1
|
Ye C, Zhang H, Chi Z, Xu Z, Cai Y, Xu Y, Tong X. Machine Learning-Based Multimodal Radiomics and Transcriptomics Models for Predicting Radiotherapy Sensitivity and Prognosis in Esophageal Cancer. J Biol Chem 2025:110242. [PMID: 40381695 DOI: 10.1016/j.jbc.2025.110242] [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: 01/24/2025] [Revised: 04/23/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025] Open
Abstract
Radiotherapy plays a critical role in treating esophageal cancer, but individual responses vary significantly, impacting patient outcomes. This study integrates machine learning-driven multimodal radiomics and transcriptomics to develop predictive models for radiotherapy sensitivity and prognosis in esophageal cancer. We applied the SEResNet101 deep learning model to imaging and transcriptomic data from the UCSC Xena and TCGA databases, identifying prognosis-associated genes such as STUB1, PEX12, and HEXIM2. Using Lasso regression and Cox analysis, we constructed a prognostic risk model that accurately stratifies patients based on survival probability. Notably, STUB1, an E3 ubiquitin ligase, enhances radiotherapy sensitivity by promoting the ubiquitination and degradation of SRC, a key oncogenic protein. In vitro and in vivo experiments confirmed that STUB1 overexpression or SRC silencing significantly improves radiotherapy response in esophageal cancer models. These findings highlight the predictive power of multimodal data integration for individualized radiotherapy planning and underscore STUB1 as a promising therapeutic target for enhancing radiotherapy efficacy in esophageal cancer.
Collapse
Affiliation(s)
- Chengyu Ye
- The Affiliated Cancer Hospital of Wenzhou Medical University, Wenzhou Central Hospital, Wenzhou, 325000, PR China
| | - Hao Zhang
- The Affiliated Cancer Hospital of Wenzhou Medical University, Wenzhou Central Hospital, Wenzhou, 325000, PR China
| | - Zhou Chi
- The Affiliated Cancer Hospital of Wenzhou Medical University, Wenzhou Central Hospital, Wenzhou, 325000, PR China
| | - Zhina Xu
- The Affiliated Cancer Hospital of Wenzhou Medical University, Wenzhou Central Hospital, Wenzhou, 325000, PR China
| | - Yujie Cai
- The Affiliated Cancer Hospital of Wenzhou Medical University, Wenzhou Central Hospital, Wenzhou, 325000, PR China
| | - Yajing Xu
- The Affiliated Cancer Hospital of Wenzhou Medical University, Wenzhou Central Hospital, Wenzhou, 325000, PR China
| | - Xiangmin Tong
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China.
| |
Collapse
|
2
|
Zeng B, Sun C, Li N, Chen Q, Rao M, Li K, Liu X, Xie S, Cheng J, Wang S, Wang X. NEK2 Control of Esophageal Squamous Cell Carcinoma Growth Based on Circadian Oscillation. Cancer Sci 2025; 116:1282-1294. [PMID: 39963019 PMCID: PMC12044676 DOI: 10.1111/cas.16461] [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: 09/16/2024] [Revised: 01/10/2025] [Accepted: 01/19/2025] [Indexed: 05/02/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a globally prevalent malignancy known for its aggressive nature and unfavorable outcomes. Identifying new biomarkers is crucial for the early detection and improved prognostication of ESCC. The circadian clock and NIMA-related kinase 2 (NEK2) are pivotal in cancer development. While the impact of circadian rhythm disruptions on ESCC progression is evident, the specific contribution of NEK2 to these changes is not well understood. Our study discovered NEK2 as a consistently differentially expressed gene across multiple datasets, with elevated expression in ESCC tissues. Notably, NEK2 overexpression was linked to increased ESCC cell proliferation, whereas its inhibition led to reduced cell growth and proliferation. Pathway analyses, including KEGG and Gene Set Enrichment Analysis (GSEA), indicated NEK2's association with established pathways like the cell cycle, and intriguingly, identified the circadian rhythm as a novel pathway influenced by NEK2. RNA sequencing data demonstrated NEK2's circadian rhythmic expression, and subsequent in vitro experiments confirmed its oscillation in synchronized ESCC cells. Moreover, we found a positive correlation between the efficacy of the NEK2 inhibitor INH6 and NEK2 expression levels in ESCC. In conclusion, our findings position NEK2 as a time-dependent oncogene and a potential biomarker in ESCC, highlighting its role in both tumorigenesis and the circadian rhythm.
Collapse
Affiliation(s)
- Boning Zeng
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
- Department of General Practice, Shenzhen Luohu People's HospitalThe Third Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Chao Sun
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Nan Li
- School of Pharmaceutical SciencesShenzhen University Medical SchoolShenzhenChina
- Department of PharmacyCancer Hospital of Shantou University Medical CollegeShantouChina
| | - Qiuling Chen
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Manni Rao
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Kai Li
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Xiaodi Liu
- Department of Infectious Disease, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Shouxia Xie
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| | - Jiwu Cheng
- Department of Trauma Orthopedic, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenGuangdongChina
| | - Shaoxiang Wang
- School of Pharmaceutical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's Hospital (The Second Clinical Medical CollegeJinan University; The First Affiliated Hospital, Southern University of Science and Technology)ShenzhenChina
| |
Collapse
|
3
|
Jing T, Tang D. Intratumoral microbiota: a new force in the development and treatment of esophageal cancer. Clin Transl Oncol 2025; 27:1921-1932. [PMID: 39455494 DOI: 10.1007/s12094-024-03757-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Esophageal cancer (EC) ranks among the most prevalent cancers worldwide, with a particularly high incidence in the Asian population. Due to the inconspicuous nature of early symptoms, patients with esophageal cancer are typically diagnosed in the middle to late stages, resulting in suboptimal overall treatment outcomes. Consequently, there is an urgent need to explore and refine therapeutic strategies. Microorganisms have been identified in numerous tumor tissues, including EC, and these microorganisms are referred to as the intratumoral microbiome. Intratumoral microbiota and their metabolic byproducts can influence the progression and treatment of esophageal cancer through various mechanisms, such as modulating tumor cell metabolism and local immune responses. Therefore, the intratumoral microbiota may potentially serve as a target for the treatment of esophageal cancer. This review delineates the composition, origin, and diagnostic significance of intratumoral microbiota in esophageal cancer tissue, and discusses the mechanisms by which intratumoral microbiota contribute to the onset of esophageal cancer. In addition, the impact of intratumoral microbiota on the treatment of esophageal cancer and its intervention measures are also addressed.
Collapse
Affiliation(s)
- Tianyang Jing
- Clinical Medical College, Yangzhou University, Yangzhou, 22500, Jiangsu Province, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
| |
Collapse
|
4
|
Liu S, Lin Z, Huang Z, Yu M, Lin Z, Hu Z. Unique Microbial Characterisation of Oesophageal Squamous Cell Carcinoma Patients with Different Dietary Habits Based on Light Gradient Boosting Machine Learning Classifier. Nutrients 2025; 17:1340. [PMID: 40284204 PMCID: PMC12030675 DOI: 10.3390/nu17081340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Objectives: The microbiome plays an important role in cancer, but the relationship between dietary habits and the microbiota in oesophageal squamous cell carcinoma (ESCC) is not clear. The aim of this study is to explore the complex relationship between the microbiota in oesophagal tissue and dietary habits in ESCC patients. Methods: 173 ESCC patients were included. The method of 16S rRNA sequencing was used to analyze microbial composition and diversity. The LEfSe and Boruta methods were used to screen important microbes, and the LightGBM algorithm distinguished microbes associated with different dietary habits. PICRUST2 and DESeq2 predicted microbial function and screened differential functions. The Pearson test was used to analyze correlations between microbes and functions, and SPARCC microbial symbiotic networks and Cytoscape were used to determine microbial interactions. Results: Significant differences in microbial composition were observed among ESCC patients with different dietary habits. LEfSe and Boruta identified three, six, and two significantly different bacteria in the FF/FP, FF/PF, and FF/PP groups, respectively, with AUC values of 0.683, 0.830, and 0.715. PICRUST2 and DESeq2 analysis revealed 3, 11, and 5 significantly different metabolic pathways in each group. Eubacterium_B sulci was positively correlated with PWY-6285, PWY-3801, and PWY-5823. PWY-6397 was positively correlated with undefinded (Fusobacterium_C). Microbial network analysis confirmed unique microbial characteristics in different diet groups. Conclusions: Different dietary habits lead to alterations in Eubacterium_B sulci and undefinded (Fusobacterium_C) and related functional pathways.
Collapse
Affiliation(s)
- Shun Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Zhifeng Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Zhimin Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Menglin Yu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Zheng Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China; (S.L.); (Z.L.); (Z.H.); (M.Y.); (Z.L.)
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
5
|
Gao Y, Wen P, Liu Y, Sun Y, Qian H, Zhang X, Peng H, Gao Y, Li C, Gu Z, Zeng H, Hong Z, Wang W, Yan R, Hu Z, Fu H. Application of artificial intelligence in the diagnosis of malignant digestive tract tumors: focusing on opportunities and challenges in endoscopy and pathology. J Transl Med 2025; 23:412. [PMID: 40205603 PMCID: PMC11983949 DOI: 10.1186/s12967-025-06428-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: 11/20/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Malignant digestive tract tumors are highly prevalent and fatal tumor types globally, often diagnosed at advanced stages due to atypical early symptoms, causing patients to miss optimal treatment opportunities. Traditional endoscopic and pathological diagnostic processes are highly dependent on expert experience, facing problems such as high misdiagnosis rates and significant inter-observer variations. With the development of artificial intelligence (AI) technologies such as deep learning, real-time lesion detection with endoscopic assistance and automated pathological image analysis have shown potential in improving diagnostic accuracy and efficiency. However, relevant applications still face challenges including insufficient data standardization, inadequate interpretability, and weak clinical validation. OBJECTIVE This study aims to systematically review the current applications of artificial intelligence in diagnosing malignant digestive tract tumors, focusing on the progress and bottlenecks in two key areas: endoscopic examination and pathological diagnosis, and to provide feasible ideas and suggestions for subsequent research and clinical translation. METHODS A systematic literature search strategy was adopted to screen relevant studies published between 2017 and 2024 from databases including PubMed, Web of Science, Scopus, and IEEE Xplore, supplemented with searches of early classical literature. Inclusion criteria included studies on malignant digestive tract tumors such as esophageal cancer, gastric cancer, or colorectal cancer, involving the application of artificial intelligence technology in endoscopic diagnosis or pathological analysis. The effects and main limitations of AI diagnosis were summarized through comprehensive analysis of research design, algorithmic methods, and experimental results from relevant literature. RESULTS In the field of endoscopy, multiple deep learning models have significantly improved detection rates in real-time polyp detection, early gastric cancer, and esophageal cancer screening, with some commercialized systems successfully entering clinical trials. However, the scale and quality of data across different studies vary widely, and the generalizability of models to multi-center, multi-device environments remains to be verified. In pathological analysis, using convolutional neural networks, multimodal pre-training models, etc., automatic tissue segmentation, tumor grading, and assisted diagnosis can be achieved, showing good scalability in interactive question-answering. Nevertheless, clinical implementation still faces obstacles such as non-uniform data standards, lack of large-scale prospective validation, and insufficient model interpretability and continuous learning mechanisms. CONCLUSION Artificial intelligence provides new technological opportunities for endoscopic and pathological diagnosis of malignant digestive tract tumors, achieving positive results in early lesion identification and assisted decision-making. However, to achieve the transition from research to widespread clinical application, data standardization, model reliability, and interpretability still need to be improved through multi-center joint research, and a complete regulatory and ethical system needs to be established. In the future, artificial intelligence will play a more important role in the standardization and precision management of diagnosis and treatment of digestive tract tumors.
