Medical College, Beijing Anzhen Nanchong Hospital of Capital Medical University & Nanchong Central Hospital, Nanchong, 637000, P.R. China.
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637100, P.R. China.
| |
Collapse
|
4
|
Song Q, Gao M, Weng Y, Zhuang X, Wu Y, Cui H, Ding N, Wang L, Bi S, Zhang L, Zhang W, Cui Y. Evolutionary adaptation and asymmetric inheritance of polyploid giant cancer cells in esophageal squamous cell carcinoma. Cancer Lett 2025:217818. [PMID: 40414521 DOI: 10.1016/j.canlet.2025.217818] [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: 01/18/2025] [Revised: 05/15/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Polyploid Giant Cancer Cells (PGCCs) play a critical role in tumor progression due to their distinctive biological behaviors. However, the mechanisms by which PGCCs regulate their composition and structure to adapt to dynamic environments during their formation remain poorly understood. In this study, we used multicolor labeling of major organelles in esophageal squamous cell carcinoma (ESCC) cells combined with high- and super-resolution time-lapse imaging to monitor induced PGCCs in three dimensions. In addition to abnormal PGCC division, we observed nuclear dynamics and transient cell-in-cell formations. PGCCs exhibited cell cycle abnormalities, including prolonged G1/S transitions, asynchronous micronuclei, and intranuclear mitosis. Notably, early progeny continued dividing despite cell cycle dysregulation, resulting in asymmetric offspring. Quantitative analysis of subcellular structures revealed asymmetric inheritance of organelles, particularly mitochondria and the Golgi apparatus, in recurrent cells. These adaptive mechanisms in PGCCs may also be relevant in the context of anticancer treatments, contributing to the heterogeneity of recurrent tumors arising from early PGCC progeny populations.
Collapse
Affiliation(s)
- Qiqin Song
- Cancer Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, P. R. China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China
| | - Mingwei Gao
- Cancer Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, P. R. China
| | - Yongjia Weng
- Cancer Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, P. R. China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China
| | - Xuehan Zhuang
- Cancer Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, P. R. China
| | - Yueguang Wu
- Cancer Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, P. R. China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China
| | - Heyang Cui
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, 999077, SAR, Hong Kong, China
| | - Ning Ding
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China
| | - Longlong Wang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China
| | - Shanshan Bi
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China
| | - Li Zhang
- Cancer Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, P. R. China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China
| | - Weimin Zhang
- Cancer Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, P. R. China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China; 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, 100142, P. R. China.
| | - Yongping Cui
- Cancer Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, 518035, P. R. China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518000, P. R. China.
| |
Collapse
|
5
|
Zhang X, Kang H, Li B, Xiong Y, Zheng S, Zhang D, Liu Y, Li S, Liu Y, Liu H, Gao Y, Ma L. Structural Optimization of 1,3-Diaryl-1,2,4-triazole-Capped Histone Deacetylase 6 Inhibitors to Obtain Novel Antiesophageal Cancer Candidates. J Med Chem 2025. [PMID: 40382720 DOI: 10.1021/acs.jmedchem.4c03231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Esophageal cancer, a leading global cancer, lacks effective therapies. Inhibition of histone deacetylase 6 (HDAC6) is a promising antitumor strategy, yet its role in esophageal cancer remains underexplored. Through structural optimization of our previously developed 1,3-diaryl-1,2,4-triazole-capped HDAC6 inhibitors, we identified compound 38k, exhibiting remarkably enhanced HDAC6 inhibition (IC50 = 3.12 nM) and 352-fold selectivity over HDAC1. Molecular docking analysis, CETSA, and BLI confirmed its strong HDAC6 binding. Moreover, 38k displayed robust in vitro and in vivo antiesophageal cancer efficacy, along with an advantageous pharmacokinetic and safety profile. Notably, combining 38k with a PI3K inhibitor synergistically enhanced the efficacy (75.02% tumor growth inhibition vs 50.94% monotherapy), likely by counteracting HDAC6 inhibition-induced PI3K/AKT activation. These findings validate HDAC6 as a therapeutic target and highlight 38k as a promising candidate for esophageal cancer treatment, particularly in combination regimens.
Collapse
Affiliation(s)
- Xinhui Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
- Newland Pharmaceutical Co., Ltd., Xuchang, Henan 461500, China
| | - Huiqin Kang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Bingqian Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuhan Xiong
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Shuxian Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Di Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuanfan Liu
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Shiyu Li
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Ying Liu
- The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, Henan 450001, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Liying Ma
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
- Key Laboratory of Cardio-cerebrovascular Drug, China Meheco Topfond Pharmaceutical Co., Zhumadian 463000, China
| |
Collapse
|
6
|
Reyila A, Gao X, Yu J, Nie Y. Insight into the role of DNA methylation in prognosis and treatment response prediction of gastrointestinal cancers. Epigenomics 2025; 17:475-488. [PMID: 40084815 PMCID: PMC12026041 DOI: 10.1080/17501911.2025.2476380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Gastrointestinal (GI) cancers impose a significant disease burden, underscoring the critical importance of accurate prognosis prediction and treatment response evaluation. DNA methylation, one of the most extensively studied epigenetic modifications, has gained prominence due to its reliable measurement across various sample types. Numerous studies have reported that DNA methylation was linked to the diagnosis, prognosis and treatment response in malignancies, including GI cancers. While its diagnostic role in GI cancers has been comprehensively reviewed. Recent research has increasingly highlighted its potential in prognosis prediction and treatment response evaluation. However, no existing reviews have exclusively focused on these two aspects. In this review, we retrieved relevant studies and included 230 of them in our discussion, thereby providing an overview of the clinical applicability of aberrant DNA methylation in these two fields among patients with esophageal, gastric, colorectal, pancreatic cancers, and hepatocellular carcinomas. Additionally, we discuss the limitations of the current literature and propose directions for future research. Specifically, we emphasize the need for standardized DNA methylation methodologies and advocate for the integration of gene panels, rather than single genes, to address tumor heterogeneity more effectively.
Collapse
Affiliation(s)
- Abudurousuli Reyila
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Fourth Military Medical University, Xi’an, Shaanxi, China
- National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xianchun Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Fourth Military Medical University, Xi’an, Shaanxi, China
- National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jun Yu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Fourth Military Medical University, Xi’an, Shaanxi, China
- National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Fourth Military Medical University, Xi’an, Shaanxi, China
- National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
7
|
Liu M, Huang Y, Tian H, Guo C, Liu Z, Liu A, Yang H, Li F, Duan L, Shen L, Wu Q, Shi C, Pan Y, Liu F, Liu Y, Chen H, Hu Z, Cai H, He Z, Ke Y. Absolute Risk Prediction for Esophageal Squamous Cell Carcinoma Adaptable to Regional Disease Burden across Diverse Regions. Cancer Epidemiol Biomarkers Prev 2025; 34:510-517. [PMID: 39869045 DOI: 10.1158/1055-9965.epi-24-1465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/03/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) exhibits a long latency period and has a significant geographic disparity in incidence, which underscores the need for models predicting the long-term absolute risk adaptable to the regional disease burden. METHODS A total of 31,883 participants in a large-scale population-based screening trial (Hua County, China) were enrolled to develop the model. Severe dysplasia and above cases identified at screening or follow-up were defined as the outcome. We calculated the absolute risk in three steps: (i) constructing a relative risk model using logistic regression, (ii) calculating the age-specific baseline hazard, and (iii) adjusting for the competing risk of all-cause death excluding ESCC. Flexible incidence rate parameters were integrated into the model to ensure its relevance across diverse regions worldwide. RESULTS A total of 295 severe dysplasia and above cases were detected. The relative risk model consisted of old age, male gender, an irregular meal pattern, a preference for hot or hard food, a BMI of less than 22 kg/m2, and ESCC family history. The AUC was 0.753 (95% confidence interval, 0.749-0.757). The averaged 5-and 10-year absolute risk were 0.53% and 1.30% among participants. Based on our model, we developed an online calculator and incorporated flexible incidence rate parameters, demonstrating ideal risk stratification tailored to regions with varying disease burdens (https://pkugenetics.shinyapps.io/escc_risk_prediction/). CONCLUSIONS We developed an absolute risk model to predict individualized long-term risk of ESCC, accounting for the local disease burden. IMPACT This model has the potential to mitigate the global burden of ESCC by enabling targeted screening and personalized prevention strategies.
Collapse
Affiliation(s)
- Mengfei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yi Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hongrui Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanhai Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhen Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Anxiang Liu
- Endoscopy Center, Anyang Cancer Hospital, Anyang, China
| | - Haijun Yang
- Department of Pathology, Anyang Cancer Hospital, Anyang, China
| | - Fenglei Li
- Hua County People's Hospital, Hua County, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing, 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, China
| | - Qi Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Endoscopy Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chao Shi
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yaqi Pan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Fangfang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ying Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Huanyu Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhe Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hong Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhonghu He
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yang Ke
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Genetics, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
8
|
Fatakhova K, Inayat F, Ali H, Patel P, Rehman AU, Afzal A, Sarfraz M, Sarfraz S, Nawaz G, Chaudhry A, Dhillon R, Dilibe A, Glazebnik B, Jones L, Glazer E. Gender disparities and woman-specific trends in Barrett's esophagus in the United States: An 11-year nationwide population-based study. World J Methodol 2025; 15:97512. [PMID: 40115400 PMCID: PMC11525896 DOI: 10.5662/wjm.v15.i1.97512] [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: 05/31/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Barrett's esophagus (BE) is a known premalignant precursor to esophageal adenocarcinoma (EAC). The prevalence rates continue to rise in the United States, but many patients who are at risk of EAC are not screened. Current practice guidelines include male gender as a predisposing factor for BE and EAC. The population-based clinical evidence regarding female gender remains limited. AIM To study comparative trends of gender disparities in patients with BE in the United States. METHODS A nationwide retrospective study was conducted using the 2009-2019 National Inpatient Sample (NIS) database. Patients with a primary or secondary diagnosis code of BE were identified. The major outcome of interest was determining the gender disparities in patients with BE. Trend analysis for respective outcomes for females was also reported to ascertain any time-based shifts. RESULTS We identified 1204190 patients with BE for the study period. Among the included patients, 717439 (59.6%) were men and 486751 (40.4%) were women. The mean age was higher in women than in men (67.1 ± 0.4 vs 66.6 ± 0.3 years, P < 0.001). The rate of BE per 100000 total NIS hospitalizations for males increased from 144.6 in 2009 to 213.4 in 2019 (P < 0.001). The rate for females increased from 96.8 in 2009 to 148.7 in 2019 (P < 0.001). There was a higher frequency of obesity among women compared to men (17.4% vs 12.6%, P < 0.001). Obesity prevalence among females increased from 12.3% in 2009 to 21.9% in 2019 (P < 0.001). A lower prevalence of smoking was noted in women than in men (20.8% vs 35.7%, P < 0.001). However, trend analysis showed an increasing prevalence of smoking among women, from 12.9% in 2009 to 30.7% in 2019 (P < 0.001). Additionally, there was a lower prevalence of alcohol abuse, Helicobacter pylori (H. pylori), and diabetes mellitus among females than males (P < 0.001). Trend analysis showed an increasing prevalence of alcohol use disorder and a decreasing prevalence of H. pylori and diabetes mellitus among women (P < 0.001). CONCLUSION The prevalence of BE among women has steadily increased from 2009 to 2019. The existing knowledge concerning BE development has historically focused on men, but our findings show that the risk in women is not insignificant.
Collapse
Affiliation(s)
- Karina Fatakhova
- Division of Gastroenterology and Hepatology, Mather Hospital and Zucker School of Medicine at Hofstra University, Port Jefferson, NY 11777, United States
| | - Faisal Inayat
- Department of Internal Medicine, Allama Iqbal Medical College, Lahore, Punjab 54550, Pakistan
| | - Hassam Ali
- Division of Gastroenterology and Hepatology, East Carolina University Brody School of Medicine, Greenville, NC 27834, United States
| | - Pratik Patel
- Division of Gastroenterology and Hepatology, Mather Hospital and Zucker School of Medicine at Hofstra University, Port Jefferson, NY 11777, United States
| | - Attiq Ur Rehman
- Division of Gastroenterology and Hepatology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, United States
| | - Arslan Afzal
- Division of Gastroenterology and Hepatology, East Carolina University Brody School of Medicine, Greenville, NC 27834, United States
| | - Muhammad Sarfraz
- Division of Gastroenterology and Hepatology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, United States
| | - Shiza Sarfraz
- Department of Internal Medicine, East Carolina University Brody School of Medicine, Greenville, NC 27834, United States
| | - Gul Nawaz
- Department of Internal Medicine, Allama Iqbal Medical College, Lahore, Punjab 54550, Pakistan
| | - Ahtshamullah Chaudhry
- Department of Internal Medicine, St. Dominic's Hospital, Jackson, MS 39216, United States
| | - Rubaid Dhillon
- Department of Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic Foundation, Cleveland, OH 44195, United States
| | - Arthur Dilibe
- Department of Internal Medicine, East Carolina University Brody School of Medicine, Greenville, NC 27834, United States
| | - Benjamin Glazebnik
- Department of Internal Medicine, Mather Hospital and Hofstra University Zucker, School of Medicine, Port Jefferson, NY 11777, United States
| | - Lindsey Jones
- Department of Internal Medicine, Mather Hospital and Hofstra University Zucker, School of Medicine, Port Jefferson, NY 11777, United States
| | - Emily Glazer
- Division of Gastroenterology and Hepatology, Mather Hospital and Zucker School of Medicine at Hofstra University, Port Jefferson, NY 11777, United States
| |
Collapse
|
9
|
Damiano OM, Stevens AJ, Kenwright DN, Seddon AR. Chronic Inflammation to Cancer: The Impact of Oxidative Stress on DNA Methylation. FRONT BIOSCI-LANDMRK 2025; 30:26142. [PMID: 40152377 DOI: 10.31083/fbl26142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 03/29/2025]
Abstract
The genomic landscape of cancer cells is complex and heterogeneous, with aberrant DNA methylation being a common observation. Growing evidence indicates that oxidants produced from immune cells may interact with epigenetic processes, and this may represent a mechanism for the initiation of altered epigenetic patterns observed in both precancerous and cancerous cells. Around 20% of cancers are linked to chronic inflammatory conditions, yet the precise mechanisms connecting inflammation with cancer progression remain unclear. During chronic inflammation, immune cells release oxidants in response to stimuli, which, in high concentrations, can cause cytotoxic effects. Oxidants are known to damage DNA and proteins and disrupt normal signalling pathways, potentially initiating a sequence of events that drives carcinogenesis. While research on the impact of immune cell-derived oxidants on DNA methylation remains limited, this mechanism may represent a crucial link between chronic inflammation and cancer development. This review examines current evidence on inflammation-associated DNA methylation changes in cancers related to chronic inflammation.