Collapse
Affiliation(s)
- Yinhu Gao
- Department of Gastroenterology, Shaanxi Province Rehabilitation Hospital, Xi'an, Shaanxi, China
| | - Peizhen Wen
- Department of General Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Yuan Liu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yahuang Sun
- Division of Colorectal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Hui Qian
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Xin Zhang
- Department of Gastrointestinal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Huan Peng
- Division of Colorectal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Yanli Gao
- Infection Control Office, Shaanxi Province Rehabilitation Hospital, Xi'an, Shaanxi, China
| | - Cuiyu Li
- Department of Radiology, The First Hospital of Nanchang, the Third Affiliated Hospital of Nanchang University, Nanchang, 330008, Jiangxi, China
| | - Zhangyuan Gu
- Tongji University School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Huajin Zeng
- Department of General Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Zhijun Hong
- Tongji University School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Weijun Wang
- Department of Gastrointestinal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Ronglin Yan
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Zunqi Hu
- Department of Gastrointestinal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Hongbing Fu
- Department of Gastrointestinal Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
6
|
Ding G, Li X, Yin H, Tang J, Sun B, Sheng JX. The role of oncostatin M in esophageal cancer and its potential as a prognostic maker. Comput Biol Med 2025; 191:110073. [PMID: 40198992 DOI: 10.1016/j.compbiomed.2025.110073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Esophageal cancer (ESCA) remains one of the most lethal malignancies worldwide, with poor survival rates. Identifying novel biomarkers like Oncostatin M (OSM) could improve early detection, prognosis, and therapeutic strategies. OSM, a cytokine from the IL-6 family, plays complex roles in cancer biology, but its specific function in ESCA is poorly understood. MATERIALS AND METHODS Gene expression data from 198 ESCA samples were retrieved from The Cancer Genome Atlas (TCGA) and GEO databases. Bioinformatic analyses, including differential expression and survival analysis, were conducted using R software. Functional experiments involved OSM knockdown in ESCA cell lines, followed by cell proliferation, migration, colony formation assays, and wound healing analysis. RESULTS OSM expression was significantly elevated in ESCA tissues compared to normal tissues. High OSM expression correlated with poor overall survival and was associated with clinical parameters such as tumor stage and lymph node metastasis. In vitro experiments demonstrated that OSM knockdown reduced cell proliferation, colony formation, and migration in ESCA cell lines. CONCLUSIONS This study found that OSM overexpression in esophageal cancer was linked to advanced stage, lymph node metastasis, and poor prognosis. A prognostic nomogram based on OSM expression and clinical features predicted patient survival. Functional analysis suggested that OSM influences cancer progression via cytokine-receptor signaling, and silencing OSM reduced cell proliferation and migration, potentially improving prognosis.
Collapse
Affiliation(s)
- Guangxiang Ding
- Department of Radiation Oncology, The First Affiliated Hospital of Dalian Medical University, No.222 Zhongshan Road, Liaoning Province, Dalian, 116011, China.
| | - Xianwei Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| | - Hongtao Yin
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| | - Jianwei Tang
- Department of Radiation Oncology, The First Affiliated Hospital of Dalian Medical University, No.222 Zhongshan Road, Liaoning Province, Dalian, 116011, China.
| | - Bing Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| | - Jun Xiu Sheng
- Department of Radiation Oncology, The First Affiliated Hospital of Dalian Medical University, No.222 Zhongshan Road, Liaoning Province, Dalian, 116011, China.
| |
Collapse
|
7
|
Li K, Yi Y, Ling R, Zhang C, Zhang Z, Wang Y, Wang G, Chen J, Cheng M, Chen S. PCIF1 drives oesophageal squamous cell carcinoma progression via m6Am-mediated suppression of MTF2 translation. Clin Transl Med 2025; 15:e70286. [PMID: 40156159 PMCID: PMC11953057 DOI: 10.1002/ctm2.70286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 04/01/2025] Open
Abstract
Oesophageal squamous cell carcinoma (OSCC) represents a highly aggressive malignancy with limited therapeutic options and poor prognosis. This study uncovers PCIF1 as a critical driver of OSCC progression via m6Am RNA modification, leading to translational repression of the tumour suppressor MTF2. Our results demonstrate that PCIF1 selectively suppresses MTF2 translation, activating oncogenic pathways that promote OSCC growth. In vitro and in vivo models confirm that PCIF1 knockdown reduces OSCC progression, whereas MTF2 knockdown counteracts this effect, highlighting the importance of the PCIF1-MTF2 axis. Clinical analyses further reveal that high PCIF1 expression and low MTF2 expression correlate with advanced tumour stage, poor treatment response and decreased overall survival. Furthermore, in a preclinical mouse model, PCIF1 knockout enhanced the efficacy of anti-PD1 immunotherapy, reducing tumour burden and improving histological outcomes. Notably, flow cytometry analysis indicated that PCIF1 primarily exerts its effects through tumour-intrinsic mechanisms rather than direct modulation of the immune microenvironment, distinguishing its mode of action from PD1 blockade. These findings establish PCIF1 and MTF2 as promising prognostic markers and therapeutic targets for OSCC, offering new avenues for treatment strategies and patient stratification. KEY POINTS: PCIF1 promotes OSCC progression via m6Am methylation at the MTF2 mRNA 5' cap. m6Am methylation suppresses MTF2 translation, enhancing tumour cell proliferation and invasion. Targeting PCIF1 holds therapeutic potential for OSCC treatment.
Collapse
Affiliation(s)
- Kang Li
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuxuan Yi
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Rongsong Ling
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Caihua Zhang
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhihui Zhang
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer MedicineGuangzhouChina
| | - Yue Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of StomatologySun Yat‐Sen UniversityGuangzhouChina
| | - Ganping Wang
- Department of Urology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jie Chen
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of StomatologySun Yat‐Sen UniversityGuangzhouChina
| | - Maosheng Cheng
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shuang Chen
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
8
|
Yan J, Liu Z, Chen H, Sun X, Ge X, Xia X. Real-World Assessment of Trilaciclib for the Prevention of Chemoradiotherapy-Induced Myelosuppression in Esophageal Squamous Cell Carcinoma: A Propensity Score Matching Study. Cancer Med 2025; 14:e70862. [PMID: 40232146 PMCID: PMC11998604 DOI: 10.1002/cam4.70862] [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: 11/12/2024] [Revised: 03/01/2025] [Accepted: 03/29/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Chemoradiotherapy-induced myelosuppression (CIM) is the most common adverse event of esophageal cancer treatment, often necessitating reductions or delays in chemotherapy. Current treatments target specific blood cells, causing adverse effects. Trilaciclib, a novel CDK4/6 inhibitor with myeloprotective effects, has not yet been evaluated for its use in esophageal cancer treatment. We aimed to investigate the efficacy and safety of trilaciclib in preventing CIM. METHODS Clinical data were retrospectively collected from 203 patients with esophageal cancer who underwent concurrent radiotherapy at the Department of Radiotherapy of Jiangsu Province People's Hospital between January 2022 and January 2024. Patients were divided into the trilaciclib group (34 patients) and control group (169 patients). Propensity score matching (PSM) was performed to balance the baseline characteristics, and the incidence of myelosuppression and adverse events was compared. RESULTS Following PSM, 34 patients were included in each group, with no significant differences in baseline characteristics. The trilaciclib group exhibited significantly higher leukocyte, neutrophil, hemoglobin, and platelet levels (p < 0.05). The trilaciclib group exhibited a lower incidence of grade III-IV neutropenia and leukopenia, and none developed febrile neutropenia. Objective remission and disease control rates were comparable between the groups, with 1-year overall survival and progression-free survival rates of 82.0% and 73.4% in the trilaciclib group and 78.9% and 72.7% in the control group (not significant). The incidence of non-hematological toxic events was similar between the groups (p > 0.05). CONCLUSION Trilaciclib prevented myelosuppression in patients with esophageal cancer undergoing concurrent chemoradiotherapy, demonstrating good safety and efficacy.
Collapse
Affiliation(s)
- Jingze Yan
- Department of Radiation OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Zeyuan Liu
- Department of Radiation OncologyThe Affiliated Jiangning Hospital of Nanjing Medical UniversityNanjingChina
- Department of OncologyKangda College of Nanjing Medical UniversityLianyungangChina
| | - Hui Chen
- Department of Radiation OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xinchen Sun
- Department of Radiation OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xiaolin Ge
- Department of Radiation OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xiaojie Xia
- Department of Radiation OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
9
|
Kong M, Xu B, Zhu G, Wang X, Kuang Z, Sun Q, Liu K, Wang Z, Zhang Y, Li J. Qizhu Yuling prescription in the prevention of postoperative metastasis and recurrence of esophagus cancer: study protocol for a randomized, double-blind, placebo-controlled, multicenter clinical trial. Front Oncol 2025; 15:1478390. [PMID: 40206581 PMCID: PMC11979610 DOI: 10.3389/fonc.2025.1478390] [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: 08/09/2024] [Accepted: 02/28/2025] [Indexed: 04/11/2025] Open
Abstract
Background Esophageal cancer (EC) is a malignant tumor with a high recurrence and metastasis rate and poor prognosis. In 2024, China ranked first in the world in terms of new EC cases and deaths. Surgery is the main treatment method for EC, but the clinical difficulty is how to prevent recurrence and metastasis after surgery. Traditional Chinese medicine as a complementary therapy has played an important role in this regard. Preclinical studies have confirmed that Qizhu Yuling Prescription (QZYLP) has anticancer effects, reduces treatment side effects, and improves quality of life, except for the lack of long-term prognostic results. Therefore, this study aims to investigate whether QZYLP can reduce the recurrence and metastasis rates of EC after surgery, improve disease-free survival (DFS), prolong overall survival, and observe the safety of the drug. Methods This study is a multicenter, randomized, double-blind, placebo-controlled clinical trial. It seeks to enroll 310 patients from 10 hospitals who have completed adjuvant therapy following R0 surgery for esophageal squamous cell carcinoma without recurrent metastasis. Using a center-randomized design, participants will be assigned to the control group (n=155, receiving placebo treatment) or experimental group (n=155, receiving QZYLP granules treatment). Treatment will last for 6 months, with follow-up every 3 months after the final treatment or endpoint event, continuing for up to 3 years postoperatively. The primary outcome measured is DFS at 1 year postoperatively. Secondary outcomes included indicators related to prognosis, fat distribution, peripheral blood inflammation, tumor markers, and quality of life scales. Discussion This study aims to further clarify the efficacy and safety of QZYLP in preventing postoperative recurrence and metastasis of EC, and to explore the mechanism of action. The results of this study will provide high-quality evidence for the participation of TCM in the comprehensive treatment program of EC, and improve the precise diagnosis and treatment system of TCM in EC. Clinical trial registration ClinicalTrials.gov, identifier NCT05626309.
Collapse
Affiliation(s)
- Miao Kong
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bowen Xu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of TCM/Integrative Medicine, Hunan Cancer Hospital, Changsha, China
| | - Guanghui Zhu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinmiao Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ziyu Kuang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qianhui Sun
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Kexin Liu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zilin Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Zhang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Hong H, He Y, Li Y, Shen Y, Qu Y. Charting the future of esophageal cancer translation: insights from clinical trial landscape. Int J Surg 2025; 111:2731-2734. [PMID: 39878068 DOI: 10.1097/js9.0000000000002254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025]
Affiliation(s)
- Hujian Hong
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Yijiang He
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Yan Li
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, China
- Graduate School, China Medical University, Shenyang, China
| | - Yongyan Shen
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, China
- Graduate School, China Medical University, Shenyang, China
| | - Yanli Qu
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang, China
| |
Collapse
|
11
|
Wu Y, Lin C, Qian Y, Huang X, Xu Y, Li J, He Y, Xie C, Su H. Identification of immune subtypes associated with CD8+ T cell-related genes providing new treatment strategies of esophageal carcinoma. Front Immunol 2025; 16:1512230. [PMID: 40083549 PMCID: PMC11903738 DOI: 10.3389/fimmu.2025.1512230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Background CD8+ T lymphocytes greatly affect the efficacy of immunotherapy, displaying promising potential in various tumors. Here, we aimed to identify immune subtypes associated with CD8+ T cell-related genes to predict the efficacy of treatment in esophageal cancer (ESCA). Methods We obtained 13 immune cell-related datasets from the Gene Expression Omnibus (GEO) database and removed batch effects. Weighted correlation network analysis (WGCNA) and co-expression analysis were performed to identify highly correlated CD8+ T cell genes. Cox analysis was used to process ESCA clinical information, and the immune clusters (ICs) were constructed through consensus cluster analysis. Furthermore, we constructed an immune risk score model to predict the prognosis of ESCA based on these CD8+ T cell genes. This model was verified using the IMvigor210 dataset, and we functionally validated the immune risk score model in vitro. Results The results revealed significant correlations between CD8+ T cell-related genes and immune-related pathways. Three ICs were identified in ESCA, with IC3 demonstrating the most favorable prognosis. The final 6-gene prognostic risk model exhibited stable predictive performance in datasets across different platforms. Compared with that in normal esophageal epithelial (HEEC cells), CHMP7 in the 6-gene prognostic risk model was upregulated in KYSE150 and TE-1 cells. Si-CHMP7 transfection led to a decrease in tumor cell migration, invasion, and proliferation, accompanied by an accelerated apoptotic process. Conclusions Collectively, we identified the immune subtypes of CD8+ T cell-related genes with different prognostic significance. We designated CHMP7 in the 6-gene prognostic risk model as a potential target to improve tumor cell prognosis. These insights provide a strong basis for improving prognosis and facilitating more personalized and accurate treatment decisions for the immunotherapy of ESCA.
Collapse
Affiliation(s)
- Youyi Wu
- Department Oncology Radiotherapy, The Third Affiliated Hospital of Wenzhou Medical University, Rui’an People Hospital, Ruian, Zhejiang, China
| | - Chen Lin
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuchen Qian
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaowei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yajing Xu
- Department of Radiation Oncology Wenzhou Central Hospital Theorem Hospital Affiliated of Wenzhou Medical University, Wenzhou, China
| | - Jiayi Li
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youdi He
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huafang Su
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Zhang M, Su Y, Wen P, Shao X, Yang P, An P, Jing W, Liu L, Yang Z, Yang M. Subtype cluster analysis unveiled the correlation between m6A- and cuproptosis-related lncRNAs and the prognosis, immune microenvironment, and treatment sensitivity of esophageal cancer. Front Immunol 2025; 16:1539630. [PMID: 40034693 PMCID: PMC11872909 DOI: 10.3389/fimmu.2025.1539630] [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: 12/04/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Objective Esophageal cancer (EC) is characterized by a high degree of malignancy and poor prognosis. N6-methyladenosine (m6A), a prominent post-transcriptional modification of mRNA in mammalian cells, plays a pivotal role in regulating various cellular and biological processes. Similarly, cuproptosis has garnered attention for its potential implications in cancer biology. This study seeks to elucidate the impact of m6A- and cuproptosis-related long non-coding RNAs (m6aCRLncs) on the prognosis of patients with EC. Methods The EC transcriptional data and corresponding clinical information were retrieved from The Cancer Genome Atlas (TCGA) database, comprising 11 normal samples and 159 EC samples. Data on 23 m6A regulators and 25 cuproptosis-related genes were sourced from the latest literature. The m6aCRLncs linked to EC were identified through co-expression analysis. Differentially expressed m6aCRLncs associated with EC prognosis were screened using the limma package in R and univariate Cox regression analysis. Subtype clustering was performed to classify EC patients, enabling the investigation of differences in clinical outcomes and immune microenvironment across patient clusters. A risk prognostic model was constructed using least absolute shrinkage and selection operator (LASSO) regression. Its robustness was evaluated through survival analysis, risk stratification curves, and receiver operating characteristic (ROC) curves. Additionally, the model's applicability across various clinical features and molecular subtypes of EC patients was assessed. To further explore the model's utility in predicting the immune microenvironment, single-sample gene set enrichment analysis (ssGSEA), immune cell infiltration analysis, and immune checkpoint differential expression analysis were conducted. Drug sensitivity analysis was performed to identify potential therapeutic agents for EC. Finally, the mRNA expression levels of m6aCRLncs in EC cell lines were validated using reverse transcription quantitative polymerase chain reaction (RT-qPCR). Results We developed a prognostic risk model based on five m6aCRLncs, namely ELF3-AS1, HNF1A-AS1, LINC00942, LINC01389, and MIR181A2HG, to predict survival outcomes and characterize the immune microenvironment in EC patients. Analysis of molecular subtypes and clinical features revealed significant differences in cluster distribution, disease stage, and N stage between high- and low-risk groups. Immune profiling further identified distinct immune cell populations and functional pathways associated with risk scores, including positive correlations with naive B cells, resting CD4+ T cells, and plasma cells, and negative correlations with macrophages M0 and M1. Additionally, we identified key immune checkpoint-related genes with significant differential expression between risk groups, including TNFRSF14, TNFSF15, TNFRSF18, LGALS9, CD44, HHLA2, and CD40. Furthermore, nine candidate drugs with potential therapeutic efficacy in EC were identified: Bleomycin, Cisplatin, Cyclopamine, PLX4720, Erlotinib, Gefitinib, RO.3306, XMD8.85, and WH.4.023. Finally, RT-qPCR validation of the mRNA expression levels of m6aCRLncs in EC cell lines demonstrated that ELF3-AS1 expression was significantly upregulated in the EC cell lines KYSE-30 and KYSE-180 compared to normal esophageal epithelial cells. Conclusion This study elucidates the role of m6aCRLncs in shaping the prognostic outcomes and immune microenvironment of EC. Furthermore, it identifies potential therapeutic agents with efficacy against EC. These findings hold significant promise for enhancing the survival of EC patients and provide valuable insights to inform clinical decision-making in the management of this disease.
Collapse
Affiliation(s)
- Ming Zhang
- Department of General Practice, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yani Su
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Pengfei Wen
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xiaolong Shao
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Peng Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Peng An
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Wensen Jing
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Lin Liu
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zhi Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Mingyi Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
13
|
Deng M, Qing Y, Qiu D, Sheng Y, Zhou J, Sun L. The prognostic value of pretreatment neutrophil-lymphocyte ratio and platelet-lymphocyte ratio in patients with esophageal cancer undergoing immunotherapy: a systematic review and meta-analysis. Front Oncol 2025; 15:1536920. [PMID: 40027124 PMCID: PMC11868166 DOI: 10.3389/fonc.2025.1536920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
Background Esophageal cancer (EC) is associated with a high morbidity and mortality rate. Immunotherapy has demonstrated effective antitumor activity in patients with EC, making it imperative to investigate easily accessible prognostic factors. Consequently, we conducted a meta-analysis to explore the prognostic significance of neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) in EC patients treated with immunotherapy. Methods The literature search was conducted across three databases: PubMed, Embase, and Web of Science. The primary deadline for literature retrieval was July 2024. Hazard ratio (HR) with a 95% confidence interval (CI) was utilized to assess the association between NLR or PLR and overall survival (OS) as well as progression-free survival (PFS). Statistical analysis was performed using Review Manager version 5.4 and STATA version 15.0. Results The meta-analysis included a total of 16 studies involving 1,481 patients. The results indicated a significant correlation between high pretreatment NLR and poor PFS (HR=1.76, 95%CI:1.38-2.25, p<0.001) as well as poor OS (HR=2.61,95%CI:1.86-3.67, p<0.001). Subgroup analyses based on tumor stage revealed that the association between elevated NLR and poor PFS was only observed in advanced EC patients. Regarding PLR, an increased PLR was found to be indicative of inferior PFS (HR=1.44, 95%CI: 1.20-1.72, p<0.001) and OS (HR=1.72,95%CI:1.08-2.74, p=0.020). However, the sensitivity analyses suggested that the observed increase in PLR lack robustness in terms of its impact on inferior OS. Conclusion Elevated NLR and PLR are associated with inferior PFS and OS in EC patients receiving immunotherapy. These findings suggest that NLR and PLR levels hold promise as prognostic biomarkers in clinical practice, offering valuable guidance for personalized immunotherapy strategies. Systematic Review Registration PROSPERO https://www.crd.york.ac.uk/prospero/, identifier CRD42024596737.
Collapse
Affiliation(s)
| | | | | | | | | | - Lan Sun
- Department of Oncology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
14
|
Yin R, Zhou G, Liu G, Hou X, Yang H, Ge J, Ying J. Pan-cancer and multi-omics analysis: NDUFA1 is a potential therapeutic target and prognostic marker for esophageal cancer. Cell Biol Toxicol 2025; 41:43. [PMID: 39937347 PMCID: PMC11821742 DOI: 10.1007/s10565-025-09993-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/28/2025] [Indexed: 02/13/2025]
Abstract
OBJECTIVES The study examined the NDUFA1 expression and its prognostic value in pan-cancer, especially in esophageal cancer (ESCA). Its carcinogenic effect on ESCA was further elucidated. MATERIALS AND METHODS TCGA database was used to examine NDUFA1 expression and its prognostic value in 33 cancer types. GO and KEGG were performed for function and pathway enrichment of NDUFA1-related genes. The carcinogenic effect on ESCA was verified in both KYSE-30 cell and Xenograft mouse model. RESULTS Abnormal high expression of NDUFA1 was detected in pan-cancer, and related to immune cell infiltration. TCGA database indicated an elevated value of NDUFA1 in ESCA tumor tissues, which was linked to patients' poor prognosis. NDUFA1-related genes were mainly enriched in oxidative stress and immune response in ESCA. NDUFA1 knockdown significantly suppressed ESCA cell proliferation, migration and invasion. Similarly, tumor growth of ESCA xenograft mice was inhibited by NDUFA1 knockdown. Activated PI3K-Akt signaling was detected in both ESCA cell lines and tumor tissues, which was reversed by NDUFA1 knockdown. CONCLUSION Multi-omics analysis showed that NDUFA1 might be adopted as a potential therapeutic goal and prognostic indicator for a variety of cancers, especially for ESCA. NDUFA1 knockdown inhibited ESCA tumor growth, which may have the participation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Rui Yin
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China
| | - Gai Zhou
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China
| | - Guanqi Liu
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China
| | - Xiaoting Hou
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China
| | - Haifeng Yang
- Department of Infectious Disease, Economic Development Zone, People's Hospital of Xuyi, No. 28, Hongwu Avenue, Xuyi, 211700, China.
| | - Jianxin Ge
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China.
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China.
| | - Jie Ying
- Department of Gastroenterology, Nanjing Jiangbei Hospital, Nanjing, 210044, China.
- Department of Gastroenterology, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, No. 552, Geguan Road, Jiangbei New District, Nanjing, 210044, China.
| |
Collapse
|
15
|
Liu D, Liu A, Guo L, Li Y, Li Y, Chi Y, Lin H, Yu J, Li M. Postoperative Adjuvant Therapy Benefits Non-pCR Patients Rather Than pCR Patients for Locally Advanced ESCC: A Multicenter Real-World Study. Thorac Cancer 2025; 16:e70021. [PMID: 39988453 PMCID: PMC11847616 DOI: 10.1111/1759-7714.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/04/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND There is no unified standard in adjuvant therapy (AT) for patients with esophageal squamous cell carcinoma (ESCC) after neoadjuvant therapy and surgery. We evaluated the significance of AT for these patients and explored its influencing factors. METHODS ESCC patients who underwent neoadjuvant therapy and surgery from 2019 to 2022 at three centers were divided into AT (n = 227) and non-AT groups (n = 435). Baseline characteristics were balanced using propensity score matching (PSM). Primary endpoints were disease-free survival (DFS) and overall survival (OS), assessed using the Kaplan-Meier method. Subgroup analyses and univariate and multivariate Cox regression analyses were conducted to identify the prognostic factors. RESULTS The median follow-up period is 36 (2-72) months. After PSM, the total population had 1-, 2-, and 3-year OS rates of 71.3%, 66.0%, and 64.1%, respectively. There were no statistically significant differences in DFS (HR: 0.79; 95% CI: 0.55-1.14, p = 0.21) or OS (HR: 0.75; 95% CI: 0.49-1.13, p = 0.17) between AT and non-AT groups. Subgroup analysis revealed that non-pCR patients benefited from AT in DFS (p = 0.042) and OS (p = 0.033). Moreover, in non-pCR patients who received AT, BMI ≥ 21.5 kg/m2 and ypN0 were independent protective factors of DFS. ypN0 was an independent protective factor of OS. In terms of AT regimens, the Kaplan-Meier analysis revealed that adjuvant immunochemotherapy (AICT) provided superior survival benefits than adjuvant radiotherapy and adjuvant chemotherapy. CONCLUSIONS Postoperative AT benefited ESCC patients with non-pCR, while AICT may be a relatively better AT regimen in real-world data, which deserves further exploration.