Collapse
Affiliation(s)
- Olivia M Damiano
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Aaron J Stevens
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Diane N Kenwright
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Annika R Seddon
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, 8011 Christchurch, New Zealand
| |
Collapse
|
10
|
Chen X, Liu HY, Zhou WB, Zhang LL, Huang J, Bao DW. Hypoxia-inducible factor 1-alpha and lactate dehydrogenase-A axis in metabolic changes and aggression in esophageal squamous-cell carcinoma. World J Gastrointest Oncol 2025; 17:103450. [PMID: 40092940 PMCID: PMC11866222 DOI: 10.4251/wjgo.v17.i3.103450] [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: 11/27/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Esophageal squamous-cell carcinoma (ESCC) is a highly aggressive cancer, predominantly affecting populations in Eastern Asia and parts of Africa. Its pathogenesis is influenced by both genetic and environmental factors. Despite recent therapeutic advances, survival rates remain dismal, underscoring an urgent need for novel therapeutic targets. AIM To investigate the role of hypoxia-inducible factor 1-alpha (HIF1A) in the progression of ESCC and its impact on the metabolic enzyme lactate dehydrogenase A (LDHA), which is crucial for the glycolytic pathway in hypoxic tumor environments. METHODS Utilizing transcriptomic data from multiple public databases, we analyzed differential gene expression and conducted gene ontology and transcription factor network analyses. The regulatory impact of HIF1A on LDHA was specifically examined through integrative analysis with HIF1A ChIP-seq data and confirmed via siRNA-mediated knockdown experiments in ESCC cell lines. RESULTS Our findings reveal a significant upregulation of HIF1A in ESCC tissues, associated with poor prognosis. HIF1A directly regulates LDHA, enhancing glycolysis under hypoxic conditions and contributing to tumor aggressiveness. Knockdown of HIF1A in cell lines not only reduced LDHA expression but also altered key pathways related to cell cycle and apoptosis. CONCLUSION The critical role of the HIF1A-LDHA axis in ESCC highlights its potential as a therapeutic target, underscoring the need for future clinical trials to validate the efficacy of HIF1A inhibitors in enhancing treatment outcomes.
Collapse
Affiliation(s)
- Xia Chen
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai’an 223300, Jiangsu Province, China
| | - Hai-Yan Liu
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai’an 223300, Jiangsu Province, China
| | - Wu-Bi Zhou
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai’an 223300, Jiangsu Province, China
| | - Li-Li Zhang
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai’an 223300, Jiangsu Province, China
| | - Jian Huang
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai’an 223300, Jiangsu Province, China
| | - Da-Wei Bao
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai’an 223300, Jiangsu Province, China
| |
Collapse
|
11
|
Subhaharan D, Kakkadasam Ramaswamy P, Jones M, John S. Implementing educational interventions and key performance measures sustains quality of endoscopic assessment in patients with Barrett's esophagus. Endosc Int Open 2025; 13:a25420618. [PMID: 40109315 PMCID: PMC11922174 DOI: 10.1055/a-2542-0618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025] Open
Abstract
Background and study aims Quality metrics for Barrett's esophagus (BE) are anticipated to improve outcomes for patients through earlier detection of neoplasia. The European Society of Gastrointestinal Endoscopy has developed guidelines to homogenize endoscopic quality in BE. Our study aimed to assess the impact of recommended key performance measures (KPMs) and their sustainability. Patients and methods A single-center, retrospective study (Phase 1) was conducted over 8 weeks. The KPMs assessed were: 1) pre-procedure metrics including indication, consent, safety checklist (target of 100%); and 2) Prague classification, Seattle protocol, or targeted biopsies, inspection time of 1 minute per cm, advanced imaging and surveillance recommendations (target of 90%). Following baseline analysis, multimodal educational interventions were implemented and repeated at 6-month intervals. Repeat analysis was performed at 6 months and 1 and 3 years (Phases 2, 3 and 4 respectively). Results In Phase 1, 39 patients with BE underwent endoscopy. Phase 2 evaluated 40 patients with BE. Phase 3 analyzed 59 patients with BE, and Phase 4 identified 34 patients with BE. Pre-procedure metrics were met in 100% of patients across the 3-year period. Baseline analysis displayed suboptimal performance at 45% to 75% for all other KPMs. However, after regular multimodal educational interventions, quality standards significantly improved and were able to be maintained over all phases, achieving pre-set targets of >9 0% for all KPMs except one. Conclusions Sustaining improvements in quality metrics in Barrett's endoscopy is important. Our study suggests that regular, replicable education interventions have a positive effect and allow sustained long-term improvements in quality metrics.
Collapse
Affiliation(s)
- Deloshaan Subhaharan
- Department of Digestive Health, Gold Coast University Hospital, Gold Coast, Australia
- Health Sciences and Medicine, Bond University Ltd, Gold Coast, Australia
| | | | - Mark Jones
- Health Sciences and Medicine, Bond University Ltd, Gold Coast, Australia
| | - Sneha John
- Department of Digestive Health, Gold Coast University Hospital, Gold Coast, Australia
- Health Sciences and Medicine, Bond University Ltd, Gold Coast, Australia
| |
Collapse
|
12
|
Veziant J, Routier B, Piessen G. [The role of surgery in preventing esophageal cancer]. Bull Cancer 2025; 112:277-285. [PMID: 40049796 DOI: 10.1016/j.bulcan.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 05/13/2025]
Abstract
The prognosis for esophageal cancer remains poor because it is often diagnosed late and patients often have unfavourable backgrounds. This is reflected in standardised 5-year net survival rates of no more than 20%, regardless of gender. Unlike gastric cancer (CDH1 gene mutation), there is currently no genetic predisposition to esophageal cancer that would justify prophylactic esophagectomy. Primary prevention by identifying and managing modifiable risk factors is therefore the best strategy for preventing esophageal cancer. The role of surgery in the prevention of esophageal cancer is discussed in this review. Although recommended, the value of antireflux surgery (fundoplication) for Barrett's esophagus with the sole aim of reducing the risk of esophageal adenocarcinoma remains controversial. With regard to bariatric surgery, national cohort studies report an equivalent or reduced incidence of esophageal adenocarcinoma in operated patients compared with non-operated obese patients. However, given the significant increase in the number of bariatric procedures, further studies with longer follow-up are needed. In addition, although surgical myotomy is a truly effective therapeutic option for the treatment of achalasia in the first-line setting, its impact on the risk of esophageal cancer remains uncertain and poorly studied.
Collapse
Affiliation(s)
- Julie Veziant
- Hôpital Claude-Huriez, université de Lille, CHU de Lille, service de chirurgie digestive et oncologique, 59000 Lille, France; University Lille, CHU de Lille, CNRS, UMR9020-U1277-CANTHER-Cancer, Inserm, 59000 Lille, France.
| | - Baptiste Routier
- Hôpital Claude-Huriez, université de Lille, CHU de Lille, service de chirurgie digestive et oncologique, 59000 Lille, France
| | - Guillaume Piessen
- Hôpital Claude-Huriez, université de Lille, CHU de Lille, service de chirurgie digestive et oncologique, 59000 Lille, France; University Lille, CHU de Lille, CNRS, UMR9020-U1277-CANTHER-Cancer, Inserm, 59000 Lille, France
| |
Collapse
|
13
|
Akhtar S, Al-Shammari A, Al-Huraiti M, Al-Anjery F. Age-period-cohort modeling of oesophageal carcinoma risk in a middle eastern country: 1980-2019. J Public Health (Oxf) 2025; 47:e59-e66. [PMID: 39674677 DOI: 10.1093/pubmed/fdae311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/29/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Understanding of the factors influencing oesophageal cancer trends is crucial. Therefore, this cross-sectional cohort study sought to disentangle the age, period and cohort effects on the trends of oesophageal cancer in Kuwait. METHODS The data on incident oesophageal carcinoma cases diagnosed between January 1, 1980, through December 31, 2019, and reference population were obtained. Age-period-cohort (APC) analysis was conducted using a loglinear Poisson regression model. RESULTS A total of 496 oesophageal carcinoma cases in 12.8 million person-years (i.e. squamous-cell carcinoma, 269, 54.23%), adenocarcinoma,147, 29.64% and unspecified cases, 80,16.13%) were diagnosed. The overall age-standardized incidence rate (per 105 person-years) of oesophageal carcinoma during the study period was 10.51 (95% CI: 6.62-14.41). The APC analysis results showed that the age and birth cohort effects were the significant determinants of declining, and subsequently steadying the oesophageal carcinoma incidence rates. CONCLUSIONS A substantial decline in oesophageal carcinoma incidence rates was recorded, which significantly varied in all three temporal dimensions. The observed birth cohort patterns suggest changing lifestyle and dietary patterns seem to be responsible for decreasing oesophageal carcinoma risk in Kuwait. Future studies may look for the component causes maintaining the endemicity of oesophageal carcinoma risk in this and similar countries in the region.
Collapse
Affiliation(s)
- Saeed Akhtar
- Department of Community Medicine and Behavioural Sciences, College of Medicine, Kuwait University, PO Box 24923, Safat 13110, Kuwait
| | - Ahmad Al-Shammari
- Department of Surgery, Faculty of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | | | - Fouzan Al-Anjery
- Ministry of Health, Jamal Abdel Nasser Street, Sulaibkhat 13001, Kuwait
| |
Collapse
|
14
|
Lawson NM, Ye L, Cho CY, Zhao B, Mitchell T, Martín-Barrio I, Beernaert B, Gupta A, Banu M, Lissanu Y, Shaffer S, Tawbi H, Li J, Gule-Monroe MK, Alvarez-Breckenridge CA, Huse JT, Murphy MB, Yin F, Lang FF, Parkes EE, Weinberg JS, Akdemir KC. Recurrent ERBB2 alterations are associated with esophageal adenocarcinoma brain metastases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.19.25322558. [PMID: 40061311 PMCID: PMC11888521 DOI: 10.1101/2025.02.19.25322558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Brain metastases in esophageal adenocarcinoma (EAC) patients are associated with poor prognosis and remain understudied. We performed multi-omics analysis with whole-genome sequencing and single-cell spatial transcriptomics on the brain metastases and matched primary tumors. Our analysis identified ERBB2 as a recurrent oncogene in EAC brain metastases, with 9 out of 10 cases harboring amplifications. Single-cell whole-genome and multi-region sequencing revealed that ERBB2 alterations, occur early during disease progression and are associated with monoclonal seeding. Although the median survival in our cohort was 13 months, one patient on HER2 antibody-drug conjugate therapy remains a long-term survivor beyond 34 months. Interestingly, the sole patient without an ERBB2 alteration had JAK2 deletion, high T cell infiltration in the brain lesion, and survived 35 months after immune checkpoint therapy. Our findings have significant clinical implications for the treatment and management of EAC brain metastases.
Collapse
Affiliation(s)
- Nora M. Lawson
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Lingqun Ye
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Chae Yun Cho
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Bo Zhao
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas Mitchell
- Department of Genetics, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Archit Gupta
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Matei Banu
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Yonathan Lissanu
- Department of Thoracic Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Sydney Shaffer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Li
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Jason T. Huse
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Feng Yin
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Frederick F. Lang
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Kadir C. Akdemir
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
- Institute for Data Science of Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
- Lead Author
| |
Collapse
|
15
|
de Melo Viana TC, Nakamura ET, Park A, Filardi KFXC, de Almeida Leite RM, Baltazar LFSR, Usón Junior PLS, Tustumi F. Molecular Abnormalities and Carcinogenesis in Barrett's Esophagus: Implications for Cancer Treatment and Prevention. Genes (Basel) 2025; 16:270. [PMID: 40149421 PMCID: PMC11942460 DOI: 10.3390/genes16030270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/16/2025] [Accepted: 02/23/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Barrett's esophagus (BE) is described by the transformation of the normal squamous epithelium into metaplastic columnar epithelium, driven by chronic gastroesophageal reflux disease (GERD). BE is a recognized premalignant condition and the main precursor to esophageal adenocarcinoma (EAC). Understanding the molecular mechanisms underlying BE carcinogenesis is crucial for improving prevention, surveillance, and treatment strategies. METHODS This narrative review examines the molecular abnormalities associated with the progression of BE to EAC. RESULTS This study highlights inflammatory, genetic, epigenetic, and chromosomal alterations, emphasizing key pathways and biomarkers. BE progression follows a multistep process involving dysplasia and genetic alterations such as TP53 and CDKN2A (p16) mutations, chromosomal instability, and dysregulation of pathways like PI3K/AKT/mTOR. Epigenetic alterations, including aberrant microRNA expression or DNA methylation, further contribute to this progression. These molecular changes are stage-specific, with some alterations occurring early in BE during the transition to high-grade dysplasia or EAC. Innovations in chemoprevention, such as combining proton pump inhibitors and aspirin, and the potential of antireflux surgery to halt disease progression are promising. Incorporating molecular biomarkers into surveillance strategies and advancing precision medicine may enable earlier detection and personalized treatments. CONCLUSIONS BE is the primary preneoplastic condition for EAC. A deeper understanding of its molecular transformation can enhance surveillance protocols, optimize the management of gastroesophageal reflux inflammation, and refine prevention and therapeutic strategies, ultimately contributing to a reduction in the global burden of EAC.
Collapse
Affiliation(s)
| | | | - Amanda Park
- Department of Evidenced-Based Medicine, Centro Universitário Lusíada, Santos 11050-071, Brazil
| | | | | | | | | | - Francisco Tustumi
- Department of Gastroenterology, Universidade de Sao Paulo, Sao Paulo 05508-220, Brazil
- Department of Health Sciences, Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
| |
Collapse
|
16
|
Yao L, Wu P, Yao F, Huang B, Zhong F, Wang X. ZCCHC4 regulates esophageal cancer progression and cisplatin resistance through ROS/c-myc axis. Sci Rep 2025; 15:5149. [PMID: 39934309 PMCID: PMC11814405 DOI: 10.1038/s41598-025-89628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Zinc finger CCHC-type containing 4 (ZCCHC4) is a newly discovered N6-methyladenosine (m6A) RNA methyltransferase (MTase), which possesses an m6A MTase domain and an RNA-binding protein (RBP) Znf domain. Aberrantly expressed ZCCHC4 has been found to be correlated with poor prognosis and chemoresistance in various tumors, such as hepatocellular carcinoma, lung cancer and colorectal cancer. However, the expression and functional analysis of the role of ZCCHC4 in esophageal cancer (ESCA) is still elusive. The expression of ZCCHC4 in esophageal cancer tissues was evaluated by qPT-PCR and western blot. Serum esophageal tumor markers are detected by electrochemiluminescence immunoassay. Relationship between ZCCHC4 expression and pathway enrichment analysis were analyzed by R. The reactive oxygen species (ROS), cell proliferation, cell cycle and apoptosis of ZCCHC4 in esophageal squamous cell carcinoma (ESCC) cells tested by CCK8 assay and flow cytometry assay. Aberrant expression of ZCCHC4 is associated with cancer stages, lymph node metastasis (LNM), and tumor histology, and poorer Overall Survival (OS) in esophageal cancer. The mRNA level of ZCCHC4 in esophageal cancer patients correlates with serum carcinoembryonic antigen (CEA) levels, Squamous Cell Carcinoma (SCC) markers, and tissue polypeptide antigen (TPA) levels. Knockdown of ZCCHC4 induces DNA damage, leading to an elevation of reactive oxygen species (ROS), which in turn triggers S-phase arrest, enhances apoptosis, augments sensitivity to cisplatin treatment, and inhibits proliferation in esophageal cancer cells. Conversely, overexpression of ZCCHC4 promotes proliferation, inhibits apoptosis, and increases resistance to cisplatin in esophageal cancer cells. Furthermore, scavenging ROS reverses the effects of ZCCHC4 downregulation on both proliferation and apoptosis in esophageal cancer cells. Additionally, downregulation of ZCCHC4 inhibits the progression of esophageal cancer and reduces cisplatin resistance in vivo. In summary, downregulation of ZCCHC4 leads to increased sensitivity of ESCC cells to cisplatin, inhibits proliferation, and promotes apoptosis in esophageal cancer cells, potentially via the ROS/c-myc axis. The study suggests a potential adjunctive role for ZCCHC4 in the diagnosis and treatment of esophageal cancer and aids in further understanding the underlying mechanisms in ESCA progression.