Collapse
Affiliation(s)
- Defeng Liu
- Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Ao Liu
- Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Longxiang Guo
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
- Department of OncologyDongying People's HospitalDongyingChina
| | - Yi Li
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Yuanlin Li
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Yuxiang Chi
- Cheeloo College of MedicineShandong UniversityJinanChina
- Institute of Oncology, Shandong Provincial HospitalShandong UniversityJinanChina
| | - Haiqun Lin
- Department of Radiation OncologyThe Second Hospital of Shandong UniversityJinanChina
| | - Jinming Yu
- Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Minghuan Li
- Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| |
Collapse
|
16
|
Wang R, Liu X, Cai H, Li B, Li Y. A nomogram to predict long-term survival after resection for esophageal cancer: An observational study in northeast China. Surgery 2025; 178:108968. [PMID: 39689614 DOI: 10.1016/j.surg.2024.108968] [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: 10/26/2023] [Revised: 10/08/2024] [Accepted: 11/13/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVE This study aims to create a prognostic nomogram by combining clinicopathologic variables that are linked to the overall survival following the surgical removal of esophageal squamous cell carcinoma. METHODS A total of 224 patients with esophageal cancer who underwent surgical R0 resection were included. The construction of the nomogram involved using a multivariable Cox proportional hazards regression model. To evaluate the model's effectiveness, Kaplan-Meier curves and calibration plots were used for discrimination and calibration, respectively. RESULTS Nearly half of the patients were >60 years old (45.1%), and 95.5% of the patients were male. After esophageal cancer resection, 35.7% of patients experienced complications, with 23.7% developing anastomotic stenosis and 4.5% developing a fistula. Using the backward selection of clinically relevant variables, we found that tumor located in middle thoracic (hazard ratio 2.299, 95% confidence interval 1.008-5.244), anastomotic fistula (3.028, 1.436-6.384), and vascular invasion (2.175, 1.496-3.108) were independently associated with mortality (all P < .05), whereas lymph node clearance ≥15 nodes is associated with longer survival (0.444, 0.278-0.710) (P = .001). On the basis of these factors, a nomogram was created to predict survival of esophageal squamous cell carcinoma after resection. Discrimination using Kaplan-Meier curves, calibration curves, and bootstrap cross-validation revealed good predictive abilities (C index, 0.673). CONCLUSIONS A nomogram was created based on the experience from northeast China to forecast overall survival following resection for esophageal squamous cell carcinoma. The validation process demonstrated accurate distinction and calibration, indicating the practical value of the nomogram in enhancing personalized survival predictions for patients who undergo esophageal squamous cell carcinoma resection in this study population.
Collapse
Affiliation(s)
- Rui Wang
- Department of Thoracic Surgery, Organ Transplantation Center, First Hospital of Jilin University, Changchun, China
| | - Xin Liu
- Department of Thoracic Surgery, Organ Transplantation Center, First Hospital of Jilin University, Changchun, China
| | - Hongfei Cai
- Department of Thoracic Surgery, Organ Transplantation Center, First Hospital of Jilin University, Changchun, China
| | - Bo Li
- School of Public Health, Jilin University, Changchun, China
| | - Yang Li
- Department of Thoracic Surgery, Organ Transplantation Center, First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
17
|
Li W, Wang W. Association between radiotherapy and risk of secondary primary malignancies in patients with esophageal cancer: A population-based study utilizing 17 Surveillance, Epidemiology, and End Results cancer registries. Cancer Radiother 2025; 29:104587. [PMID: 40049061 DOI: 10.1016/j.canrad.2024.07.020] [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: 03/21/2024] [Revised: 05/18/2024] [Accepted: 07/17/2024] [Indexed: 04/01/2025]
Abstract
PURPOSE Despite the frequent use of radiotherapy as a standard treatment for esophageal cancer, the potential risk of developing second primary malignancies as a result of radiation-related side effects remains underinvestigated. METHODS We conducted a comprehensive analysis using data from the large cohort of 17 cancer registries within the Surveillance, Epidemiology, and End Results (SEER) database. To estimate the incidence of second primary malignancies associated with radiotherapy, we employed Poisson regression and competing risk regression models. RESULTS A total of 54,844 patients diagnosed with esophageal cancer were identified from the SEER database. Among them, 8351 patients did not receive radiotherapy, while 15,555 patients were treated with radiation. The incidence of second primary malignancies among patients who had radiotherapy was 6.28 % (977 patients) compared to an incidence of 7.09 % (592 patients) for those who had not (P<0.0001). After adjusting for potential confounders, such as age, gender, year of diagnosis, tumor grade, and chemotherapy, the incidence rates of patients who received radiotherapy were significantly higher compared to those who had not (odds ratio [OR] Poissonregression=1.18, 95 % confidence interval [CI]: 1.05-1.32, P=0.018; OR multivariablecompetingriskregression=1.15, 95 %CI: 1.04-1.26, P=0.018). Notably, patients who received radiotherapy were found to be at an increased risk of developing lung cancer, breast cancer, and gastric cancer. Additionally, younger patients with esophageal cancer appeared to be more susceptible, with the highest risk of developing subsequent primary malignancies observed more than 10 years after the esophageal cancer diagnosis. CONCLUSION Our findings indicated that radiotherapy for esophageal cancer was associated with an increased risk of developing overall cancer. It is crucial to address and mitigate the risk of second primary malignancies associated with radiotherapy, while also improving the prognosis for patients affected by these secondary malignancies.
Collapse
Affiliation(s)
- Wenjie Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Baiyun District, Guangzhou, Guangdong 510515, China.
| | - Wei Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Baiyun District, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
18
|
Li J, Wu C, Song Y, Fan Y, Li C, Li H, Zhang S. Exploring the Clinical Value of Perioperative ctDNA-Based Detection of Molecular Residual Disease in Patients With Esophageal Squamous Cell Carcinoma. Thorac Cancer 2025; 16:e70017. [PMID: 39966084 PMCID: PMC11835505 DOI: 10.1111/1759-7714.70017] [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: 11/12/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE To explore the clinical value of molecular residual disease detection based on circulating tumor DNA (ctDNA-MRD) in the perioperative period of esophageal squamous cell carcinoma (ESCC) and to analyze the tumor escape mechanisms in MRD-positive cases. METHODS A total of 35 ESCC patients were prospectively enrolled. Preoperative and postoperative (1 month after surgery) blood and surgical tissue samples were analyzed. ctDNA variants were tracked in plasma to assess ctDNA-MRD, and whole-transcriptome sequencing was performed on MRD-positive and MRD-negative tissue samples. RESULTS Preoperative blood ctDNA was positive in 54.3% of patients, with a 31.6% positive predictive value for recurrence. One month postsurgery, the positive rate of ctDNA was 17.1%, with an 83.3% predictive value for recurrence. Both preoperative and postoperative ctDNA positivity were significant prognostic indicators (HR = 2.78, p < 0.05; HR = 4.42, p < 0.001). Multivariate analysis confirmed ctDNA as an independent prognostic factor (HR = 303.75, p < 0.001). Transcriptomic analysis revealed increased macrophage (W = 15 848; p < 0.01) and follicular helper T (Tfh) cell (W = 10 935; p < 0.01) levels in MRD-positive patients, suggesting a potential link to immune escape in tumors. CONCLUSIONS Plasma ctDNA measured 1 month postoperatively in ESCC patients can effectively detect MRD, and ctDNA-MRD serves as an independent risk factor for postoperative recurrence. The mechanism underlying MRD positivity may involve the polarization of Tfh cells and macrophages, aiding tumor cells in immune escape through the bloodstream.
Collapse
Affiliation(s)
- Jimin Li
- Department of Thoracic SurgeryCancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical SciencesTaiyuanChina
| | - Congcong Wu
- Department of Thoracic SurgeryCancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical SciencesTaiyuanChina
| | - Yongming Song
- Department of Thoracic SurgeryShanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Yuhui Fan
- Department of Thoracic SurgeryShanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Chao Li
- Department of Thoracic SurgeryShanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Haibo Li
- Department of Thoracic SurgeryJincheng Second People's HospitalJinchengChina
| | - Shuangping Zhang
- Department of Thoracic SurgeryCancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical SciencesTaiyuanChina
- Department of Thoracic SurgeryYuncheng Central HospitalYunchengChina
| |
Collapse
|
19
|
Lv H, Zhou J, Qiu L, Tang X, Huang C. AURKB and circAURKB_288aa enhance Esophageal cancer drug resistance through inducing abnormal centrosome separation. Biochem Pharmacol 2025; 232:116691. [PMID: 39638069 DOI: 10.1016/j.bcp.2024.116691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Esophageal cancer (EC) is one of the most fatal malignancies worldwide, with a dramatic increase in incidence in the western world occurring over the past few decades. Chromosome instability (CIN) is a major contributor to EC progression, drug resistance, relapse, and the development of intratumoral heterogeneity. This study revealed a striking elevation of AURKB expression in EC patients, with a strong correlation to poor clinical outcomes. AURKB overexpression promoted cellular proliferation and induced drug resistance in both cell culture and animal models. Conversely, genetic targeting of AURKB abrogated these effects. Mechanistically, enforced AURKB expression triggered CIN, a key driver of poor EC outcomes, primarily through CEP250 phosphorylation. Interestingly, we identified a novel circular form of AURKB (circAURKB_288aa) harboring the AURKB kinase domain and encoding a 288-amino acid protein. Elevated levels of circAURKB_288aa in EC peripheral blood samples mirrored poor patient outcomes and synergistically enhanced CIN alongside AURKB. Furthermore, EC cells were capable of secreting circAURKB_288aa, influencing tumor microenvironmental cells similarly to full-length AURKB protein. Notably, AURKB siRNA targeting the shared kinase domain of both AURKB and circAURKB_288aa significantly inhibited EC malignancy. Collectively, these findings establish AURKB and circAURKB_288aa as promising targets for EC prognosis and therapy.
Collapse
Affiliation(s)
- Hongzhen Lv
- School of Basic Medical Sciences, Jiangsu Medical College, Yancheng, China
| | - Jing Zhou
- General Medicine Department, Yancheng Third People's Hospital, Yancheng, China
| | - Limin Qiu
- Thoracic Surgery Department, Yancheng NO.1 People's Hospital, Yancheng, China
| | - Xiaozhu Tang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Cheng Huang
- School of Traditional Chinese Medicine, Jiangsu Medical College, Yancheng, China.
| |
Collapse
|
20
|
Qu H, Yu Q, Ye L, Zheng J. SLC39A14 promotes the development of esophageal squamous cell carcinoma through PI3K/Akt/mTOR signaling pathway. Int Immunopharmacol 2025; 146:113831. [PMID: 39700956 DOI: 10.1016/j.intimp.2024.113831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/25/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVES This study aims to investigate the expression of solute carrier family 39 member 14 (SLC39A14) in esophageal squamous cell carcinoma (ESCC) tissues and its prognosis, as well as the impact of SLC39A14 expression on the biological behavior of ESCC cells and associated mechanisms. METHODS Bioinformatics analysis was utilized to compare the differential expression of SLC39A14 mRNA between esophageal cancer tissues and adjacent non-cancerous tissues. Immunohistochemistry was employed to evaluate SLC39A14 protein expression in human ESCC tissues and normal esophageal tissues, followed by an analysis of its association with clinicopathological parameters in esophageal cancer patients. Through cell proliferation, migration, invasion, and Western blot assays, we deeply evaluated the specific effects of SLC39A14 gene knockdown (or overexpression) on ESCC cells and explored its potential biological functions in ESCC. Subsequently, we validated the role of SLC39A14 in ESCC in a xenograft model. Furthermore, LY294002 drug intervention was used to verify the regulatory effect of SLC39A14 on PI3K/Akt/mTOR signaling pathway. RESULTS Both mRNA and protein levels of SLC39A14 were significantly elevated in tumor tissues from ESCC patients compared to adjacent normal tissues. Notably, higher levels of SLC39A14 expression positively correlated with ESCC tumor size (p = 0.010) and clinical T stage (p = 0.025), while exhibiting a negative correlation with overall patient survival rates (p = 0.023). In vitro experiments demonstrated that knocking down SLC39A14 significantly inhibited cell proliferation, migration and invasion. In vivo study showed that SLC39A14 facilitated progression within murine models bearing ESCC tumors. Mechanistic analyses suggested that pro-carcinogenic effects exerted by SLC39A14 are mediated through activation of the PI3K/Akt/mTOR signaling pathway. CONCLUSIONS Our findings suggest that SLC39A14 may serve as a potential biomarker for ESCC due to its pro-oncogenic role during ESCC progression.
Collapse
Affiliation(s)
- Hangshuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Qingxin Yu
- Department of pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province, China
| | - Luxia Ye
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Jingmin Zheng
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China.
| |
Collapse
|
21
|
Yan G, Li J, Su Y, Li G, Feng G, Liu J, Gao X, Zhou H. Risk factors analysis of hypokalemia after radical resection of esophageal cancer and establishment of a nomogram risk prediction model. Front Surg 2025; 11:1433751. [PMID: 39840263 PMCID: PMC11747289 DOI: 10.3389/fsurg.2024.1433751] [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: 05/20/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
Objective This study aimed to explore the risk factors of hypokalemia after radical resection of esophageal cancer (EC) and establish a nomogram risk prediction model to evaluate hypokalemia risk after esophagectomy. Thus, this study provides a reference for the clinical development of intervention measures. Methods Clinical data of EC patients who underwent radical surgery from January 2020 to November 2022 in the First Affiliated Hospital of Guangxi Medical University were retrospectively collected. The relevant variables were screened using multivariate logistic regression analysis with IBM SPSS 25.0 and R 4.2.0 software, and a nomogram for predicting hypokalemia risk was established. The established nomogram was evaluated by receiver operating characteristic (ROC), calibration, and decision curves. The model was also internally validated by 1000 bootstrap resampling methods. Results After radical EC resection, the incidence rate of hypokalemia in 213 patients was 19.2% (41/213). The hemoglobin levels, total serum protein, serum albumin, calcium ion concentration, direct bilirubin, prothrombin time (PT), and activated partial thromboplastin time (APTT) were related (p < 0.05). The multivariate logistic analysis showed that the white blood cell count, serum albumin level, direct bilirubin, and operation time were risk factors for hypokalemia after radical EC resection (p < 0.05). The area under the ROC curve (AUC) was 0.764, demonstrating the good discriminative ability of the established nomogram for hypokalemia prediction. The calibration curve showed a good fit between the predicted and actual observed probabilities. The model maintained a high C-index in the internal validation (C-index = 0.758), supporting that the nomogram can be widely used for hypokalemia prediction. Conclusion The prediction model for hypokalemia risk with individualized scores based on the patient's white blood cell count, serum albumin level, direct bilirubin, and operation time can screen out high-risk patients who might develop hypokalemia. It is of certain reference value for clinicians to screen and follow up with patients with emphasis and to formulate preoperative and postoperative intervention strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Huafu Zhou
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
22
|
Ye C, Xu C, Tang Y, Qi Y, Peng X, Wei G, Jiang L. A novel disulfidptosis-related LncRNA prognostic risk model: predicts the prognosis, tumor microenvironment and drug sensitivity in esophageal squamous cell carcinoma. BMC Gastroenterol 2024; 24:437. [PMID: 39604874 PMCID: PMC11603746 DOI: 10.1186/s12876-024-03530-2] [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: 10/07/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Disulfidptosis is a newly discovered type of cell death that differs from apoptosis, necrosis, ferroptosis and other death modes and is closely related to the occurrence and progression of tumors. However, the predictive potential and biological characteristics of disulfidptosis-related lncRNAs (DRGs-lncRNAs) in esophageal squamous cell carcinoma (ESCC) are unclear. METHODS RNA transcriptome data, clinical information and mutation data for ESCC patients were obtained from The Cancer Genome Atlas (TCGA) database. Pearson correlation and Cox regression analyses were used to identify the DRGs-lncRNAs associated with overall survival (OS). LASSO regression analysis was used to construct the prognostic model. A nomogram was created to predict the prognosis of patients with ESCC. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) were used to identify the signaling pathways associated with the model. TIMER, CIBERSORT, ESTIMATE and other methods were used to analyze immune infiltration, immune function, immune checkpoints and drug sensitivity. The tumor mutation burden (TMB) were assessed between different risk groups. Real-time polymerase chain reaction (RT‒PCR) was used to detect the expression of DRGs-lncRNAs in ESCC cell lines. RESULTS A total of 155 lncRNAs significantly associated with disulfidptosis were identified. Through univariate Cox regression analysis, LASSO regression analysis and multivariate Cox regression analysis, 9 lncRNAs with independent prognostic significance were selected, and a prognosis model was established. Survival analysis with the prognostic model revealed that there were obvious differences in survival between the high- and low-risk groups. Further analysis revealed that the immune microenvironment, immune infiltration, immune function, immune checkpoints, and drug sensitivity significantly differed between the high-risk and low-risk groups. Patients who exhibited both high risk and high tumor mutation burden (TMB) survived shorter, while those who fell into the low risk and low TMB categories survived longer. In addition, RT‒PCR analysis revealed differential expression of DRG lncRNAs between ESCC cell lines and esophageal epithelial cell lines. CONCLUSIONS We established a DRG-lncRNA prognostic model that can be used to predict the prognosis, tumor mutation burden, immune cell infiltration, and drug sensitivity of ECSS patients. The results of this study provide valuable insights into the understanding of ESCC and provide valuable assistance for the individualized treatment of ESCC patients.
Collapse
Affiliation(s)
- Chunlin Ye
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Chuan Xu
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Yongchao Tang
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Yingcheng Qi
- Department of Gastroenterological Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Xiaoyue Peng
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China
| | - Guangxia Wei
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
| | - Lei Jiang
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, Jiangxi, 330000, People's Republic of China.
| |
Collapse
|
23
|
Yang C, Wei W, Hu F, Zhao X, Yang H, Song X, Sun Z. Dihydroartemisinin suppresses the tumorigenesis of esophageal carcinoma by elevating DAB2IP expression in a NFIC-dependent manner. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8117-8128. [PMID: 38789636 DOI: 10.1007/s00210-024-03163-y] [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: 04/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Dihydroartemisinin (DHA) has been identified to have the anticancer and anti-inflammatory activities. Disabled homolog 2 interacting protein (DAB2IP) is a well-recognized tumor suppressor. Both DHA and DAB2IP were proven to have suppressing effects on esophageal carcinoma (ESCA) tumorigenesis. However, whether DHA regulated ESCA cells via DAB2IP and its mechanism are still vague. Functional analyses were conducted using MTT, tube formation, sphere formation, and transwell assays in vitro as well as Tumor formation experiments in mice. Levels of genes and proteins were assayed by qRT-PCR and western blotting analyses. The interaction between DAB2IP and Nuclear Factor I C (NFIC) was confirmed using bioinformatics analysis and dual-luciferase reporter assay. DHA treatment suppressed ESCA cell angiogenesis, stemmess, migration, and invasion. DAB2IP level was decreased in ESCA tissues and cells, and DHA elevated DAB2IP expression in ESCA cells. Functionally, DAB2IP overexpression impaired ESCA cell angiogenesis, stemmess, migration and invasion. Mechanistically, NFIC had binding sites on the promoter region and directly targeted DAB2IP. DHA could up-regulate DAB2IP expression via NFIC. Moreover, NFIC was also decreased in ESCA tissues and cells, and its overexpression had anticancer activity in ESCA cells. In addition, DAB2IP knockdown reversed the anticancer effects of NFIC or DHA on ESCA cells. In further in vivo analysis, DHA also suppressed ESCA growth by regulating DAB2IP expression. DHA suppressed the tumorigenesis of ESCA by elevating DAB2IP expression in an NFIC-dependent manner, suggesting the potential clinical application of DHA in ESCA treatment.
Collapse
Affiliation(s)
- Chao Yang
- Department of Thoracic Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, Hubei Province, People's Republic of China
| | - Wei Wei
- Department of Thoracic Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, Hubei Province, People's Republic of China
| | - Fen Hu
- Department of Thoracic Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, Hubei Province, People's Republic of China
| | - Xing Zhao
- Department of Thoracic Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, Hubei Province, People's Republic of China
| | - Hanxue Yang
- Department of Thoracic Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, Hubei Province, People's Republic of China
| | - Xiujun Song
- Department of Thoracic Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, Hubei Province, People's Republic of China
| | - Zhihua Sun
- Department of Thoracic Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, Hubei Province, People's Republic of China.
| |
Collapse
|
24
|
Zhu X, Li M, Gan H, Guo Y. Causal association between hyperthyroidism and risk of gastroesophageal reflux or esophageal cancer: a bidirectional Mendelian randomization investigation. Front Endocrinol (Lausanne) 2024; 15:1411629. [PMID: 39355614 PMCID: PMC11442246 DOI: 10.3389/fendo.2024.1411629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024] Open
Abstract
Background Emerging observational studies indicated an association between hyperthyroidism and gastrointestinal disorders. However, it remains unclear whether this association is causal, particularly in the case of gastroesophageal reflux (GERD) and esophageal cancer. Methods To assess the potential causal relationship between hyperthyroidism and GERD or esophageal cancer, we conducted a bidirectional 2-sample Mendelian randomization study. Independent genetic instruments for hyperthyroidism from the UK Biobank (N case=3,545 and N control=459,388) and public genome-wide association study (GWAS) dataset (N case=3,731 and N control=480,867) were used to investigate the association with esophageal cancer in the UK Biobank study (N case=740 and N control=372,016) and GERD in the public GWAS database (N case=20,381 and N control=464,217). Four different approaches (inverse variance weighted (IVW), weighted mode, MR-Egger, and weighted median regression) were used to ensure that our results more reliable. Additional sensitivity analyses were also performed to validate our results. Results When hyperthyroidism was considered as the exposure factor, it appeared to act as a protective factor for GERD (ORIVW = 0.88, 95% CI, 0.79-0.99, P = 0.039), while as a risk factor for esophageal cancer (ORIVW = 1.03, 95% CI, 1.01-1.06, P = 0.003). However, there is no evidence supporting a reverse causal relationship between genetic susceptibility to hyperthyroidism and GERD or esophageal cancer. Conclusion Our findings provided genetic evidence supporting bidirectional causal relationships between hyperthyroidism and GERD or esophageal cancer. These results substantiate certain discoveries from previous observational studies on a causal level and provide insight into relevant genetic susceptibility factors.