Collapse
Affiliation(s)
- Lihua Yao
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China
| | - Piao Wu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Fangyi Yao
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China
| | - Fangmin Zhong
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China.
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, , Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
17
|
Laun SE, Kann L, Braun J, Pierre F, Kim S, Gilbert S, Lunz D, Kalra A, Ma K, Cheng Y, Leggett CL, Zaidi AH, Omstead AN, Korman L, Jobe B, Perpetua L, Greenwald BD, Maddala T, Meltzer SJ. Spatiotemporal Study of a Risk-Stratification Epigenetic-Based Biomarker Assay in Patients With Barrett Esophagus. Am J Gastroenterol 2025; 120:00000434-990000000-01586. [PMID: 39933887 PMCID: PMC12124209 DOI: 10.14309/ajg.0000000000003367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION Barrett esophagus (BE) is the strongest known risk factor for developing esophageal adenocarcinoma (EAC), the second-most lethal cancer in the United States. Esopredict is a novel validated methylation-based biomarker assay that provides precise quantification of neoplastic progression risk in BE patients. Inherit challenges, including tissue heterogeneity, sampling error, interobserver variability, and inconsistent adherence to surveillance biopsy guidelines, may affect the predictive value results of Esopredict obtained at different anatomic locations or different sampling time points. METHODS To investigate the spatiotemporal performance of Esopredict across multiple spatiotemporal sampling points, we profiled 220 biopsies obtained from 58 BE patients, including 11 patients with overlapping spatial and temporal biopsies. We focused on spatial profiling (i.e., multiple biopsies obtained at several anatomic locations during a single endoscopy) and temporal profiling (i.e., biopsies obtained from multiple endoscopies performed at different time points). Each patient had an initial histologic diagnosis of nondysplastic Barrett esophagus, indefinite for dysplasia, or low-grade dysplasia. Final follow-up (endpoint) biopsies showed either high-grade dysplasia or EAC (progressors), or nondysplastic Barrett esophagus, indefinite for dysplasia, or low-grade dysplasia (nonprogressors). Biopsies were analyzed with Esopredict to compute a progression risk score, which quantified the likelihood of future progression to high-grade dysplasia or EAC within 5 years. RESULTS In 52 spatially profiled patients, Esopredict demonstrated a sensitivity of 81% (17/21 progressor patients), based on the highest-scoring biopsy from each patient; sensitivity increased to 100% (12/12) when end point biopsies occurred within 5 years of the index (initial) biopsy. In 28 temporally profiled patients, sensitivity was 100% (8/8 patients), based on the biopsy performed at the time point closest to the end point biopsy. DISCUSSION Esopredict showed high predictive performance in multiple spatiotemporal samples in BE patients. These data further support the use of Esopredict as a robust test to distinguish high-risk BE patients, who may benefit from endoscopic eradication therapy or increased surveillance frequency, from low-risk patients, who may be candidates for less frequent surveillance and noninterventional observation.
Collapse
Affiliation(s)
| | | | | | | | - Suji Kim
- Previse, Baltimore, Maryland, USA
| | | | | | - Andrew Kalra
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ke Ma
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Jefferson Einstein Philadelphia Hospital, Philadelphia, Pennsylvania, USA
| | - Yulan Cheng
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cadman L. Leggett
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ali H. Zaidi
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Ashten N. Omstead
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Louis Korman
- Capital Digestive Care, Chevy Chase, Maryland, USA
| | - Blair Jobe
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Esophageal Institute, Allegheny Health Network, Pittsburg, Pennsylvania, USA
| | - Lorrie Perpetua
- Research Tissue Biorepository Core Facility, University of Connecticut, Storrs, Connecticut, USA
| | - Bruce D. Greenwald
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Stephen J. Meltzer
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Gu J, Zhang S, Lin D, Wang W, Cheng J, Zheng Q, Wang H, Tan L. Suppressing SENP1 inhibits esophageal squamous carcinoma cell growth via SIRT6 SUMOylation. Cell Oncol (Dordr) 2025; 48:67-81. [PMID: 38954215 PMCID: PMC11850494 DOI: 10.1007/s13402-024-00956-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2024] [Indexed: 07/04/2024] Open
Abstract
PURPOSE Esophageal squamous cell carcinoma (ESCC) is a prevalent tumor in the gastrointestinal tract, but our understanding of the molecular mechanisms underlying ESCC remains incomplete. Existing studies indicate that SUMO specific peptidase 1 (SENP1) plays a crucial role in the development and progression of various malignant tumors through diverse molecular mechanisms. However, the functional mechanism and clinical implications of SENP1 in the progression of ESCC remain unclear. METHODS Bulk RNA-Sequencing (RNA-seq) was used to compare potential genes in the esophageal tissues of mice with ESCC to the control group. The up-regulated SENP1 was selected. The protein level of SENP1 in ESCC patient samples was analyzed by immunohistochemistry and western blot. The potential prognostic value of SENP1 on overall survival of ESCC patients was examined using tissue microarray analysis and the Kaplan-Meier method. The biological function was confirmed through in vitro and in vivo knockdown approaches of SENP1. The role of SENP1 in cell cycle progression and apoptosis of ESCC cells was analyzed by flow cytometry and western blot. The downstream signaling pathways regulated by SENP1 were investigated via using RNA-Seq. SENP1-associated proteins were identified through immunoprecipitation. Overexpression of Sirtuin 6 (SIRT6) wildtype and mutant was performed to investigate the regulatory role of SENP1 in ESCC progression in vitro. RESULTS Our study discovered that SENP1 was upregulated in ESCC tissues and served as a novel prognostic factor. Moreover, SENP1 enhanced cell proliferation and migration of ESCC cell lines in vitro, as well as promoted tumor growth in vivo. Thymidine kinase 1 (TK1), Geminin (GMNN), cyclin dependent kinase 1(CDK1), and cyclin A2 (CCNA2) were identified as downstream genes of SENP1. Mechanistically, SENP1 deSUMOylated SIRT6 and subsequently inhibited SIRT6-mediated histone 3 lysine 56 (H3K56) deacetylation on those downstream genes. SIRT6 SUMOylation mutant (4KR) rescued the growth inhibition upon SENP1 depletion. CONCLUSIONS SENP1 promotes the malignant progression of ESCC by inhibiting the deacetylase activity of SIRT6 pathway through deSUMOylation. Our findings suggest that SENP1 may serve as a valuable biomarker for prognosis and a target for therapeutic intervention in ESCC.
Collapse
Affiliation(s)
- Jianmin Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shaoyuan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dong Lin
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Wenhan Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinke Cheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Quan Zheng
- Center for Singl-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hao Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
19
|
Chen L, Tang J, Chang Y, Hang D, Ji J, Chen G. SMURF1 leads to the β-catenin signaling-mediated progression of esophageal squamous carcinoma by losing PATZ1-induced CCNG2 transcription. Biochem Pharmacol 2025; 232:116688. [PMID: 39617210 DOI: 10.1016/j.bcp.2024.116688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/10/2024] [Accepted: 11/28/2024] [Indexed: 12/16/2024]
Abstract
Cyclin G2 (CCNG2), a known inhibitor of cell cycle progression, has been identified as a suppressor for the canonical β-catenin pathway. This study explores the impact of CCNG2 on β-catenin activity and malignant characteristics of esophageal squamous cell carcinoma (ESCC) cells, and the mechanism behind CCNG2 dysregulation. In ESCC tissues and cells, CCNG2 was under-expressed and associated with poor clinical outcomes, whereas β-catenin showed an opposite trend. Inducing CCNG2 overexpression in ESCC cells led to a reduction in β-catenin levels, which in turn suppressed proliferation, cell cycle progression, migration, invasion, stemness, and tumorigenesis. Additionally, it enhanced the cytotoxicity and proliferation of T cells in co-culture systems. However, these beneficial effects were negated by the Wnt signaling agonist BML-284. Furthermore, PATZ1 was found as a transcription factor promoting CCNG2 transcription. However, the PATZ1 protein in ESCC cells was degraded by SMURF1. Silencing of SMURF1 restored CCNG2 expression and inhibited β-catenin, thereby suppressing the malignant phenotype of ESCC cells and reducing T cell exhaustion. Yet, these effects were blocked by further silencing of PATZ1. In summary, this research demonstrates that SMURF1 activates β-catenin signaling by suppressing the PATZ1/CCNG2 axis, thereby promoting the progression of ESCC.
Collapse
Affiliation(s)
- Lingling Chen
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China
| | - Jie Tang
- Department of Gastroenterology, Jiangwan Hospital, Hongkou District, Shanghai 200434, PR China
| | - Yunli Chang
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China
| | - Dongyun Hang
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China
| | - Jieru Ji
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China.
| | - Guoyu Chen
- Department of Gastroenterology, Pudong New Area People's Hospital, Shanghai 201299, PR China.
| |
Collapse
|
20
|
Li J, Wang Y, Wei S, Xu S, Dai S, Zhang L, Tian Z, Zhao L, Lv H. NEK2 Promotes ESCC Malignant Progression by Inhibiting Cellular Senescence via the FOXM1/c-Myc/p27 Signaling Pathway. Mol Carcinog 2025; 64:244-259. [PMID: 39503194 DOI: 10.1002/mc.23839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 01/15/2025]
Abstract
Never in mitosis gene A (NIMA)-related kinase 2 (NEK2) is a crucial serine-threonine kinase involved in the process of cell mitosis. However, the precise relationship between NEK2 and esophageal squamous cell carcinoma (ESCC) remains inadequately understood. NEK2 expression in ESCC tissues was assessed through bioinformatics analysis, reverse transcription-quantitative PCR (RT-qPCR) and immunohistochemistry, revealing a correlation with ESCC patient prognosis. Cultured ESCC cells and human normal esophageal epithelial cells (HEEC) were used to investigate the effects of NEK2 knockdown on the development and progression of ESCC by integrated confluence algorithm, colony formation, wound-healing, transwell, and ESCC xenograft tumor model, in vitro and in vivo. In ESCC tissues, NEK2 was found to be significantly upregulated, and its expression correlated with poor prognosis in ESCC patients. NEK2 may facilitate ESCC development by regulating cell proliferation, migration, and invasion. Additionally, results from in vivo experiments suggested that NEK2 knockdown can inhibit tumor growth. Moreover, forkhead box M1 (FOXM1) was identified as a potential downstream target of NEK2 in the regulation of ESCC, with its overexpression reversing the effects of NEK2 knockdown on ESCC. Mechanistic studies also indicated that NEK2 may promote the malignant progression of ESCC by inhibiting cellular senescence through the activation of the FOXM1/c-Myc/p27 signaling pathways, which may provide a novel perspective for the management of ESCC.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yaojie Wang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sisi Wei
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shi Xu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Suli Dai
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li Zhang
- Department of Geriatric, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Tumor Gene Diagnosis, Prevention, and Therapy of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huilai Lv
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
21
|
Zheng H, Wu R, Zhang G, Wang Q, Li Q, Zhang L, Li H, Wang Y, Xie L, Guo X. Nomograms for prognosis prediction in esophageal adenocarcinoma: realities and challenges. Clin Transl Oncol 2025; 27:449-457. [PMID: 39083141 DOI: 10.1007/s12094-024-03589-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: 04/01/2024] [Accepted: 06/30/2024] [Indexed: 02/01/2025]
Abstract
Prognostic assessment is of great significance for individualized treatment and care of cancer patients. Although the TNM staging system is widely used as the primary prognostic classifier for solid tumors in clinical practice, the complexity of tumor occurrence and development requires more personalized probability prediction models than an ordered staging system. By integrating clinical, pathological, and molecular factors into digital models through LASSO and Cox regression, a nomogram could provide more accurate personalized survival estimates, helping clinicians and patients develop more appropriate treatment and care plans. Esophageal adenocarcinoma (EAC) is a common pathological subtype of esophageal cancer with poor prognosis. Here, we screened and comprehensively reviewed the studies on EAC nomograms for prognostic prediction, focusing on performance evaluation and potential prognostic factors affecting survival. By analyzing the strengths and limitations of the existing nomograms, this study aims to provide assistance in constructing high-quality prognostic models for EAC patients.
Collapse
Affiliation(s)
- Hong Zheng
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
| | - Rong Wu
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
| | - Guosen Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
| | - Qiang Wang
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
- School of Software, Henan University, Kaifeng, China
| | - Qiongshan Li
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
| | - Lu Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
| | - Huimin Li
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
| | - Yange Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
| | - Longxiang Xie
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute of Biomedical Informatics, Henan University, Kaifeng, China
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China
| | - Xiangqian Guo
- School of Basic Medical Sciences, Henan University, Kaifeng, China.
- Institute of Biomedical Informatics, Henan University, Kaifeng, China.