Collapse
Affiliation(s)
- Xingyu Zhu
- Department of Cardiovascular Surgery, West China School of Medicine, West China
Hospital, Sichuan University, Chengdu, China
- Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan
University, Chengdu, China
| | - Ming Li
- Department of Cardiovascular Surgery, West China School of Medicine, West China
Hospital, Sichuan University, Chengdu, China
- Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan
University, Chengdu, China
| | - Hanghang Gan
- Department of Cardiovascular Surgery, West China School of Medicine, West China
Hospital, Sichuan University, Chengdu, China
- Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan
University, Chengdu, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China School of Medicine, West China
Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Fang P, Zhou J, Liu Y, Liang Z, Yang Y, Luan S, Xiao X, Li X, Zhang H, Shang Q, Chen L, Zeng X, Yuan Y. Substitute or coexistence? Mediastinoscopy-assisted versus thoracoscope-assisted esophagectomy in esophageal cancer: a meta-analysis of perioperative outcomes and long-term survival. Int J Surg 2024; 110:5802-5817. [PMID: 38869981 PMCID: PMC11392080 DOI: 10.1097/js9.0000000000001777] [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: 03/30/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Currently, mediastinoscopy-assisted esophagectomy (MAE) and thoracoscope-assisted esophagectomy (TAE) represent two prevalent forms of minimally invasive esophagectomy extensively employed in the management of esophageal cancer (EC). The aim of this meta-analysis is to assess and compare these two surgical approaches concerning perioperative outcomes and long-term survival, offering valuable insights for refining surgical strategies and enhancing patient outcomes in this field. METHODS Adhering to PRISMA guidelines, the authors systematically searched PubMed, Web of Science, Cochrane Library, Embase, and CNKI databases until 1 March 2024, for studies comparing MAE and TAE. Outcomes of interest included perioperative outcomes (intraoperative outcomes, postoperative recovery, postoperative complications) and survival rates. Statistical analyses were performed using RevMan 5.4, with heterogeneity dictating the use of fixed or random-effects models. RESULTS A total of 21 relevant studies were finally included. MAE was associated with significantly shorter operation times [mean difference (MD)=-59.58 min, 95% CI: -82.90 to -36.26] and less intraoperative blood loss (MD=-68.34 ml, 95% CI: -130.45 to -6.23). However, MAE resulted in fewer lymph nodes being dissected (MD=-3.50, 95% CI: -6.23 to -0.78). Postoperative recovery was enhanced following MAE, as evidenced by reduced hospital stays and tube times. MAE significantly reduced pulmonary complications [odds ratio (OR)=0.59, 95% CI: 0.44, 0.81] but increased the incidence of recurrent laryngeal nerve injury (OR=1.84, 95% CI: 1.30, 2.60). No significant differences were observed in anastomotic leakage, chylothorax, cardiac complications, wound infections, and gastric retention between MAE and TAE. The long-term survival outcomes showed no statistical difference [hazard ratio (HR)=1.05, 95% CI: 0.71, 1.54]. CONCLUSIONS MAE offers advantages in reducing operation time, blood loss, and specific postoperative complications, particularly pulmonary complications, with a shorter recovery period compared to TAE. However, it poses a higher risk of recurrent laryngeal nerve injury and results in fewer lymph nodes being dissected. No difference in long-term survival was observed, indicating that both techniques have distinct benefits and limitations. These findings underscore the need for personalized surgical approaches in EC treatment, considering individual patient characteristics and tumor specifics.
Collapse
Affiliation(s)
- Pinhao Fang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Jianfeng Zhou
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Yixin Liu
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Zhiwen Liang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Yushang Yang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Siyuan Luan
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Xin Xiao
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Xiaokun Li
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Hanlu Zhang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Qixin Shang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Longqi Chen
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Yuan
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University
| |
Collapse
|
26
|
Meredith LT, Grenda TR. Putting Wind in the Sails of Neoadjuvant Therapy for Locally Advanced Esophageal Adenocarcinoma: Perioperative Outcomes. Ann Surg Oncol 2024; 31:5495-5496. [PMID: 38858293 DOI: 10.1245/s10434-024-15556-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/12/2024]
Affiliation(s)
- Luke T Meredith
- Department of Surgery, Sidney Kimmel Medical College, Philadelphia, PA, USA
| | - Tyler R Grenda
- Division of Thoracic Surgery, Department of Surgery, Sidney Kimmel Medical College, Philadelphia, PA, USA.
| |
Collapse
|
27
|
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
|
28
|
Shibata R, Konishi H, Arita T, Yamamoto Y, Matsuda H, Yamamoto T, Ohashi T, Shimizu H, Komatsu S, Shiozaki A, Kubota T, Fujiwara H, Otsuji E. Extracellular glypican-1 affects tumor progression and prognosis in esophageal cancer. Cancer Med 2024; 13:e70212. [PMID: 39300946 PMCID: PMC11413415 DOI: 10.1002/cam4.70212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/03/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
INTRODUCTION Cells are covered with a glycan surface layer that is referred to as the glycocalyx (GCX). It has been reported that the formation of the GCX is promoted on cancer cells and is associated with tumor growth and metastasis. Heparan sulfate proteoglycan glypican-1 (GPC1) is a core protein of the GCX that is overexpressed in esophageal squamous cell carcinoma (ESCC) and is involved in the development and progression of cancer cells. The purpose of the present study is to analyze the utility of GPC1 as a new biomarker ralated to glycocalyx that reflects therapeutic effect and prognosis of ESCC. METHODS We measured the concentration of GPC1 protein in preoperative plasma from advanced esophageal cancer patients and examined its relationships with clinicopathological factors and therapeutic efficacy, and the effects of extracellular GPC1 were investigated. RESULTS The following clinical factors were significantly correlated with the preoperative high GPC1 concentration: male, tumor size ≥30 mm, venous invasion, pT factor ≥2, pStage ≥3, residual tumor, and distant metastatic recurrence. Both overall and recurrence-free survival were significantly worse in the high GPC1 group. Extracellular GPC1 protein concentration reflected intracellular GPC1 expression. Furthermore, we examined the effects of extracellular recombinant human (rh)GPC1 on ESCC cells, and found that extracellular rhGPC1 affects cell motility, including migration and invasion. CONCLUSIONS These results demonstrated the utility of extracellular GPC1 as a biomarker, which can be assayed from a less invasive blood sample-based liquid biopsy. Extracellular GPC1 protein plays a role in both tumor cell motility and cancer progression. Thus, plasma GPC1 is a useful biomarker for esophageal cancer progression and may be a potential candidate of therapeutic target.
Collapse
Affiliation(s)
- Rie Shibata
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Yusuke Yamamoto
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Hayato Matsuda
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Taiga Yamamoto
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of SurgeryKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
29
|
Ke J, Xie Y, Huang S, Wang W, Zhao Z, Lin W. Comparison of esophageal cancer survival after neoadjuvant chemoradiotherapy plus surgery versus definitive chemoradiotherapy: A systematic review and meta-analysis. Asian J Surg 2024; 47:3827-3840. [PMID: 38448293 DOI: 10.1016/j.asjsur.2024.02.099] [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: 08/10/2023] [Revised: 12/31/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Surgery after neoadjuvant chemoradiotherapy remains the gold standard for the treatment of resectable esophageal cancer (EC); however, chemoradiotherapy without surgery has been recommended in specific cases. The aim of this meta-analysis is to analyse the survival between surgeries after neoadjuvant chemoradiotherapy compared with definitive chemoradiotherapy in order to provide a theoretical basis for clinically individualised differential treatment. We conducted an initial search of MEDLINE (PubMed), the Cochrane Library, and Embase for English-only articles that compared treatment regimens and provided survival data. According to the final I2 value of the two survival indicators, the random effect model or fixed effect model was used to calculate the overall hazard ratio (HR) and 95% confidence intervals (CI). Cochrane's Q test was used to judge the heterogeneity of the studies, and a funnel plot was used to evaluate for publication bias. A sensitivity analysis was performed to verify the stability of the included studies. A total of 38 studies involving 29161 patients (neoadjuvant therapy: 15401, definitive chemoradiotherapy: 13760) were included in the analysis. The final pooled results (HR = 0.74, 95% CI: 0.67-0.82) showed a statistically significant increase in overall survival with neoadjuvant chemoradiotherapy plus surgery compared with definitive chemoradiotherapy. Subgroup analyses were performed to determine the effects of heterogeneity, additional treatment regimens, study types, and geographic regions, as well as histologic differences, complications, and recurrence, on the overall results. For people with esophageal cancer that can be removed, neoadjuvant chemoradiotherapy combined with surgery improves survival compared to definitive chemoradiotherapy. However, more research is needed to confirm these results and help doctors make decisions about treatment.
Collapse
Affiliation(s)
- Junli Ke
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China
| | - Yujie Xie
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China
| | - Shenyang Huang
- Department of Cardiothoracic Surgery, Guangdong Medical University, Zhanjiang, China
| | - Wei Wang
- Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Zhengang Zhao
- Department of Cardiothoracic Surgery, Guangdong Medical University, Zhanjiang, China
| | - Wanli Lin
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China.
| |
Collapse
|
30
|
Wu Y, Liu X, Li H, Wang W, Ye L, Zhou Y, Chen D. D-dimer levels predict the treatment efficacy and prognosis of esophageal squamous cell carcinoma treated with PD-1/PD-L1 inhibitors. Int J Biol Markers 2024; 39:209-216. [PMID: 38887052 DOI: 10.1177/03936155241262045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
OBJECTIVES This study aimed to explore the value of D-dimer levels in predicting the treatment efficacy and prognosis of advanced esophageal squamous cell carcinoma (ESCC) treated with programmed cell death protein-1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors. METHODS The study retrospectively analyzed 233 ESCC patients who received PD-1/PD-L1 inhibitors. The optimal cut-off values for platelets, fibrinogen, and D-dimer were calculated based on maximally selected rank statistics for patients' overall survival. Univariate and multivariate analyses of progression-free survival and overall survival were conducted by Cox proportional hazards regression model. Subgroup analyses of D-dimer levels in different fibrinogen levels were performed by log-rank test. RESULTS The multivariate Cox regression analyses demonstrated that ESCC patients with D-dimer levels > 236 ng/mL exhibited both poorer progression-free survival (P = 0.004) and overall survival (P < 0.0001) compared to those with low D-dimer levels. The subgroup analyses further indicated that in the group of low fibrinogen levels, the higher D-dimer levels of ESCC patients exhibited significantly shorter progression-free survival (P = 0.0021) and overall survival (P < 0.0001). CONCLUSIONS The study revealed that the D-dimer levels possess predictive value for the treatment efficacy and prognosis of ESCC patients treated with PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Yuchen Wu
- Department of Laboratory Medicine, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xin Liu
- Department of Respiratory, Yichun Central Hospital, Yichun, China
| | - Huihui Li
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, China
| | - Wenjing Wang
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, China
| | - Lisha Ye
- Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou, China
| | - Yun Zhou
- Department of Laboratory Medicine, Zhejiang Cancer Hospital, Hangzhou, China
| | - Da Chen
- Department of Oncological Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
31
|
Zhang J, Liu X, Zeng L, Hu Y. GABRP inhibits the progression of oesophageal cancer by regulating CFTR: Integrating bioinformatics analysis and experimental validation. Int J Exp Pathol 2024; 105:118-132. [PMID: 38989629 PMCID: PMC11263814 DOI: 10.1111/iep.12513] [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: 03/03/2024] [Revised: 05/23/2024] [Accepted: 06/08/2024] [Indexed: 07/12/2024] Open
Abstract
Oesophageal cancer (EC) is a malignancy which accounts for a substantial number of cancer-related deaths worldwide. The molecular mechanisms underlying the pathogenesis of EC have not been fully elucidated. GSE17351 and GSE20347 data sets from the Gene Expression Omnibus (GEO) database were employed to screen differentially expressed genes (DEGs). Reverse transcription quantitative PCR (RT-qPCR) was used to examine hub gene expression. ECA-109 and TE-12 cells were transfected using the pcDNA3.1 expression vector encoding GABRP. The cell counting kit-8 (CCK-8), cell scratch and Transwell assays were performed to assess the effect of GABRP on EC cell proliferation, migration and invasion. Epithelial-mesenchymal transition (EMT)-associated protein levels were measured by Western blotting. Subsequently, CFTR was knocked down to verify whether GABRP affected biological events in EC cells by targeting CFTR. Seven hub genes were identified, including GABRP, FLG, ENAH, KLF4, CD24, ABLIM3 and ABLIM1, which all could be used as diagnostic biomarkers for EC. The RT-qPCR results indicated that the expression levels of GABRP, FLG, KLF4, CD24, ABLIM3 and ABLIM1 were downregulated, whereas the expression level of ENAH was upregulated. In vitro functional assays demonstrated that GABRP overexpression suppressed the proliferation, migration, invasion and EMT of EC cells. Mechanistically, GABRP promoted the expression of CFTR, and CFTR knockdown significantly counteracted the influence of GABRP overexpression on biological events in EC cells. Overexpression of GABRP inhibited EC progression by increasing CFTR expression, which might be a new target for EC treatment.