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, China.
| |
Collapse
|
22
|
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
|
23
|
Du C, Hu Y, Yang X, Zhang Z, Gu J, Zhang T, Wang R, Zhang S, Tan L, Yu G. SUMO-Specific Peptidase 5 Promotes Oesophageal Squamous Cell Carcinoma Growth through the NF-κB- SLC1A3 Axis. FRONT BIOSCI-LANDMRK 2025; 30:27047. [PMID: 39862098 DOI: 10.31083/fbl27047] [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: 10/20/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 01/30/2025]
Abstract
BACKGROUND This study investigates the role of small ubiquitin-like modifier (SUMO)-specific peptidase 5 (SENP5), a key regulator of SUMOylation, in esophageal squamous cell carcinoma (ESCC), a lethal disease, and its underlying molecular mechanisms. METHODS Differentially expressed genes between ESCC mouse oesophageal cancer tissues and normal tissues were analysed via RNA-seq; among them, SENP5 expression was upregulated, and this gene was selected for further analysis. Immunohistochemistry and western blotting were then used to validate the increased protein level of SENP5 in both mouse and human ESCC samples. The Kaplan‒Meier method and multivariate analysis were used to analyse the relationship between SENP5 expression and ESCC prognosis. Stable SENP5-knockdown (KD) cell lines and conditional knockout (cKO) mice were established to verify the biological function of SENP5. Further RNA-seq comparisons between short hairpin SENP5 (shSENP5)- and short hairpin negative control (shNC)-transfected ESCC cell lines were conducted, and the nuclear factor kappa B (NF-κB)-SLC1A3 axis was identified through bioinformatics analysis. The correlation of SENP5 with signalling pathway components was validated via real-time quantitative PCR (qPCR), western blotting (WB), and immunoprecipitation. RESULTS Our study revealed that SENP5 was upregulated in human and mouse ESCC samples, and clinical data analysis revealed a correlation between high SENP5 expression and poor patient prognosis. SENP5 knockdown inhibited tumorigenesis and growth in vivo and suppressed the proliferation, migration, and invasion of ESCC cell lines in vitro. Our study also revealed that SENP5 knockdown enhanced the SUMO1-mediated SUMOylation of NF-kappa-B inhibitor alpha (IκBα), thereby inhibiting the activation of the NF-κB-SLC1A3 axis, which subsequently suppresses ESCC cell energy metabolism and impedes ESCC progression. CONCLUSIONS Suppression of SENP5 slows the development of ESCC by inhibiting the NF-κB‒SLC1A3 axis through SUMO1-mediated SUMOylation of IκBα. Our research suggests that SENP5 could serve as a prognostic indicator and a target for therapeutic intervention for ESCC patients.
Collapse
Affiliation(s)
- Chaoxiang Du
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Department of Thoracic Surgery, Zhongshan Hospital (Xiamen), Fudan University, 361006 Xiamen, Fujian, China
| | - Yunfan Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Xinyu Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Zhe Zhang
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, 214400 Jiangyin, Jiangsu, China
| | - Jianmin Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Tao Zhang
- Department of Thoracic Surgery, Zhongshan Hospital (Xiamen), Fudan University, 361006 Xiamen, Fujian, China
| | - Renfeng Wang
- Department of Thoracic Surgery, Zhongshan Hospital (Xiamen), Fudan University, 361006 Xiamen, Fujian, China
| | - Shaoyuan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, 214400 Jiangyin, Jiangsu, China
| |
Collapse
|
24
|
Zhang C, Chen L, Xiu Y, Zhang H, Zhang Y, Ying W. Burden of esophageal cancer in global, regional and national regions from 1990 to 2021 and its projection until 2050: results from the GBD study 2021. Front Oncol 2025; 14:1518567. [PMID: 39902130 PMCID: PMC11788179 DOI: 10.3389/fonc.2024.1518567] [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: 10/28/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025] Open
Abstract
Background Esophageal cancer (EC) is a major global health issue characterized by high morbidity and mortality rates, with a notably low five-year survival rate. Comprehensive analyses of the global burden of EC remain limited and outdated, despite its global significance. This study aimed to systematically assess the global burden and trends of esophageal cancer across diverse populations. Methods Data on the burden of EC were collected from the Global Burden of Disease (GBD) 2021 study, including estimates of incidence, mortality, and disability-adjusted life years (DALYs), as well as risk factors, spanning 204 countries and territories. Age-standardized rates (ASRs) were calculated to allow comparisons across populations. The study further explored the relationship between EC burden and socioeconomic development by utilizing the Socio-demographic Index (SDI), aggregating data by regions. The Bayesian age-period-cohort model was applied to project future trends until 2050. Results In 2021, there were 576,529 new esophageal cancer cases, with an age-standardized incidence rate (ASIR) of 6.65 per 100,000, reflecting a 24.87% decrease since 1990. The global number of deaths reached 538,602, with an age-standardized death rate (ASDR) of 6.25 per 100,000, representing a 30.67% decline. DALYs totaled 12,999,264, corresponding to an estimated annual percentage change (EAPC) of a 1.73% decrease in the age-standardized DALYs rate. East Asia accounted for nearly two-thirds of global EC cases and deaths, while Central Sub-Saharan Africa recorded the highest ASIR and ASDR. Central Asia experienced the largest reductions, whereas Western Sub-Saharan Africa showed increasing trends. Middle-SDI countries, such as Malawi and Lesotho, had disproportionately high burdens, while high-SDI countries, including Tunisia and Kuwait, had lower burdens. Males had higher incidence and mortality rates across all age groups. By 2050, the ASIR is projected to decrease to 6.17 per 100,000, and the ASDR to 5.23 per 100,000, though the absolute number of cases and deaths is expected to rise. Conclusions The global burden of EC remains significant, with ongoing challenges in regions such as Africa and East Asia. These findings highlight the need for sustained and targeted prevention efforts, particularly in high-risk populations, to address the increasing absolute number of cases and deaths.
Collapse
Affiliation(s)
- Chengcheng Zhang
- Institute of Nursing Research, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Linzhi Chen
- Department of Nursing, Shantou University Medical College, Shantou, Guangdong, China
| | - Yuqi Xiu
- Department of Nursing, Shantou University Medical College, Shantou, Guangdong, China
| | - Hongling Zhang
- Department of Nursing, Shantou University Medical College, Shantou, Guangdong, China
| | - Yuejuan Zhang
- Nursing Research Office, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wenjuan Ying
- Department of Nursing, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
25
|
Noh JH, Park H, Kim DH, Na HK, Ahn JY, Lee JH, Jung KW, Choi KD, Song HJ, Lee GH, Jung HY. Sex Differences in Clinical Features and Survival Outcomes of Esophageal Cancer: A Comparative Study in the Korean Population. World J Mens Health 2025; 43:43.e3. [PMID: 39843176 DOI: 10.5534/wjmh.240192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/14/2024] [Accepted: 11/03/2024] [Indexed: 01/24/2025] Open
Abstract
PURPOSE Esophageal cancer is a predominantly male disease. However, the sex differences associated with esophageal cancer have not been thoroughly investigated. This study aimed to evaluate the differences between esophageal cancer in males and females in the Korean population. MATERIALS AND METHODS We assessed patients diagnosed with esophageal cancer between 2005 and 2015 at a tertiary referral center. The clinical features of patients, histopathologic characteristics of tumors, and treatment and survival outcomes were compared between male and female patients. RESULTS We enrolled 2,068 patients, comprising 1,924 (93.0%) males and 144 (7.0%) females. The median age at diagnosis was younger for females than males (65 vs. 63 years, p=0.004). Squamous cell carcinoma was the predominant pathological type (99.0% in males and 93.1% in females); however, the proportion of adenocarcinoma cases was higher in females than males (0.8% vs. 5.6%, p<0.001). Multivariate analysis indicated favorable overall survival for female patients (hazard ratio [HR], 0.685; 95% confidence interval [CI], 0.548-0.857) and patients with high body mass index (≥25 kg/m², HR, 0.432; 95% CI, 0.355-0.526), and in early tumor stage (Stage 4, HR, 12.684; 95% CI, 7.451-21.591). The 5-year overall survival (44.8% vs. 53.5%, p=0.016) and recurrence-free survival rates (74.0% vs. 84.3%, p=0.036) were higher in females than in males. CONCLUSIONS We found significant sex differences in esophageal cancer among the Korean population, with female patients demonstrating distinct clinical characteristics and more favorable survival outcomes compared to male patients. These findings underscore the importance of considering sex-specific factors in the management and prognosis of esophageal cancer.
Collapse
Affiliation(s)
- Jin Hee Noh
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Hyungchul Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Do Hoon Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Hee Kyong Na
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji Yong Ahn
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong Hoon Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kee Wook Jung
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kee Don Choi
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho June Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gin Hyug Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hwoon-Yong Jung
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Sun Z, Wang X. Absence of genetic association between insulin-like growth factors and esophageal cancer. Medicine (Baltimore) 2024; 103:e40899. [PMID: 39969361 PMCID: PMC11688069 DOI: 10.1097/md.0000000000040899] [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: 09/11/2024] [Accepted: 11/22/2024] [Indexed: 02/20/2025] Open
Abstract
This study aimed to explore the causal relationship between concentrations of various insulin-like growth factors (IGFs) and IGF-binding proteins (IGFBPs) and esophageal cancer (ESC), addressing the gap in understanding the genetic link between IGF1 and ESC. A two-sample Mendelian randomization (MR) analysis was conducted using single nucleotide polymorphisms linked to IGFs/IGFBPs and ESC from the IEU Open GWAS Project. This analysis included ESC GWAS data from 1996 individuals of European descent and genetic variant data from 3310 individuals of European ancestry. Various methods, such as inverse variance weighting, weighted median, weighted mode, and MR-Egger regression, were applied for analysis, with sensitivity assessments including MR-PRESSO, Cochran Q, and leave-one-out analysis to ensure the robustness of results and detect biases. The genetic predictions indicated no significant association between IGFs/IGFBPs and ESC. When ESC was the outcome measure, the odds ratios with 95% confidence intervals were as follows: IGF1 = 1.00 (0.89-1.12, P = .936), IGF1R = 1.07 (0.90-1.27, P = .453), IGFBP3 = 1.00 (0.79-1.26, P = .975), and IGFBPL1 = 0.91 (0.75-1.12, P = .372). MR-Egger regression confirmed the absence of horizontal pleiotropy, and no outliers were identified by MR-PRESSO. Leave-one-out analysis supported the stability of the results. The study did not find a causal connection between IGFs/IGFBPs and ESC. These results suggest the need for further validation and potentially highlight the complex interplay of factors involved in the development of ESC.
Collapse
Affiliation(s)
- Zhengliang Sun
- Department of Thoracic Surgery, East Hospital of Tongji University, Pudong District, Shanghai, China
| | - Xiaohong Wang
- Longhua Street Community Health Center, Xuhui District, Shanghai, China
| |
Collapse
|
27
|
Hu B, Yin G, Zhu J, Bai Y, Sun X. Continuous prediction for tumor mutation burden based on transcriptional data in gastrointestinal cancers. BMC Med Inform Decis Mak 2024; 24:384. [PMID: 39695561 DOI: 10.1186/s12911-024-02794-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Tumor mutation burden (TMB) has been considered a biomarker for utilization of immune checkpoint inhibitors(ICIs), but whole exome sequencing(WES) and cancer gene panel(CGP) based on next generation sequencing for TMB detection are costly. Here, we use transcriptome data of TCGA to construct a model for TMB prediction in gastrointestinal tumors. METHODS Transcriptome data, somatic mutation data and clinical data of four gastrointestinal tumors from TCGA, including esophageal cancer (ESCA), stomach adenocarcinoma (STAD), colon adenocarcinoma (COAD) and rectal adenocarcinoma (READ). Using R, we performed visual analysis of somatic mutation data, differentially expressed genes (DEGs) function enrichment analysis, gene set enrichment analysis (GSEA), and estimated TMB value in clinic. Finally, a deep neural network (DNN) model was constructed for TMB prediction. RESULTS Visualization of somatic mutation data summarized the classification of mutation, frequency of each mutation type, and top-mutated genes. GSEA showed the enrichment of CD4+/CD8+ T cells in the high TMB group and the activation of tumor suppressing pathways. Single-sample GSEA (ssGSEA) manifested that the high-TMB group had higher level of multiple immune cells infiltration. In addition, distribution of TMB was related to clinical parameters. Like age, M stage, N stage, AJCC stage, and overall survival(OS). After model optimization using genetic algorithm, in the training set, validation set, and testing set, the Pearson relevance coefficient r between predicted values and actual values reaches 0.98, 0.82, and 0.92, respectively; the coefficient of determination R2 is 0.95, 0.82, and 0.7, respectively. CONCLUSION TMB correlates with clinicopathological parameters in gastrointestinal carcinoma, and patients with high TMB have higher levels of immune infiltration. In addition, the DNN model based on 31 genes predicts TMB of gastrointestinal tumors in a high accuracy.
Collapse
Affiliation(s)
- Beibei Hu
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guohui Yin
- Key Laboratory of Traffic Safety On Track (Central South University), Ministry of Education, School of Traffic and Transportation Engineering, Central South University, Changsha, 410075, China
| | - Jialin Zhu
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yi Bai
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuren Sun
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
28
|
Gambella A, Fiocca R, Lugaresi M, D’Errico A, Malvi D, Spaggiari P, Tomezzoli A, Albarello L, Ristimäki A, Bottiglieri L, Bonora E, Krishnadath KK, Raulli GD, Rosati R, Romario UF, De Manzoni G, Räsänen J, Mattioli S, Grillo F, Mastracci L. Pre-Surgical Endoscopic Biopsies Are Representative of Esophageal and Esophago-Gastric Junction Adenocarcinoma Histologic Classes and Survival Risk. Cancers (Basel) 2024; 16:4045. [PMID: 39682231 PMCID: PMC11640587 DOI: 10.3390/cancers16234045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Background and Objectives: The Esophageal Adenocarcinoma Study Group Europe (EACSGE) recently proposed a granular histologic classification of esophageal-esophago-gastric junctional adenocarcinomas (EA-EGJAs) based on the study of naïve surgically resected specimens that, when combined with the pTNM stage, is an efficient indicator of prognosis, molecular events, and response to treatment. In this study, we compared histologic classes of endoscopic biopsies taken before surgical resection with those of the surgical specimen, to evaluate the potential of the EACSGE classification at the initial diagnostic workup. Methods: A total of 106 EA-EGJA cases with available endoscopic biopsies and matched surgical resection specimens were retrieved from five Italian institutions. Histologic classification was performed on all specimens to identify well-differentiated glandular adenocarcinoma (WD-GAC), poorly differentiated glandular adenocarcinoma (PD-GAC), mucinous muconodular carcinoma (MMC), infiltrative mucinous carcinoma (IMC), diffuse desmoplastic carcinoma, diffuse anaplastic carcinoma (DAC), and mixed subtypes. Related risk subgroups (low-risk versus high-risk) were also assessed. The correlations of histologic classes and risk subgroups between diagnostic biopsies and surgical resection specimens were explored with Spearman's correlation test. Sensitivity, specificity, accuracy, positive predictive value, negative predictive value, true positives, true negatives, false positives, and false negatives were also calculated. Results: A strong positive correlation between biopsies and surgical specimens occurred for both histologic classes (coefficient: 0.75, p < 0.001) and risk subgroups (coefficient: 0.65, p < 0.001). The highest sensitivities and specificities were observed for MMC, IMC, and DAC (100% and 99% for all), followed by WD-GAC (sensitivity 91%, specificity 79%) and PD-GAC (sensitivity 722%, specificity 86%). The low-risk and high-risk groups presented a sensitivity and specificity of 89% and 76% (low-risk) and 76% and 89% (high-risk). Conclusions: The EACSGE histologic classification of EA-EGJAs and associated prognostic subgroups can be reliably assessed on pre-operative diagnostic biopsies. Further studies on larger and more representative cohorts of EA-EGJAs will allow us to validate our findings and confirm if the EA-EGJA biopsy histomorphology and clinical TNM staging will be as efficient as the surgical specimen histomorphology and pTNM in predicting patient prognoses and tailoring personalized therapeutic approaches.