Collapse
Affiliation(s)
- Jingzhi Zhang
- Department of GastroenterologyThe Affiliated Ganzhou Hospital of Nanchang UniversityGanzhou CityChina
| | - Xue Liu
- Department of GastroenterologyThe Affiliated Ganzhou Hospital of Nanchang UniversityGanzhou CityChina
| | - Ling Zeng
- Department of GastroenterologyThe Affiliated Ganzhou Hospital of Nanchang UniversityGanzhou CityChina
| | - Ying Hu
- Department of GastroenterologyThe First Affiliated Hospital of Gannan Medical UniversityGanzhou CityChina
| |
Collapse
|
32
|
Jin X, Zhang J, Zhang Y, He J, Wang M, Hei Y, Guo S, Xu X, Liu Y. Different origin-derived exosomes and their clinical advantages in cancer therapy. Front Immunol 2024; 15:1401852. [PMID: 38994350 PMCID: PMC11236555 DOI: 10.3389/fimmu.2024.1401852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Exosomes, as a class of small extracellular vesicles closely related to the biological behavior of various types of tumors, are currently attracting research attention in cancer diagnosis and treatment. Regarding cancer diagnosis, the stability of their membrane structure and their wide distribution in body fluids render exosomes promising biomarkers. It is expected that exosome-based liquid biopsy will become an important tool for tumor diagnosis in the future. For cancer treatment, exosomes, as the "golden communicators" between cells, can be designed to deliver different drugs, aiming to achieve low-toxicity and low-immunogenicity targeted delivery. Signaling pathways related to exosome contents can also be used for safer and more effective immunotherapy against tumors. Exosomes are derived from a wide range of sources, and exhibit different biological characteristics as well as clinical application advantages in different cancer therapies. In this review, we analyzed the main sources of exosomes that have great potential and broad prospects in cancer diagnosis and therapy. Moreover, we compared their therapeutic advantages, providing new ideas for the clinical application of exosomes.
Collapse
Affiliation(s)
- Xiaoyan Jin
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Jing Zhang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
- The Second Affiliated Hospital of Xi‘an Medical University, Xi’an, Shaanxi, China
| | - Yufu Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Jing He
- Laboratory of Obstetrics and Gynecology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Mingming Wang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yu Hei
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Shutong Guo
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Xiangrong Xu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yusi Liu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
33
|
Li X, Xu B, Yang H, Zhu Z. Gut Microbiota, Human Blood Metabolites, and Esophageal Cancer: A Mendelian Randomization Study. Genes (Basel) 2024; 15:729. [PMID: 38927665 PMCID: PMC11203100 DOI: 10.3390/genes15060729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Unbalances in the gut microbiota have been proposed as a possible cause of esophageal cancer (ESCA), yet the exact causal relationship remains unclear. PURPOSE To investigate the potential causal relationship between the gut microbiota and ESCA with Mendelian randomization (MR) analysis. METHODS Genome-wide association studies (GWASs) of 207 gut microbial taxa (5 phyla, 10 classes, 13 orders, 26 families, 48 genera, and 105 species) and 205 gut microbiota metabolic pathways conducted by the Dutch Microbiome Project (DMP) and a FinnGen cohort GWAS of esophageal cancer specified the summary statistics. To investigate the possibility of a mediation effect between the gut microbiota and ESCA, mediation MR analyses were performed for 1091 blood metabolites and 309 metabolite ratios. RESULTS MR analysis indicated that the relative abundance of 10 gut microbial taxa was associated with ESCA but all the 12 gut microbiota metabolic pathways with ESCA indicated no statistically significant association existing. Two blood metabolites and a metabolite ratio were discovered to be mediating factors in the pathway from gut microbiota to ESCA. CONCLUSION This research indicated the potential mediating effects of blood metabolites and offered genetic evidence in favor of a causal correlation between gut microbiota and ESCA.
Collapse
Affiliation(s)
- Xiuzhi Li
- State Key Laboratory of Oncology in South China, Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China;
| | - Bingchen Xu
- State Key Laboratory of Oncology in South China, Department of Minimally Invasive Intervention, Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China;
| | - Han Yang
- State Key Laboratory of Oncology in South China, Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China;
| | - Zhihua Zhu
- State Key Laboratory of Oncology in South China, Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
34
|
Han L, Xiang X, Fu Y, Wei S, Zhang C, Li L, Liu Y, Lv H, Shan B, Zhao L. Periplcymarin targets glycolysis and mitochondrial oxidative phosphorylation of esophageal squamous cell carcinoma: Implication in anti-cancer therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155539. [PMID: 38522311 DOI: 10.1016/j.phymed.2024.155539] [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: 10/16/2023] [Revised: 01/28/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is the predominant histological subtype of esophageal cancer (EC) in China, and demonstrates varying levels of resistance to multiple chemotherapeutic agents. Our previous studies have proved that periplocin (CPP), derived from the extract of cortex periplocae, exhibiting the capacity to hinder proliferation and induce apoptosis in ESCC cells. Several studies have identified additional anti-cancer constituents in the extract of cortex periplocae, named periplcymarin (PPM), sharing similar compound structure with CPP. Nevertheless, the inhibitory effects of PPM on ESCC and their underlying mechanisms remain to be further elucidated. PURPOSE The aim of this study was to investigate function of PPM inhibiting the growth of ESCC in vivo and in vitro and to explore its underlying mechanism, providing the potential anti-tumor drug for ESCC. METHODS Initially, a comparative analysis was conducted on the inhibitory activity of three naturally compounds obtained from the extract of cortex periplocae on ESCC cells. Among these compounds, PPM was chosen for subsequent investigation owing to its comparatively structure and anti-tumor activity simultaneously. Subsequently, a series of biological functional experiments were carried out to assess the impact of PPM on the proliferation, apoptosis and cell cycle arrest of ESCC cells in vitro. In order to elucidate the molecular mechanism of PPM, various methodologies were employed, including bioinformatics analyses and mechanistic experiments such as high-performance liquid chromatography combined with mass spectrometry (HPLC-MS), cell glycolysis pressure and mitochondrial pressure test. Additionally, the anti-tumor effects of PPM on ESCC cells and potential toxic side effects were evaluated in vivo using the nude mice xenograft assay. RESULTS Our study revealed that PPM possesses the ability to impede the proliferation of ESCC cells, induce apoptosis, and arrest the cell cycle of ESCC cells in the G2/M phase in vitro. Mechanistically, PPM exerted its effects by modulating glycolysis and mitochondrial oxidative phosphorylation (OXPHOS), as confirmed by glycolysis pressure and mitochondrial pressure tests. Moreover, rescue assays demonstrated that PPM inhibits glycolysis and OXPHOS in ESCC cells through the PI3K/AKT and MAPK/ERK signaling pathways. Additionally, we substantiated that PPM effectively suppresses the growth of ESCC cells in vivo, with only modest potential toxic side effects. CONCLUSION Our study provides novel evidence that PPM has the potential to simultaneously target glycolysis and mitochondrial OXPHOS in ESCC cells. This finding highlights the need for further investigation into PPM as a promising therapeutic agent that targets the tumor glucose metabolism pathway in ESCC.
Collapse
Affiliation(s)
- Lujuan Han
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Department of Pathogenic Biology, Hebei Medical University, Zhongshan Road 361, Shijiazhuang, 050017, PR China
| | - Xiaohan Xiang
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Yuhui Fu
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Sisi Wei
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Cong Zhang
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Lei Li
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China
| | - Yueping Liu
- Department of Pathology, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China
| | - Huilai Lv
- Department of Thoracic Surgery, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China
| | - Baoen Shan
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China.
| | - Lianmei Zhao
- Research Center, the Fourth Hospital of Hebei Medical University, Jiankang Road 12, Shijiazhuang, 050011, PR China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, Hebei Province, Shijiazhuang, 050011, PR China.
| |
Collapse
|
35
|
Sun X, Li L, Yang X, Ke D, Zhong Q, Zhu Y, Yang L, Zhang Z, Lin J. Identification of a novel prognostic cuproptosis-associated LncRNA signature for predicting prognosis and immunotherapy response in patients with esophageal cancer. Heliyon 2024; 10:e30277. [PMID: 38707466 PMCID: PMC11068819 DOI: 10.1016/j.heliyon.2024.e30277] [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: 09/20/2023] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
Nowadays, effective prognostic models for esophageal cancer (ESCA) are still lacking. Long noncoding RNAs (lncRNAs) are commonly utilized as indicators for diagnosing cancer and forecasting patient outcomes. Cuproptosis is regulated by multiple genes and is crucial to the progression of ESCA. However, it is not yet clear what role the cuproptosis-associated lncRNAs (CuALs) play in ESCA. To tackle this problem, a prognostic signature incorporating three CuALs was created. This signature was constructed by the use of the least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression. Subsequently, the signature effectively stratified ESCA samples into a high-risk group and a low-risk group. Those in the low-risk group demonstrated extended overall survival (OS), as well as increased infiltration of T cells, macrophages, and NK cells, suggesting a potentially enhanced response to immunotherapy. The ROC curve analysis demonstrated that this prognostic signature outperformed conventional clinical factors in predicting patient prognosis (AUC = 0.708). K-M survival analysis and correlation analysis identified UGDH-AS1 (a CuAL) as a protective factor positively associated with patient prognosis. The results of RT-qPCR and wound healing assays indicated that UGDH-AS1 is overexpressed in ESCA and could inhibit cancer cell migration. In general, the prognostic signature of CuALs demonstrated a robust capability in forecasting the immune environment and patient prognosis, highlighting its potential as a tool for enhancing personalized treatment strategies in ESCA.
Collapse
Affiliation(s)
- Xinhai Sun
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Liming Li
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojie Yang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Ke
- Heilongjiang Key Laboratory of tissue damage and repair, College of life sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Qihong Zhong
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuanchang Zhu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Litao Yang
- Department of Thoracic Surgery, Baoji Traditional Chinese Medicine Hospital, Shaanxi, China
| | - Zhenyang Zhang
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiangbo Lin
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
36
|
Sun X, Li L, Yang X, Ke D, Zhong Q, Zhu Y, Yang L, Zhang Z, Lin J. Identification of a novel prognostic cuproptosis-associated LncRNA signature for predicting prognosis and immunotherapy response in patients with esophageal cancer. Heliyon 2024; 10:e30277. [PMID: 38707466 DOI: 10.1016/j.heliyon.2024.e30277if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 09/15/2024] Open
Abstract
Nowadays, effective prognostic models for esophageal cancer (ESCA) are still lacking. Long noncoding RNAs (lncRNAs) are commonly utilized as indicators for diagnosing cancer and forecasting patient outcomes. Cuproptosis is regulated by multiple genes and is crucial to the progression of ESCA. However, it is not yet clear what role the cuproptosis-associated lncRNAs (CuALs) play in ESCA. To tackle this problem, a prognostic signature incorporating three CuALs was created. This signature was constructed by the use of the least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression. Subsequently, the signature effectively stratified ESCA samples into a high-risk group and a low-risk group. Those in the low-risk group demonstrated extended overall survival (OS), as well as increased infiltration of T cells, macrophages, and NK cells, suggesting a potentially enhanced response to immunotherapy. The ROC curve analysis demonstrated that this prognostic signature outperformed conventional clinical factors in predicting patient prognosis (AUC = 0.708). K-M survival analysis and correlation analysis identified UGDH-AS1 (a CuAL) as a protective factor positively associated with patient prognosis. The results of RT-qPCR and wound healing assays indicated that UGDH-AS1 is overexpressed in ESCA and could inhibit cancer cell migration. In general, the prognostic signature of CuALs demonstrated a robust capability in forecasting the immune environment and patient prognosis, highlighting its potential as a tool for enhancing personalized treatment strategies in ESCA.