Collapse
Affiliation(s)
- Alessandro Gambella
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy; (R.F.); (F.G.); (L.M.)
| | - Roberto Fiocca
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy; (R.F.); (F.G.); (L.M.)
| | - Marialuisa Lugaresi
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (M.L.); (E.B.); (S.M.)
- Division of Thoracic Surgery, Maria Cecilia Hospital, GVM Care & Research Group, Cotignola, 48022 Ravenna, Italy
| | - Antonietta D’Errico
- Pathology Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (A.D.); (D.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Deborah Malvi
- Pathology Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (A.D.); (D.M.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Paola Spaggiari
- Unit of Anatomic Pathology, Humanitas University, 20089 Milan, Italy;
| | - Anna Tomezzoli
- Unit of Anatomic Pathology, Azienda Ospedaliera di Verona, 37122 Verona, Italy;
| | - Luca Albarello
- Pathology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Ari Ristimäki
- Department of Pathology, HUSLAB and HUS Diagnostic Center, University of Helsinki, 00170 Helsinki, Finland;
- Helsinki University Hospital, 00170 Helsinki, Finland
| | - Luca Bottiglieri
- Unit of Anatomic Pathology, Istituto Europeo di Oncologia, 20122 Milan, Italy;
| | - Elena Bonora
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (M.L.); (E.B.); (S.M.)
- IRCCS Azienda Ospedaliero Universitaria di Bologna, 40126 Bologna, Italy
| | - Kausilia K. Krishnadath
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Department of Gastroenterology and Hepatology, University Hospital Antwerp, 2650 Antwerp, Belgium;
| | | | - Riccardo Rosati
- Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, Vita Salute San Raffaele University, 20132 Milan, Italy;
| | | | - Giovanni De Manzoni
- Department of Surgery, General and Upper G.I. Surgery Division, University of Verona, 37126 Verona, Italy;
| | - Jari Räsänen
- Department of General Thoracic and Esophageal Surgery, Helsinki University Hospital, Helsinki University, 00170 Helsinki, Finland;
| | - Sandro Mattioli
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (M.L.); (E.B.); (S.M.)
- Division of Thoracic Surgery, Maria Cecilia Hospital, GVM Care & Research Group, Cotignola, 48022 Ravenna, Italy
| | - Federica Grillo
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy; (R.F.); (F.G.); (L.M.)
- IRCCS San Martino Policlinic Hospital of Genoa, 16132 Genoa, Italy
| | - Luca Mastracci
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy; (R.F.); (F.G.); (L.M.)
- IRCCS San Martino Policlinic Hospital of Genoa, 16132 Genoa, Italy
| |
Collapse
|
29
|
Aoki T, Watson DI, Bulamu NB. Cost-effective identification of Barrett's esophagus in the community: A first step towards screening. J Gastroenterol Hepatol 2024; 39:2654-2663. [PMID: 39385742 PMCID: PMC11660199 DOI: 10.1111/jgh.16762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND AND AIM The first step towards developing a screening strategy for Barrett's esophagus (BE) is the identification of individuals in the community. Currently available tools include endoscopy, less-invasive non-endoscopic devices, and non-invasive risk stratification models. We evaluated the cost of potential strategies for identification of BE as a first step towards screening. METHODS Two hypothetical cohorts of the general population aged ≥ 50 years with BE prevalence rates of 1.9% and 6.8% were modeled. Four potential screening tools were evaluated: (i) risk stratification based on non-weighted clinical factors according to US/European guidelines, (ii) weighted risk stratification using algorithmic models, (iii) less-invasive devices such as Cytosponge + trefoil factor 3 (TFF3), and (iv) endoscopy. Using a decision-analytic model, the cost per BE case identified and the cost-effectiveness were compared for six potential BE screening strategies based on combinations of the four screening tools; (i) + (iv), (ii) + (iv), (iii) + (iv), (i) + (iii) + (iv), (ii) + (iii) + (iv), and only (iv). RESULTS The cost per BE case identified was lowest for the weighted risk stratification followed by Cytosponge-TFF3 then endoscopy strategy at both 1.9% and 6.8% BE prevalences (US$9282 and US$3406, respectively) although it was sensitive to the cost of less-invasive devices. This strategy was also most cost-effective for a BE prevalence of 1.9%. At BE prevalence of 6.8%, the Cytosponge-TFF3 followed by endoscopy strategy was most cost-effective. CONCLUSIONS Incorporating weighted risk stratification and less-invasive devices such as Cytosponge-TFF3 into BE screening strategies has a potential to cost-effectively identify BE in the community although device cost and the community prevalence of BE will impact the optimal strategy.
Collapse
Affiliation(s)
- Tomonori Aoki
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - David I. Watson
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
- Department of SurgeryFlinders Medical CentreAdelaideSouth AustraliaAustralia
| | - Norma B. Bulamu
- Flinders Health and Medical Research Institute, College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| |
Collapse
|
30
|
Ghasemi M, Bahrami Koutenaei M, Ghasemi A, Alizadeh-Navaei R, Moosazadeh M. A systematic review and dose‒response meta-analysis of the association between nitrate & nitrite intake and gastroesophageal cancer risk. Nitric Oxide 2024; 153:61-71. [PMID: 39401565 DOI: 10.1016/j.niox.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/07/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
OBJECTIVE The objective of this systematic review and dose‒response meta-analysis was to assess the associations between the dietary consumption of nitrate and nitrite and the risk of gastric and esophageal cancer. METHODS MEDLINE, Scopus, Embase, Web of Science, Proquest, and Google Scholar were searched until April 1, 2024. Articles were selected by two independent researchers on the basis of the inclusion and exclusion criteria. Data regarding the study design, type of exposure and outcomes, intervals of intake of nitrate or nitrite in each layer, OR/RR/HR of the relationship for each layer of intake, total sample size, and number of cases of gastric or esophageal cancer were extracted. The certainty of the evidence was rated via the GRADE method. The pooled odds ratios, risk ratios, and dose‒response analyses were calculated via Stata version 17.0. The best-fit dose‒response model was assessed by the P value for linearity and nonlinearity. Study heterogeneity was assessed via the I2 and Q tests. RESULTS We found 2124 nonredundant studies, 234 of which were potentially relevant. Eighteen articles met the inclusion criteria and were included in the review. The results of the meta-analysis revealed a significant positive association between nitrite intake and gastric cancer in both case‒control studies (OR = 1.29, 95 % CI = 1.09-1.52, P value = 0.001, I2 = 1.91 %) and cohort studies (RR = 1.17, 95 % CI = 1.00-1.37, P value = 0.04, I2 = 0.00 %). In addition, case‒control studies revealed a nonsignificant inverse association between nitrate intake and gastric cancer incidence (OR = 0.71, 95 % CI = 0.50-1.01, P value = 0.06, I2 = 74.89 %), and cohort studies (RR = 0.89, 95 % CI = 0.73-1.09, P value = 0.27, I2 = 0.00 %). Case‒control studies also revealed no significant correlation between nitrite intake and esophageal cancer incidence (OR = 1.48, 95 % CI = 0.91 to 2.42, P value = 0.12, I2 = 0.001 %). Nitrites correlated linearly with gastric cancer (linearity P value = 0.001). The most appropriate fit models for the relationship between nitrate and gastric cancer were both piecewise linear and natural polynomial regression (quadratic) models (P values = 0.003 and 0.005, respectively). There was no significant publication bias. CONCLUSION According to this meta-analysis, high consumption of nitrites was associated with an increased risk of gastric cancer in case‒control and cohort studies with a linear regression model, and dietary nitrate intake was not associated with the risk of gastric cancer in either case‒control or cohort studies. These findings are inconclusive and require confirmation in future prospective studies with robust methodologies and adjustments for potential confounders.
Collapse
Affiliation(s)
| | - Mohammad Bahrami Koutenaei
- Gastrointestinal Cancer Research Center, Noncommunicable Disease Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Ghasemi
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Reza Alizadeh-Navaei
- Gastrointestinal Cancer Research Center, Noncommunicable Disease Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahmood Moosazadeh
- Gastrointestinal Cancer Research Center, Noncommunicable Disease Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
31
|
Huang G, Xu J, Li Y, Song L, Wen C, Ruan Q, Wen Z, Qi J, Deng J, Liu Y. Corynoxine exerts the anti-tumor effect on esophageal squamous cell carcinoma principally via the EZH2-DUSP5-ERK1/2-mediated cell growth inhibition. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156103. [PMID: 39383633 DOI: 10.1016/j.phymed.2024.156103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Esophageal cancer is one of the most prevalent malignant tumors and the sixth largest cause of tumor-associated death worldwide. Squamous cell carcinoma (ESCC) accounts for 85 % of all esophageal cancer cases. ESCC treatment remains to be significantly difficult. Corynoxine (Cory) is a tetracyclic hydroxyindole alkaloid isolated from Uncaria macrophylla. It is unclear whether Cory has an anti-tumor effect on ESCC. PURPOSE To determine the anti-tumor activity of Cory and the associated mechanisms in ESCC. STUDY DESIGN Cory's effects on proliferation, apoptosis, migration, and invasion, as well as the underlying molecular causes were assessed using two ESCC cell lines, KYSE150 and TE-1. A xenograft mouse model was then applied to evaluate the anti-tumor activity of Cory in vivo. METHODS Western blot, assays including CCK-8, colony formation, EdU staining, TUNEL staining, cell scratch and Transwell, and a xenograft mouse model were used in this study. RESULTS Cory suppressed cell growth, provoked cell apoptosis, and hindered cell migration and invasion of ESCC cells. DUSP5 knockdown reduced the Cory-induced cell death and restored cell migration and invasion through ERK1/2 activation. Further analyses showed that Cory promoted DUSP5 expression via inhibiting EZH2 expression, leading to inactivation of ERK1/2 signaling and the subsequent cell growth inhibition of ESCC. In vivo experiments disclosed that Cory suppressed tumor growth of ESCC through upregulating DUSP5 expression. CONCLUSIONS Cory plays an anti-tumor role in ESCC by regulating EZH2-DUSP5-ERK1/2 signaling pathway. Cory may be promising to be a novel therapy for treating ESCC.
Collapse
Affiliation(s)
- Gang Huang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; The People's Hospital of Beilun District, Ningbo 315000, China
| | - Jiale Xu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yingchao Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Liangtao Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chunmei Wen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Qingqing Ruan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Zhikai Wen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035 China
| | - Jinxia Qi
- Biobank, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jie Deng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Yu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
32
|
Yu Z, Chen T, Peng H, Li A, Wei Y, Xiao S. Trends in incidence, treatment modalities and prognosis of esophageal adenocarcinoma in the US population. Cancer Epidemiol 2024; 93:102683. [PMID: 39366329 DOI: 10.1016/j.canep.2024.102683] [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: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) was the predominant subtype of esophageal cancer in the Western population. However, an updated and comprehensive analysis of epidemiologic, clinical, and prognostic characteristics of esophageal adenocarcinoma is lacking. MATERIALS AND METHODS This was a population-based cohort study using the Surveillance Epidemiology and End-Results (SEER) Database. Patients diagnosed with EAC between 1988 and 2020 were included. Incidence trends, clinical characteristics, treatment patterns, and relative survival were systematically analyzed. RESULTS The overall age-standardized incidence rate of EAC significantly increased from 1.7 per 100000 persons in 1988 to 3.6 per 100000 persons in 2020. There were no significant changes in the distribution of age group, sex, and primary site of EAC over time. However, the proportion of EAC clinically staged as I or II decreased from 35.1 % to 27.9 %. Over time, palliative chemotherapy in metastatic EAC increased from 26.7 % to 41.3 %, combination therapy was still the main treatment strategy for nonmetastatic EAC. Despite the 5-year survival rate was less than 20 %, 1-year survival has experienced a moderate increase from 46.7 % to 53.7 %. Specifically, 1-year survival rate for nonmetastatic EAC undergoing surgery only experienced a significant increase from 80.2 % in 2004-2006 to 94.7 % in 2019-2020. For metastatic EAC, obvious improvement in 1-year survival rate was observed in those treated with systematic therapy (from 26.6 % in 2004-2006 to 41.2 % in 2019-2020). In the multivariable analysis, older age, male sex, lower household income, living without a partner, advanced TNM stage, and receiving no cancer treatment were significantly associated with poor survival. CONCLUSION In summary, this population-based study of EAC patients in the US showed an increase in incidence, a shift in treatment modalities for metastatic EAC, and moderately improved 1-year survival. The search for more effective surveillance and treatment strategies should be continued in the future.
Collapse
Affiliation(s)
- Zhuoyang Yu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tong Chen
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Haoyu Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Anyuan Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yutong Wei
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shiyu Xiao
- Department of Gastroenterology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
33
|
Wang C, Shi ZZ. Exosomes in esophageal cancer: function and therapeutic prospects. Med Oncol 2024; 42:18. [PMID: 39601925 DOI: 10.1007/s12032-024-02543-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024]
Abstract
Esophageal cancer (EC) is one of the most common malignant tumors worldwide. Exosomes are a type of extracellular vesicles produced by eukaryotic cells and present in all body fluids. Recent studies have revealed that exosomes can be used as a tool for cell signaling and have great potential in cancer diagnosis and treatment strategies. This article reviews the research progress of exosomes in EC in recent years, mainly including the mechanism of action, diagnostic markers, therapeutic targets, and drug carriers. The challenges faced are discussed to provide guidelines for further research in future.
Collapse
Affiliation(s)
- Chong Wang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Zhi-Zhou Shi
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
34
|
Shaheen NJ, Othman MO, Taunk J, Chang KJ, Jaganmohan S, Yachimski PS, Fang JC, Spataro JS, Verma S, Lee VT, deGuzman BJ, Aklog L. Use of a Two-Gene Methylated DNA Biomarker Assay and Nonendoscopic Balloon for Detection of Barrett Esophagus Among High-Risk Individuals in a Screening Population. Am J Gastroenterol 2024:00000434-990000000-01475. [PMID: 39588974 DOI: 10.14309/ajg.0000000000003238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 11/01/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Barrett esophagus (BE) is the precursor to esophageal adenocarcinoma (EAC). We aimed to assess performance, safety, and tolerability of the EsoGuard (EG) assay on samples collected nonendoscopically with the EsoCheck (EC) device (EG/EC) for BE detection in the intended-use population meeting American College of Gastroenterology guideline criteria (chronic gastroesophageal reflux disease and 3+ additional risk factors). METHODS We performed a prospective, multicenter study (NCT04293458) to assess EG performance (primary endpoint) on cells collected with EC, for detection of BE and EAC using esophagogastroduodenoscopy (EGD) and biopsies as the comparator. Twenty-four sites across the United States and Spain participated. EC safety and usability were assessed as secondary endpoints. RESULTS 180 male subjects aged >50 years with chronic gastroesophageal reflux disease met eligibility criteria, of which 163 (90.6%) had EGD and successful EC administration. Mean age was 60.5 years, 34.4% were obese, 56.7% had tobacco history, and 3.9% had a 1st degree relative with BE or EAC. Of 122 samples analyzed, 93 contributed to the primary endpoint analysis. Eight subjects (8.6%) in the Primary Analysis Population had BE on EGD, none with dysplasia. Sensitivity of EG for BE was 87.5% (95% confidence interval [CI] 47.4-99.7), specificity was 81.2% (95% CI 71.2-88.8), positive predictive value was 30.4% (95% CI 13.2-52.9), and negative predictive value was 98.6% (95% CI 92.3-99.96). Mild esophageal abrasions were observed in 1.5%; no serious adverse events were reported. DISCUSSION This study in the intended-use population suggests that EG/EC is promising for BE screening. While future work is necessary to define its performance characteristics with more precision, this approach may provide a safe, accurate, and well-tolerated nonendoscopic alternative in high-risk patients.
Collapse
Affiliation(s)
- Nicholas J Shaheen
- Division of Gastroenterology and Hepatology, Center for Esophageal Diseases and Swallowing, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mohamed O Othman
- Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, Texas, USA
| | - Jawar Taunk
- Advanced Gastroenterology Associates, Palm Harbor, Florida, USA
| | - Kenneth J Chang
- Irvine Medical Center, University of California, Orange, California, USA
| | | | - Patrick S Yachimski
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John C Fang
- Division of Gastroenterology, Hepatology and Nutrition, University of Utah, Salt Lake City, Utah, USA
| | | | - Suman Verma
- Lucid Diagnostics Inc., New York, New York, USA
| | | | | | | |
Collapse
|
35
|
Yang H, Wang F, Hallemeier CL, Lerut T, Fu J. Oesophageal cancer. Lancet 2024; 404:1991-2005. [PMID: 39550174 DOI: 10.1016/s0140-6736(24)02226-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/16/2024] [Accepted: 10/07/2024] [Indexed: 11/18/2024]
Abstract
Oesophageal cancer is the seventh leading cause of cancer mortality worldwide. Two major pathological subtypes exist: oesophageal squamous cell carcinoma and oesophageal adenocarcinoma. Epidemiological studies in the last decade have shown a gradual increase in the incidence of oesophageal adenocarcinoma worldwide. The prognosis of oesophageal cancer has greatly improved due to breakthroughs in screening, surgical procedures, and novel treatment modalities. The success achieved with combined modality therapies, including surgery, chemotherapy, and radiotherapy, to treat locally advanced oesophageal cancer is particularly notable. Immunotherapy has become a crucial treatment for oesophageal cancer, with immune checkpoint inhibitor-based therapies now established as the standard of care in adjuvant and metastatic first-line settings. This Seminar provides an overview of advances in the screening, diagnosis, and treatment of oesophageal squamous cell carcinoma and oesophageal adenocarcinoma, with a particular focus on neoadjuvant therapies for locally advanced oesophageal cancer and immune checkpoint inhibitor-based therapies.
Collapse
Affiliation(s)
- Hong Yang
- Department of Thoracic Surgery, Sun Ya University Cancer Center, Guangzhou, China; Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China; Guangdong Esophageal Cancer Institute, Guangzhou, China
| | - Feng Wang
- Department of Medical Oncology, Sun Ya University Cancer Center, Guangzhou, China; Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
| | | | - Toni Lerut
- Department of Thoracic Surgery, University Hospital Leuven, Leuven, Belgium
| | - Jianhua Fu
- Department of Thoracic Surgery, Sun Ya University Cancer Center, Guangzhou, China; Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China; Guangdong Esophageal Cancer Institute, Guangzhou, China.
| |
Collapse
|
36
|
Zhang P, Zhang W, Hong Z, Jiang J, Wu N, Lin J, Kang M. Elucidating the role of CYFIP2 in conferring cisplatin resistance in esophageal squamous cell carcinoma. Sci Rep 2024; 14:27130. [PMID: 39511293 PMCID: PMC11544133 DOI: 10.1038/s41598-024-77420-8] [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: 01/24/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
Cisplatin (CDDP) serves as a vital component in the chemotherapeutic approach to treat esophageal squamous cell carcinoma (ESCC). However, prolonged CDDP application frequently culminates in resistance, compromising therapeutic outcomes. Through genome-wide CRISPR library screening, our study elucidates the mechanisms underlying this resistance, pinpointing CYFIP2 as a pivotal mediator. Notably, the involvement CYFIP2 is characterized by pronounced autophagic activity and the modulation of multiple cellular pathways. Empirical validation was achieved by treating ESCC cell lines with CDDP, which resulted in an upsurge of CYFIP2 expression. The functional impact of CYFIP2 was further delineated through knockdown experiments, where a marked suppression in cell proliferation was observed, alongside a discernible decline in reactive oxygen species levels. This was complemented by a suite of assays and microscopic techniques, including GFP-LC3, mRFP-GFP-LC3, electron microscopy and western blot, which collectively affirmed the inhibitory effect of CYFIP2 knockdown on autophagic processes, particularly impeding autophagosome formation and their subsequent fusion with lysosomes. In vivo studies have also confirmed that CYFIP2 knockdown limits tumor progression and increases CDDP efficacy. Conclusively, our findings introduce CYFIP2 as a novel contributor to CDDP resistance in ESCC, underscoring its potential as a therapeutic target. This revelation not only deepens our understanding of resistance mechanisms but also paves the way for novel oncotherapeutic strategies, promising enhanced treatment efficacy against ESCC.
Collapse
Affiliation(s)
- Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Weiguang Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Zhinuan Hong
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Junfei Jiang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Ningzi Wu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China
| | - Jihong Lin
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China.
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China.
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
- Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China.
| |
Collapse
|
37
|
Zheng H, Huang L, An G, Guo L, Wang N, Yang W, Zhu Y. A Nanoreactor Based on Metal-Organic Frameworks With Triple Synergistic Therapy for Hepatocellular Carcinoma. Adv Healthc Mater 2024; 13:e2401743. [PMID: 39015058 DOI: 10.1002/adhm.202401743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/17/2024] [Indexed: 07/18/2024]
Abstract
The transformation of monotherapy into multimodal combined targeted therapy to fully exploit synergistic efficacy is of increasing interest in tumor treatment. In this work, a novel nanodrug-carrying platform based on iron-based MOFs, which is loaded with doxorubicin hydrochloride (DOX), dihydroartemisinin (DHA), and glucose oxidase (GOx), and concurrently covalently linked to the photosensitizer 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) in polydopamine (PDA)-encapsulated MIL-101(Fe) (denoted as MIL-101(Fe)-DOX-DHA@TCPP/GOx@PDA, MDDTG@P), is successfully developed. Upon entering the tumor microenvironment, MDDTG@P catalyzes the hydrogen peroxide (H2O2) into hydroxyl radicals (·OH) and depletes glutathione (GSH); thus, exerting the role of chemodynamic therapy (CDT). The reduced Fe2+ can also activate DHA, further expanding CDT and promoting tumor cell apoptosis. The introduced GOx will rapidly consume glucose and oxygen (O2) in the tumor; while, replenishing H2O2 for Fenton reaction, starving the cancer cells; and thus, realizing starvation and chemodynamic therapy. In addition, the covalent linkage of TCPP endows MDDTG@P with good photodynamic therapeutic (PDT) properties. Therefore, this study develops a nanocarrier platform for triple synergistic chemodynamic/photodynamic/starvation therapy, which has promising applications in the efficient treatment of tumors.
Collapse
Affiliation(s)
- Heming Zheng
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Lei Huang
- School of Stomatology, Minzhu Clinic of Stomatology Hospital Affiliated to Guangxi Medical University, Guangxi, 530007, China
| | - Guanghui An
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Lianshan Guo
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Wenhui Yang
- Department of Medical Laboratory, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QF, UK
| |
Collapse
|
38
|
Chen Y, Zheng Z, Wang L, Chen R, He M, Zhao X, Jin L, Yao J. Deciphering STAT3's negative regulation of LHPP in ESCC progression through single-cell transcriptomics analysis. Mol Med 2024; 30:192. [PMID: 39468431 PMCID: PMC11520558 DOI: 10.1186/s10020-024-00962-0] [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: 06/15/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Esophageal Squamous Cell Carcinoma (ESCC) remains a predominant health concern in the world, characterized by high prevalence and mortality rates. Advances in single-cell transcriptomics have revolutionized cancer research by enabling a precise dissection of cellular and molecular diversity within tumors. OBJECTIVE This study aims to elucidate the cellular dynamics and molecular mechanisms in ESCC, focusing on the transcriptional influence of STAT3 (Signal Transducer and Activator of Transcription 3) and its interaction with LHPP, thereby uncovering potential therapeutic targets. METHODS Single-cell RNA sequencing was employed to analyze 44,206 cells from tumor and adjacent normal tissues of ESCC patients, identifying distinct cell types and their transcriptional shifts. We conducted differential gene expression analysis to assess changes within the tumor microenvironment (TME). Validation of key regulatory interactions was performed using qPCR in a cohort of 21 ESCC patients and further substantiated through experimental assays in ESCC cell lines. RESULTS The study revealed critical alterations in cell composition and gene expression across identified cell populations, with a notable shift towards pro-tumorigenic states. A significant regulatory influence of STAT3 on LHPP was discovered, establishing a novel aspect of ESCC pathogenesis. Elevated levels of STAT3 and suppressed LHPP expression were validated in clinical samples. Functional assays confirmed that STAT3 directly represses LHPP at the promoter level, and disruption of this interaction by promoter mutations diminished STAT3's repressive effect. CONCLUSION This investigation underscores the central role of STAT3 as a regulator in ESCC, directly impacting LHPP expression and suggesting a regulatory loop crucial for tumor behavior. The insights gained from our comprehensive cellular and molecular analysis offer a deeper understanding of the dynamics within the ESCC microenvironment. These findings pave the way for targeted therapeutic interventions focusing on the STAT3-LHPP axis, providing a strategic approach to improve ESCC management and prognosis.
Collapse
Affiliation(s)
- Yurao Chen
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Zemao Zheng
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Luoshai Wang
- Department of Thoracic Surgery, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
| | - Ronghuai Chen
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Ming He
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Xiang Zhao
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Liyan Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213000, Jiangsu, China.
- Department of Oncology, The Wujin Clinical college of Xuzhou Medical University, Changzhou, 213000, Jiangsu, China.
| | - Juan Yao
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China.
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China.
| |
Collapse
|
39
|
Wang Y, Xing S, Xu YW, Xu QX, Ji MF, Peng YH, Wu YX, Wu M, Xue N, Zhang B, Xie SH, Zhu RD, Ou XY, Huang Q, Tian BY, Li HL, Jiang Y, Yao XB, Li JP, Ling L, Cao SM, Zhong Q, Liu WL, Zeng MS. Highly sensitive detection platform-based diagnosis of oesophageal squamous cell carcinoma in China: a multicentre, case-control, diagnostic study. Lancet Digit Health 2024; 6:e705-e717. [PMID: 39332854 DOI: 10.1016/s2589-7500(24)00153-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/01/2024] [Accepted: 07/07/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Early detection and screening of oesophageal squamous cell carcinoma rely on upper gastrointestinal endoscopy, which is not feasible for population-wide implementation. Tumour marker-based blood tests offer a potential alternative. However, the sensitivity of current clinical protein detection technologies is inadequate for identifying low-abundance circulating tumour biomarkers, leading to poor discrimination between individuals with and without cancer. We aimed to develop a highly sensitive blood test tool to improve detection of oesophageal squamous cell carcinoma. METHODS We designed a detection platform named SENSORS and validated its effectiveness by comparing its performance in detecting the selected serological biomarkers MMP13 and SCC against ELISA and electrochemiluminescence immunoassay (ECLIA). We then developed a SENSORS-based oesophageal squamous cell carcinoma adjunct diagnostic system (with potential applications in screening and triage under clinical supervision) to classify individuals with oesophageal squamous cell carcinoma and healthy controls in a retrospective study including participants (cohort I) from Sun Yat-sen University Cancer Center (SYSUCC; Guangzhou, China), Henan Cancer Hospital (HNCH; Zhengzhou, China), and Cancer Hospital of Shantou University Medical College (CHSUMC; Shantou, China). The inclusion criteria were age 18 years or older, pathologically confirmed primary oesophageal squamous cell carcinoma, and no cancer treatments before serum sample collection. Participants without oesophageal-related diseases were recruited from the health examination department as the control group. The SENSORS-based diagnostic system is based on a multivariable logistic regression model that uses the detection values of SENSORS as the input and outputs a risk score for the predicted likelihood of oesophageal squamous cell carcinoma. We further evaluated the clinical utility of the system in an independent prospective multicentre study with different participants selected from the same three institutions. Patients with newly diagnosed oesophageal-related diseases without previous cancer treatment were enrolled. The inclusion criteria for healthy controls were no obvious abnormalities in routine blood and tumour marker tests, no oesophageal-associated diseases, and no history of cancer. Finally, we assessed whether classification could be improved by integrating machine-learning algorithms with the system, which combined baseline clinical characteristics, epidemiological risk factors, and serological tumour marker concentrations. Retrospective SYSUCC cohort I (randomly assigned [7:3] to a training set and an internal validation set) and three prospective validation sets (SYSUCC cohort II [internal validation], HNCH cohort II [external validation], and CHSUMC cohort II [external validation]) were used in this step. Six machine-learning algorithms were compared (the least absolute shrinkage and selector operator regression, ridge regression, random forest, logistic regression, support vector machine, and neural network), and the best-performing algorithm was chosen as the final prediction model. Performance of SENSORS and the SENSORS-based diagnostic system was primarily assessed using accuracy, sensitivity, specificity, and area under the receiver operating characteristic curve (AUC). FINDINGS Between Oct 1, 2017, and April 30, 2020, 1051 participants were included in the retrospective study. In the prospective diagnostic study, 924 participants were included from April 2, 2022, to Feb 2, 2023. Compared with ELISA (108·90 pg/mL) and ECLIA (41·79 pg/mL), SENSORS (243·03 fg/mL) showed 448 times and 172 times improvements, respectively. In the three retrospective validation sets, the SENSORS-based diagnostic system achieved AUCs of 0·95 (95% CI 0·90-0·99) in the SYSUCC internal validation set, 0·93 (0·89-0·97) in the HNCH external validation set, and 0·98 (0·97-1·00) in the CHSUMC external validation set, sensitivities of 87·1% (79·3-92·3), 98·6% (94·4-99·8), and 93·5% (88·1-96·7), and specificities of 88·9% (75·2-95·8), 74·6% (61·3-84·6), and 92·1% (81·7-97·0), respectively, successfully distinguishing between patients with oesophageal squamous cell carcinoma and healthy controls. Additionally, in three prospective validation cohorts, it yielded sensitivities of 90·9% (95% CI 86·1-94·2) for SYSUCC, 84·8% (76·1-90·8) for HNCH, and 95·2% (85·6-98·7) for CHSUMC. Of the six machine-learning algorithms compared, the random forest model showed the best performance. A feature selection step identified five features to have the highest performance to predictions (SCC, age, MMP13, CEA, and NSE) and a simplified random forest model using these five features further improved classification, achieving sensitivities of 98·2% (95% CI 93·2-99·7) in the internal validation set from retrospective SYSUCC cohort I, 94·1% (89·9-96·7) in SYSUCC prospective cohort II, 88·6% (80·5-93·7) in HNCH prospective cohort II, and 98·4% (90·2-99·9) in CHSUMC prospective cohort II. INTERPRETATION The SENSORS system facilitates highly sensitive detection of oesophageal squamous cell carcinoma tumour biomarkers, overcoming the limitations of detecting low-abundance circulating proteins, and could substantially improve oesophageal squamous cell carcinoma diagnostics. This method could act as a minimally invasive screening tool, potentially reducing the need for unnecessary endoscopies. FUNDING The National Key R&D Program of China, the National Natural Science Foundation of China, and the Enterprises Joint Fund-Key Program of Guangdong Province. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Yu Wang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shan Xing
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Qing-Xia Xu
- Department of Clinical Laboratory, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, China
| | - Ming-Fang Ji
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ya-Xian Wu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Meng Wu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ning Xue
- Department of Clinical Laboratory, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, China
| | - Biao Zhang
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Shang-Hang Xie
- Department of Cancer Prevention Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rui-Dan Zhu
- The Second Clinical Faculty of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xin-Yuan Ou
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qi Huang
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bo-Yu Tian
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui-Lan Li
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yu Jiang
- Department of Clinical Laboratory, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, China
| | - Xiao-Bin Yao
- Department of Clinical Laboratory, Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, China
| | - Jian-Pei Li
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Ling
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Su-Mei Cao
- Department of Cancer Prevention Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qian Zhong
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wan-Li Liu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mu-Sheng Zeng
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
40
|
Zhu J, Qiu X, Jin X, Nie X, Ou S, Wu G, Shen J, Zhang R. ZNF468-mediated epigenetic upregulation of VEGF-C facilitates lymphangiogenesis and lymphatic metastasis in ESCC via PI3K/Akt and ERK1/2 signaling pathways. Cell Oncol (Dordr) 2024; 47:1927-1942. [PMID: 39141315 DOI: 10.1007/s13402-024-00976-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Dysfunctional lymphangiogenesis is pivotal for various pathological processes including tumor lymph node metastasis which is a crucial cause of therapeutic failure for ESCC. In this study, we aim to elucidate the molecular mechanisms and clinical relevance of Zinc-finger protein ZNF468 in lymphangiogenesis and lymphatic metastasis in ESCC. METHODS Immunohistochemistry, Western blot, Kaplan-Meier plotter analysis and Gene Set Enrichment Analysis were preformed to detect the association of ZNF468 with lymphangiogenesis and poor prognosis in ESCC patients. Foot-pads lymph node metastasis model, tube formation assay, 3D-culture assay and invasion assay were preformed to verify the effect of ZNF468 on lymphangiogenesis and lymph node metastasis. CUT&Tag analysis, immunoprecipitation and mass spectrometry analysis and ChIP-PCR assay were preformed to study the molecular mechanisms of ZNF468 in lymphangiogenesis. RESULTS We found that ectopic expression of ZNF468 was correlated with higher microlymphatic vessel density in ESCC tissues, leading to poorer prognosis of ESCC patients. ZNF468 enhanced the capacity of lymphangiogenesis and promoted lymphatic metastasis in ESCC both in vitro and in vivo. However, silencing ZNF468 reversed these phenotypes in ESCC. Mechanically, we demonstrated that ZNF468 recruits the histone modification factors (PRMT1/HAT1) to increase the levels of H4R2me2a and H3K9ac, which then leads to the recruitment of the transcription initiation complex on the VEGF-C promoter, ultimately promoting the upregulation of VEGF-C transcription. Strikingly, the promoting effect of lymphatic metastasis induced by ZNF468 in ESCC was abrogated by targeting PRMT1 using Arginine methyltransferase inhibitor-1 or silencing VEGF-C. Furthermore, we found that the activation of PI3K/AKT and ERK1/2 signaling is required for ZNF468-medicated lymphatic metastasis in ESCC. Importantly, the clinical relevance between ZNF468 and VEGF-C were confirmed not only in ESCC samples and but also in multiple cancer types. CONCLUSION Our results identified a precise mechanism underlying ZNF468-induced epigenetic upregulation of VEGF-C in facilitating lymphangiogenesis and lymph node metastasis of ESCC, which might provide a novel prognostic biomarker and potential therapeutic for ESCC patients.
Collapse
Affiliation(s)
- Jinrong Zhu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiangyu Qiu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xin Jin
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoya Nie
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shengming Ou
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Geyan Wu
- Biomedicine Research Centre, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provicial Clinical Research Center for Obsterics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Jianfei Shen
- Department of Cardiothoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China.
| | - Rongxin Zhang
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
41
|
Zhao XH, Zhang WC, Wang X, Chen JQ, Xu YJ, Zhao KL, Huang W, Qian PD, Liu YT, Ge XL, Xia XJ, Weng CG, Gai CY, Wang HS, Gao HM, Shen WB, Zhu SC. Dose escalation in radical radio(chemo)therapy for cervical and upper thoracic esophageal cancer with 3DCRT/IMRT (ChC&UES): a multicenter retrospective study. Radiat Oncol 2024; 19:126. [PMID: 39334163 PMCID: PMC11429629 DOI: 10.1186/s13014-024-02521-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Cervical and upper thoracic esophageal cancer (ESCA) presents treatment challenges due to limited clinical evidence. This multi-center study (ChC&UES) explores radical radio(chemo)therapy efficacy and safety, especially focusing on radiation dose. METHOD We retrospectively analyzed clinical data from 1,422 cases across 8 medical centers. According to the radiation dose for primary gross tumor, patients were divided into standard dose radiotherapy (SD, 50-55 Gy) or high dose (HD, > 55 Gy) radiotherapy. HD was further subdivided into conventional- high-dose group (HD-conventional, 55-63 Gy) and ultra-high-dose group (HD-ultra, ≥ 63 Gy). Primary outcome was Overall Survival (OS). RESULTS The median OS was 33.0 months (95% CI: 29.401-36.521) in the whole cohort. Compared with SD, HD shown significant improved survival in cervical ESCA in Kaplan-Meier (P = 0.029) and cox multivariate regression analysis (P = 0.024) while shown comparable survival in upper thoracic ESCA (P = 0.735). No significant difference existed between HD-conventional and HD-ultra in cervical (P = 0.976) and upper thoracic (P = 0.610) ESCA. Incidences of radiation esophagitis and pneumonia from HD were comparable to SD (P = 0.097, 0.240), while myosuppression risk was higher(P = 0.039). The Bonferroni method revealed that, for both cervical and upper thoracic ESCA, HD-ultra enhance the objective response rate (ORR) compared to SD (P < 0.05). CONCLUSION HD radiotherapy benefits cervical but not upper thoracic ESCA, while increasing bone marrow suppression risk. Further dose escalating (≥ 63 Gy) doesn't improve survival but enhances ORR.
Collapse
Affiliation(s)
- Xiao-Han Zhao
- Department of Radiation Oncology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - Wen-Cheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi District, Tianjin, 300060, China
| | - Xin Wang
- Department of Radiotherapy, National Cancer Center/National Cancer Clinical Medical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun-Qiang Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical Univercity, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Yuan-Ji Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical Univercity, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Kuai-Le Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wei Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Pu-Dong Qian
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Ya-Tian Liu
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xiao-Lin Ge
- Department of Radiation Oncology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 300, Guangzhou Road, Nanjing, Jiangsu, China
| | - Xiao-Jie Xia
- Department of Radiation Oncology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 300, Guangzhou Road, Nanjing, Jiangsu, China
| | - Chen-Gang Weng
- Department of Throacic Surgery Department, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chun-Yue Gai
- Department of Throacic Surgery Department, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - He-Song Wang
- Department of Radiation Oncology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - Hong-Mei Gao
- Department of Radiation, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Wen-Bin Shen
- Department of Radiation Oncology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China.
| | - Shu-Chai Zhu
- Department of Radiation Oncology, the Fourth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China.
| |
Collapse
|
42
|
Chen H, Ren C. Helicobacter pylori Eradication Treatment Might Help Reduce the Risk of Esophageal Adenocarcinoma. Gastroenterology 2024:S0016-5085(24)05477-5. [PMID: 39306253 DOI: 10.1053/j.gastro.2024.08.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 10/09/2024]
Affiliation(s)
- Hui Chen
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Chuanli Ren
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| |
Collapse
|
43
|
Tran P, Ancha A, Tjahja M, Shell M, Naumann C. Poor adherence to proper Barrett's esophagus screening and surveillance guidelines in patients with newly diagnosed esophageal adenocarcinoma. Proc AMIA Symp 2024; 37:922-926. [PMID: 39440080 PMCID: PMC11492693 DOI: 10.1080/08998280.2024.2397936] [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: 04/04/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 10/25/2024] Open
Abstract
Background Screening for Barrett's esophagus (BE) remains controversial, even for high-risk populations. Our study aimed to evaluate the proportion of patients diagnosed with esophageal adenocarcinoma (EAC) who were not screened for BE or did not receive recommended BE surveillance screening. We then evaluated the relationship between cancer staging and screening/surveillance opportunities. Methods This single-center retrospective study included 187 patients from January 2016 to January 2022 with newly diagnosed EAC. Data extracted from patient charts included BE risk factors, and BE, endoscopic, and histologic history. Results A total of 187 patients had a new diagnosis of EAC. Among this group, 44% had appropriate BE surveillance adherence, and 47% of patients met the criteria for BE screening but had not been screened prior to EAC diagnosis. Adherence to BE surveillance was associated with earlier stages of cancer on biopsy. No significant difference in cancer staging was found in those with missed BE screening opportunities. Discussion Patients with a diagnosis of BE who adhered to surveillance guidelines had earlier stage EAC at diagnosis, which emphasizes the importance of surveillance. Most of those with an initial diagnosis of EAC had not received any BE screening.
Collapse
Affiliation(s)
- Phi Tran
- Department of Internal Medicine, Baylor Scott and White Medical Center, Temple, Texas, USA
| | - Anupama Ancha
- Department of Internal Medicine, Baylor Scott and White Medical Center, Temple, Texas, USA
| | - Matthew Tjahja
- Department of Internal Medicine, Baylor Scott and White Medical Center, Temple, Texas, USA
| | - Mark Shell
- Division of Gastroenterology, Baylor Scott and White Medical Center, Temple, Texas, USA
| | - Christopher Naumann
- Division of Gastroenterology, Baylor Scott and White Medical Center, Temple, Texas, USA
| |
Collapse
|
44
|
Li X, Sun T, Li H, Liu J, Huang N, Liu S. The Novel-B-Cell-Related Gene Signature Predicts the Prognosis and Immune Status of Patients with Esophageal Carcinoma. J Gastrointest Cancer 2024; 55:1313-1323. [PMID: 38963643 PMCID: PMC11347472 DOI: 10.1007/s12029-024-01083-x] [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: 06/15/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND The current understanding of the prognostic significance of B cells and their role in the tumor microenvironment (TME) in esophageal carcinoma (ESCA) is limited. METHODS We conducted a screening for B-cell-related genes through the analysis of single-cell transcriptome data. Subsequently, we developed a B-cell-related gene signature (BRGrisk) using LASSO regression analysis. Patients from The Cancer Genome Atlas cohort were divided into a training cohort and a test cohort. Patients were categorized into high- and low-risk groups based on their median BRGrisk scores. The overall survival was assessed using the Kaplan-Meier method, and a nomogram based on BRGrisk was constructed. Immune infiltration profiles between the risk groups were also compared. RESULTS The BRGrisk prognostic model indicated significantly worse outcomes for patients with high BRGrisk scores (p < 0.001). The BRGrisk-based nomogram exhibited good prognostic performance. Analysis of immune infiltration revealed that patients in the high-BRGrisk group had notably higher levels of immune cell infiltration and were more likely to be in an immunoresponsive state. Enrichment analysis showed a strong correlation between the prognostic gene signature and cancer-related pathways. IC50 results indicated that patients in the low-BRGrisk group were more responsive to common drugs compared to those in the high-BRGrisk group. CONCLUSIONS This study presents a novel BRGrisk that can be used to stratify the prognosis of ESCA patients and may offer guidance for personalized treatment strategies aimed at improving prognosis.
Collapse
Affiliation(s)
- Xinhong Li
- Department of Oncohematology, Norinco General Hospital, Xi'an, Shaanxi, 710061, China
| | - Tongyu Sun
- Hepatobiliary and Vascular Surgery, Norinco General Hospital, Xi'an, Shaanxi, 710061, China
| | - Hongyan Li
- Department of Radiology, Norinco General Hospital, Xi'an, Shaanxi, 710061, China
| | - Juan Liu
- Department of Oncohematology, Norinco General Hospital, Xi'an, Shaanxi, 710061, China
| | - Na Huang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Surong Liu
- Department of Oncohematology, Norinco General Hospital, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
45
|
Liu D, van der Zalm AP, Koster J, Bootsma S, Oyarce C, van Laarhoven HWM, Bijlsma MF. Predictive biomarkers for response to TGF- β inhibition in resensitizing chemo(radiated) esophageal adenocarcinoma. Pharmacol Res 2024; 207:107315. [PMID: 39059615 DOI: 10.1016/j.phrs.2024.107315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/26/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Epithelial-mesenchymal transition (EMT) has been identified as a driver of therapy resistance, particularly in esophageal adenocarcinoma (EAC), where transforming growth factor beta (TGF-β) can induce this process. Inhibitors of TGF-β may counteract the occurrence of mesenchymal, resistant tumor cell populations following chemo(radio)therapy and improve treatment outcomes in EAC. Here, we aimed to identify predictive biomarkers for the response to TGF-β targeting. In vitro approximations of neoadjuvant treatment were applied to publicly available primary EAC cell lines. TGF-β inhibitors fresolimumab and A83-01 were employed to inhibit EMT, and mesenchymal markers were quantified via flow cytometry to assess efficacy. Our results demonstrated a robust induction of mesenchymal cell states following chemoradiation, with TGF-β inhibition leading to variable reductions in mesenchymal markers. The cell lines were clustered into responders and non-responders. Genomic expression profiles were obtained through RNA-seq analysis. Differentially expressed gene (DEG) analysis identified 10 positively- and 23 negatively-associated hub genes, which were bioinformatically identified. Furthermore, the correlation of DEGs with response to TGF-β inhibition was examined using public pharmacogenomic databases, revealing 9 positively associated and 11 negatively associated DEGs. Among these, ERBB2, EFNB1, and TNS4 were the most promising candidates. Our findings reveal a distinct gene expression pattern associated with the response to TGF-β inhibition in chemo(radiated) EAC. The identified DEGs and predictive markers may assist patient selection in clinical studies investigating TGF-β targeting.
Collapse
Affiliation(s)
- Dajia Liu
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| | - Amber P van der Zalm
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Jan Koster
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Sanne Bootsma
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| | - Cesar Oyarce
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands.
| |
Collapse
|
46
|
Idalsoaga F, Díaz LA, Ayares G, Cabrera D, Chahuan J, Monrroy H, Halawi H, Arrese M, Arab JP. Review article: Oesophageal disorders in chronic liver disease. Aliment Pharmacol Ther 2024; 60:715-726. [PMID: 39082463 DOI: 10.1111/apt.18193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/03/2024] [Accepted: 07/20/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Oesophageal disorders and chronic liver disease are common worldwide and significantly impact quality of life. The intricate link between these conditions, including how oesophageal disorders like GERD, Barrett's oesophagus and oesophageal cancer affect and are affected by chronic liver disease, remains poorly understood. AIMS To review the relationship between oesophageal disorders and chronic liver disease, evaluating epidemiology, pathophysiology and therapeutic factors. METHODS We reviewed the literature on the relationship between oesophageal disorders and chronic liver disease, including cirrhosis, using the PubMed database RESULTS: Oesophageal disorders such as gastroesophageal reflux disease, Barrett's oesophagus, oesophageal cancer, oesophageal motor disorders and oesophageal candidiasis are prevalent among individuals with cirrhosis, exacerbating the burden of liver disease. These diseases have a multifaceted symptomatology and pathogenic basis, posing a significant challenge in cirrhotic patients that necessitates careful diagnosis and management. Additionally, therapies frequently used for these diseases, such as proton pump inhibitors, require careful consideration in cirrhotic patients due to potential adverse effects and altered pharmacokinetics. Managing oesophageal disorders in cirrhotic patients requires a cautious approach due to possible interactions with medications and the risk of adverse effects. Furthermore, symptoms associated with these conditions are often exacerbated by common interventions in patients with cirrhosis, such as band ligation for oesophageal varices. CONCLUSIONS Oesophageal disorders are common in cirrhosis and increase the disease burden. These conditions require careful management due to complex symptoms and treatment risks. Proton pump inhibitors and other therapies must be used cautiously, as cirrhosis interventions can worsen symptoms.
Collapse
Affiliation(s)
- Francisco Idalsoaga
- Departamento De Gastroenterología, Escuela De Medicina, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Luis Antonio Díaz
- Departamento De Gastroenterología, Escuela De Medicina, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Gustavo Ayares
- Departamento De Gastroenterología, Escuela De Medicina, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Daniel Cabrera
- Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Centro de Estudios e Investigación en Salud y Sociedad, Escuela de Medicina, Facultad de Ciencias Médicas, Universidad Bernardo O Higgins, Santiago, Chile
| | - Javier Chahuan
- Departamento De Gastroenterología, Escuela De Medicina, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Hugo Monrroy
- Departamento De Gastroenterología, Escuela De Medicina, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Houssam Halawi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Marco Arrese
- Departamento De Gastroenterología, Escuela De Medicina, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento De Gastroenterología, Escuela De Medicina, Pontificia Universidad Católica De Chile, Santiago, Chile
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
47
|
Papaefthymiou A, Norton B, Telese A, Ramai D, Murino A, Gkolfakis P, Vargo J, Haidry RJ. Efficacy and Safety of Cryoablation in Barrett's Esophagus and Comparison with Radiofrequency Ablation: A Meta-Analysis. Cancers (Basel) 2024; 16:2937. [PMID: 39272792 PMCID: PMC11394299 DOI: 10.3390/cancers16172937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND The mainstay approach in endoscopic eradication therapy (EET) for dysplastic Barrett's esophagus (BE) includes the endoscopic resection of visible lesions, accompanied by ablation of the residual metaplastic epithelium. Cryoablation therapy is one such emerging ablation technique in this field. This systematic review with a meta-analysis aims to accumulate pooled data on cryoablation performance in the treatment of patients with BE and to compare this technique to the standard of care radiofrequency ablation (RFA). METHODS The MEDLINE, Cochrane, and Scopus databases were searched until June 2024 for studies evaluating BE management using cryoablation for cumulative results. The primary outcome was the complete eradication of dysplasia (CED) and intestinal metaplasia (CEIM) in BE compared to RFA, while secondary outcomes included the respective pooled rates using cryoablation, recurrence, and adverse events, with a separate analysis for strictures. The meta-analyses were based on a random-effects model, and the results were reported as odds ratios (ORs) with 95% confidence intervals (CIs). Subgroup analyses by type of cryoablation were also performed. RESULTS Twenty-three studies (1604 patients) were finally included, four of which were comparative. CED and CEIM did not differ significantly between cryoablation and RFA [OR= 0.95 (95%CI: 0.50-1.81) and OR = 0.57 (95%CI: 0.20-1.63), respectively)]. The pooled rates of CED, CEIM, and recurrence after cryoablation were 84.2% (95%CI: 79.1-89.3), 64.1% (95%CI: 49.2-79.0), and 8.3% (95%CI: 4.7-11.9), accompanied by high rates of heterogeneity. Adverse events were noted in 14.5% (95%CI: 9.9-19.2) of cases, and 6.5% (95%CI: 4.1-9.0) developed strictures. In the subgroup analysis, the cryoballoon achieved a reduction in heterogeneity in CED, adverse events, and stricture formation, whereas spray catheters provided homogenous results in terms of recurrence. CONCLUSIONS Cryoablation provides equal outcomes compared to RFA in the treatment of patients with BE, with the cryoballoon achieving relatively homogenous rates of CED and adverse events.
Collapse
Affiliation(s)
| | - Benjamin Norton
- Digestive Diseases and Surgery Institute, Cleveland Clinic, London SW1X 7HY, UK
| | - Andrea Telese
- Digestive Diseases and Surgery Institute, Cleveland Clinic, London SW1X 7HY, UK
| | - Daryl Ramai
- Gastroenterology and Hepatology, University of Utah Health, Salt Lake City, UT 84132, USA
| | - Alberto Murino
- Digestive Diseases and Surgery Institute, Cleveland Clinic, London SW1X 7HY, UK
| | - Paraskevas Gkolfakis
- Department of Gastroenterology, "Konstantopoulio-Patision" General Hospital of Nea Ionia, 142 33 Athens, Greece
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme University Hospital, Université Libre De Bruxelles (ULB), 1070 Brussels, Belgium
| | - John Vargo
- Department of Gastroenterology, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rehan J Haidry
- Digestive Diseases and Surgery Institute, Cleveland Clinic, London SW1X 7HY, UK
| |
Collapse
|
48
|
Wu X, Liu S, Li F, Chen Y. Association between preoperative neutrophil-to-lymphocyte ratio and the survival outcomes of esophageal cancer patients underwent esophagectomy: a systematic review and meta-analysis. Front Oncol 2024; 14:1404711. [PMID: 39224809 PMCID: PMC11366628 DOI: 10.3389/fonc.2024.1404711] [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: 03/21/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Objectives The purpose of this study was to assess the association between preoperative neutrophil-to-lymphocyte ratio (NLR) and the survival outcomes of esophageal cancer patients who underwent esophagectomy, the latest and comprehensive systematic review performed. Methods Related literature retrieved from PubMed, Web of Science, Embase, and Cochrane before January 2024, according to the inclusion criteria. Outcomes measured were overall survival (OS), disease-free survival (DFS), relapse-free survival (RFS), and cancer-specific survival (CSS). Results Eighteen studies with 6,119 esophageal cancer patients were retained for analysis. Meta-analysis demonstrated that OS (HR: 1.47; 95% CI: 1.29, 1.67; P < 0.00001), DFS (HR: 1.62; 95% CI: 1.29, 2.05; P < 0.0001), and CSS (HR: 1.62; 95% CI: 1.29, 2.05; P < 0.0001) were significantly shorter in the high NLR group compared with the low NLR group. In addition, meta-analysis revealed a similar RFS (HR: 1.47; 95% CI: 0.92, 2.35; P = 0.10) among the two groups. Subgroup analysis of OS and DFS based on mean/median age, NLR cutoff, and region found that all subgroups remained significant difference between two groups. Conclusion Among esophageal cancer patients who underwent esophagectomy, preoperative NLR can be used as prognostic factor independently. High-preoperative NLR is associated with poor prognosis. More large-scale, multicenter prospective clinical studies are needed to further validate the relationship between preoperative NLR and prognosis of esophageal cancer.
Collapse
Affiliation(s)
| | | | | | - YingTai Chen
- Department of Thoracic Surgery, Beijing Aerospace General Hospital, Beijing, China
| |
Collapse
|
49
|
Laun SE, Kann L, Braun J, Gilbert S, Lunz D, Pierre F, Kalra A, Ma K, Tsai HL, Wang H, Jit S, Cheng Y, Ahmed Y, Wang KK, Leggett CL, Cellini A, Ioffe OB, Zaidi AH, Omstead AN, Jobe B, Korman L, Cornish D, Zellenrath P, Spaander M, Kuipers E, Perpetua L, Greenwald BD, Maddala T, Meltzer SJ. Validation of an Epigenetic Prognostic Assay to Accurately Risk-Stratify Patients with Barrett's Esophagus. Am J Gastroenterol 2024; 120:00000434-990000000-01289. [PMID: 39140473 PMCID: PMC11825890 DOI: 10.14309/ajg.0000000000003030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/20/2024] [Indexed: 08/15/2024]
Abstract
INTRODUCTION: Esophageal adenocarcinoma (EAC) is the second-most lethal cancer in the United States, with Barrett esophagus (BE) being the strongest risk factor. Assessing the future risk of neoplastic progression in patients with BE is difficult; however, high-grade dysplasia (HGD) and early EAC are treatable by endoscopic eradication therapy (EET), with survival rates of 90%. Thus, it would be beneficial to develop a molecular assay to identify high-risk patients, who merit more frequent endoscopic surveillance or EET, as well as low-risk patients, who can avoid EET and undergo less frequent surveillance. METHODS: Deidentified endoscopic biopsies were acquired from 240 patients with BE at 6 centers and confirmed as future progressors or nonprogressors. Tissues were analyzed by a set of methylation-specific biomarker assays. Test performance was assessed in an independent validation set using 4 stratification levels: low risks, low-moderate risks, high-moderate risks, and high risks. RESULTS: Relative to patients in the low-risk group, high-risk patients were 15.2 times more likely to progress within 5 years to HGD or EAC. For patients in the high-risk category, the average risk of progressing to HGD or EAC within 5 years was 21.5%, 4-fold the BE population prevalence within 5 years, whereas low-risk patients had a progression risk of only 1.85%. DISCUSSION: This clinical assay, Esopredict, stratifies future neoplastic progression risk to identify higher-risk patients with BE who can benefit from EET or more frequent surveillance and lower-risk patients who can benefit from reduced surveillance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andrew Kalra
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ke Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Biostatistics, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hao Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Biostatistics, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Simran Jit
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yulan Cheng
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yousra Ahmed
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kenneth K. Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Cadman L. Leggett
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ashley Cellini
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Olga B. Ioffe
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ali H. Zaidi
- Esophageal Institute, Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Ashten N. Omstead
- Esophageal Institute, Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Blair Jobe
- Department of Surgery, Esophageal Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Drexel University, Philadelphia, Pennsylvania, USA
| | - Louis Korman
- Capital Digestive Care, Chevy Chase, Maryland, USA
| | - Drew Cornish
- Capital Digestive Care, Chevy Chase, Maryland, USA
| | - Pauline Zellenrath
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Manon Spaander
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Ernst Kuipers
- Department of Gastroenterology & Hepatology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Lorrie Perpetua
- Research Tissue Biorepository Core Facility, University of Connecticut, Storrs, Connecticut, USA
| | - Bruce D. Greenwald
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Stephen J. Meltzer
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
50
|
Zhang J, Han H, Liu Y, Xu J, Zhang D, Wang W, Gao Y, Li Z, Qin Y. SKF96365 Inhibits Tumor Proliferation by Inducing Apoptosis and Autophagy in Human Esophageal Squamous Cell Carcinoma. Int J Genomics 2024; 2024:4501154. [PMID: 39165489 PMCID: PMC11335422 DOI: 10.1155/2024/4501154] [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: 03/14/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 08/22/2024] Open
Abstract
Calcium channel blockers are emerging as a new generation of attractive anticancer drugs. SKF96365, originally thought to be a store-operated calcium entry (SOCE) inhibitor, is now often used as a TRPC channel blocker and is widely used in medical diagnostics. SKF96365 has shown antitumor effects on a variety of cancer cell lines. The objective of this study was to investigate the anticancer effect of SKF96365 on esophageal cancer in vivo and in vitro. Cell Counting Kit-8 (CCK-8) and colony formation were used to test the proliferation inhibition of SKF96365 on cell lines. Western blot and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining were used to detect cell apoptosis rates. In addition, we demonstrated the antitumor effect of SKF96365 in vivo in xenografted mice. As a result, SKF96365 significantly inhibited the proliferation of K510, K30, and EC9706 in vitro. SKF96365 induces apoptosis in three cell lines through the poly(adenosine diphosphate-ribose) polymerase (PARP), caspase-9, and BCL-2 pathways in a dose-dependent and time-dependent manner. Moreover, SKF96365 treatment also induced apoptosis and inhibited tumor growth in nude mice. The calcium channel TRPC1 was significantly downregulated by SKF96365. Autophagy was also induced during the treatment of SKF96365. In summary, SKF96365 induces apoptosis (PARP, caspase-9, and BCL-2) and autophagy (LC3-A/B) by inhibiting TRPC1 in esophageal cancer cells, thereby inhibiting tumor growth.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Huiqiong Han
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yihan Liu
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jiayao Xu
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Daidi Zhang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wenjia Wang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yaping Gao
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanru Qin
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|