Collapse
Affiliation(s)
- Xinhai Sun
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Liming Li
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojie Yang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Ke
- Heilongjiang Key Laboratory of tissue damage and repair, College of life sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Qihong Zhong
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuanchang Zhu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Litao Yang
- Department of Thoracic Surgery, Baoji Traditional Chinese Medicine Hospital, Shaanxi, China
| | - Zhenyang Zhang
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiangbo Lin
- Department of Thoracic Surgery, Fujian Institute of Thoracic and Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
37
|
Xu J, Jiang W, Hu T, Long Y, Shen Y. NEDD4 and NEDD4L: Ubiquitin Ligases Closely Related to Digestive Diseases. Biomolecules 2024; 14:577. [PMID: 38785984 PMCID: PMC11117611 DOI: 10.3390/biom14050577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Protein ubiquitination is an enzymatic cascade reaction and serves as an important protein post-translational modification (PTM) that is involved in the vast majority of cellular life activities. The key enzyme in the ubiquitination process is E3 ubiquitin ligase (E3), which catalyzes the binding of ubiquitin (Ub) to the protein substrate and influences substrate specificity. In recent years, the relationship between the subfamily of neuron-expressed developmental downregulation 4 (NEDD4), which belongs to the E3 ligase system, and digestive diseases has drawn widespread attention. Numerous studies have shown that NEDD4 and NEDD4L of the NEDD4 family can regulate the digestive function, as well as a series of related physiological and pathological processes, by controlling the subsequent degradation of proteins such as PTEN, c-Myc, and P21, along with substrate ubiquitination. In this article, we reviewed the appropriate functions of NEDD4 and NEDD4L in digestive diseases including cell proliferation, invasion, metastasis, chemotherapeutic drug resistance, and multiple signaling pathways, based on the currently available research evidence for the purpose of providing new ideas for the prevention and treatment of digestive diseases.
Collapse
Affiliation(s)
| | | | | | | | - Yueming Shen
- Department of Digestive Diseases, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, 161 Shaoshan Road, Changsha 410000, China; (J.X.); (W.J.); (T.H.); (Y.L.)
| |
Collapse
|
38
|
Han S, Chen Y, Huang Y, Jin L, Ma Y. Arecoline promotes Akt-c-Myc-driven aerobic glycolysis in esophageal epithelial cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:2794-2802. [PMID: 38282581 DOI: 10.1002/tox.24159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/02/2024] [Accepted: 01/18/2024] [Indexed: 01/30/2024]
Abstract
Aerobic glycolysis is a typical metabolic rearrangement for tumorigenesis. Arecoline is of explicit carcinogenicity, numerous works demonstrate its mutagenicity, genotoxicity, and cytotoxicity. However, the effects of arecoline on aerobic glycolysis of esophageal epithelial cells remain unclear. In the present study, 5 μM arecoline efficiently increased HK2 expression to induce aerobic glycolysis in Het-1A-Are and NE2-Are cells. The mechanistic analysis showed that arecoline activated the Akt-c-Myc signaling pathway and reduced the GSK3β-mediated phosphorylation of c-Myc on Thr58 to prevent its ubiquitination and destruction, subsequently promoting HK2 transcription and expression. Taken together, these results suggest that arecoline can induce aerobic glycolysis of esophageal epithelial cells and further confirm that arecoline is a carcinogen harmful to human health.
Collapse
Affiliation(s)
- Shuangze Han
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingji Chen
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu Huang
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Longyu Jin
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yuchao Ma
- Department of Cardiothoracic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
39
|
Zhang XJ, Yu Y, Zhao HP, Guo L, Dai K, Lv J. Mechanisms of tumor immunosuppressive microenvironment formation in esophageal cancer. World J Gastroenterol 2024; 30:2195-2208. [PMID: 38690024 PMCID: PMC11056912 DOI: 10.3748/wjg.v30.i16.2195] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/05/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
As a highly invasive malignancy, esophageal cancer (EC) is a global health issue, and was the eighth most prevalent cancer and the sixth leading cause of cancer-related death worldwide in 2020. Due to its highly immunogenic nature, emer-ging immunotherapy approaches, such as immune checkpoint blockade, have demonstrated promising efficacy in treating EC; however, certain limitations and challenges still exist. In addition, tumors may exhibit primary or acquired resistance to immunotherapy in the tumor immune microenvironment (TIME); thus, understanding the TIME is urgent and crucial, especially given the im-portance of an immunosuppressive microenvironment in tumor progression. The aim of this review was to better elucidate the mechanisms of the suppressive TIME, including cell infiltration, immune cell subsets, cytokines and signaling pathways in the tumor microenvironment of EC patients, as well as the downregulated expression of major histocompatibility complex molecules in tumor cells, to obtain a better understanding of the differences in EC patient responses to immunotherapeutic strategies and accurately predict the efficacy of immunotherapies. Therefore, personalized treatments could be developed to maximize the advantages of immunotherapy.
Collapse
Affiliation(s)
- Xiao-Jun Zhang
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Yan Yu
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - He-Ping Zhao
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Lei Guo
- Department of Spinal Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| | - Kun Dai
- Department of Clinical Laboratory, Yanliang Railway Hospital of Xi’an, Xi’an 710089, Shaanxi Province, China
| | - Jing Lv
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, Shaanxi Province, China
| |
Collapse
|
40
|
Ma R, Okugawa Y, Shimura T, Yamashita S, Sato Y, Yin C, Uratani R, Kitajima T, Imaoka H, Kawamura M, Morimoto Y, Okita Y, Yoshiyama S, Ohi M, Toiyama Y. Clinical implications of C-reactive protein-albumin-lymphocyte (CALLY) index in patients with esophageal cancer. Surg Oncol 2024; 53:102044. [PMID: 38335851 DOI: 10.1016/j.suronc.2024.102044] [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: 09/25/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
PURPOSE The C-reactive protein-albumin-lymphocyte (CALLY) index is a novel inflammatory nutritional biomarker. This study aimed to investigate the potential clinical significance and oncological prognostic role of the preoperative CALLY index in patients with esophageal cancer. METHODS We analyzed the preoperative CALLY index in 146 patients with esophageal cancer. The CALLY index and clinicopathological variables were analyzed by the Mann-Whitney U test, and associations between the CALLY index and survival outcomes were analyzed by Kaplan-Meier analysis and log-rank tests. Univariate and multivariate analyses of prognostic variables were conducted using Cox proportional hazards regression. RESULTS A lower preoperative CALLY index was significantly correlated with patient age, advanced T stage, presence of lymph node metastasis, neoadjuvant therapy, lymphatic invasion, and advanced stage classification. The preoperative CALLY index decreased significantly in a stage-dependent manner. Patients with esophageal cancer with a low CALLY index had poorer overall survival, disease-free survival than those with a high CALLY index. Multivariate analysis showed that a low CALLY index was an independent prognostic factor for overall survival, disease-free survival and an independent predictor of postoperative surgical site infection. CONCLUSIONS Preoperative CALLY index is a useful marker to guide the perioperative and postoperative management of patients with esophageal cancer.
Collapse
Affiliation(s)
- Ruiya Ma
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Yoshinaga Okugawa
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan; Department of Genomic Medicine, Mie University Hospital, Mie, Japan.
| | - Tadanobu Shimura
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Shinji Yamashita
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Yuhki Sato
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Chengzeng Yin
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Ryo Uratani
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Takahito Kitajima
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan; Department of Genomic Medicine, Mie University Hospital, Mie, Japan
| | - Hiroki Imaoka
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Mikio Kawamura
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Yuhki Morimoto
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Yoshiki Okita
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Shigeyuki Yoshiyama
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Masaki Ohi
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Mie University Graduate School of Medicine, Mie, Japan.
| |
Collapse
|
41
|
Yan J, Zhou Y, Wang Y, Liu Y. PARG Promotes Esophagus Cancer Cell Metastasis by Activation of the Wnt/β-Catenin Pathway. Biochem Genet 2024; 62:761-774. [PMID: 37429965 DOI: 10.1007/s10528-023-10434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/15/2023] [Indexed: 07/12/2023]
Abstract
Esophagus cancer (EC) is a highly malignant and metastatic cancer. Poly(ADP-ribose) glycohydrolase (PARG), a DNA replication and repair regulator, inhibits cancer cell replication defects. This study aimed to explore the role of PARG in EC. The biological behaviors were analyzed using MTT assay, Transwell assay, scratch test, cell adhesion assay, and western blot. PARG expression was detected using quantitative PCR and immunohistochemical assay. The regulation of the Wnt/β-catenin pathway was assessed using western blot. The results showed that PARG was highly expressed in EC tissues and cells. Knockdown of PARG suppressed cell viability, invasion, migration, adhesion, and epithelial-mesenchymal transition. Conversely, overexpression of PARG promoted the biological behaviors mentioned above. Moreover, overexpression of PARG promoted the activation of the Wnt/β-catenin pathway rather than the STAT and Notch pathways. XAV939, the Wnt/β-catenin pathway inhibitor, partly abolished the biological behaviors mediated by PARG overexpression. In conclusion, PARG promoted the malignant advancement of EC via activating the Wnt/β-catenin pathway. These findings suggested that PARG might be a new therapeutic target for EC.
Collapse
Affiliation(s)
- Jiaxin Yan
- Department of Pathology, Sichuan Cancer Hospital, 55 Renmin South Road, Wuhou District, Chengdu City, 610000, Sichuan, China
| | - Yehan Zhou
- Department of Pathology, Sichuan Cancer Hospital, 55 Renmin South Road, Wuhou District, Chengdu City, 610000, Sichuan, China
| | - Yalan Wang
- Department of Pathology, School of Basic Medical Sciences, Chongqing Medical University, Chengdu City, China
| | - Yang Liu
- Department of Pathology, Sichuan Cancer Hospital, 55 Renmin South Road, Wuhou District, Chengdu City, 610000, Sichuan, China.
| |
Collapse
|
42
|
Shang H, Chen Y, Wang Q, Yang Y, Zhang J. A Correlation Evaluation Between the Peripheral Blood Index and the Prognosis of Advanced Esophageal Squamous Cell Carcinoma Patients Treated with Camrelizumab. J Inflamm Res 2024; 17:2009-2021. [PMID: 38566981 PMCID: PMC10986412 DOI: 10.2147/jir.s450669] [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: 11/19/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Purpose This study aimed to investigate the relationship between peripheral blood indices and the efficacy and prognosis of advanced esophageal squamous cell carcinoma (ESCC) patients treated with camrelizumab. Patients and Methods We retrospectively analyzed 64 patients who received camrelizumab for advanced ESCC at the Second People's Hospital of Lianyungang City between July 2020 and June 2022. The study included examination of the neutrophil-to-lymphocyte ratio (NLR), the platelet-to-lymphocyte ratio (PLR), the systemic inflammation index (SII), the lymph-to-monocytes ratio (LMR), the absolute lymphocyte count (ALC), and lactate dehydrogenase (LDH). We used multivariate logistic regression analysis to explore the link existing between peripheral blood and the efficacy of treatment. Determination of potential prognostic factors for Progression-free survival (PFS) and Overall survival (OS) using Cox regression analysis. The nomogram model was developed based on the results of the Cox multivariate analysis. Patients were divided into three groups according to the reduction in LDH and LDL levels before treatment, and the Kaplan-Meier survival curves for the three groups were compared and ROC curves for LDH combined with PLR were plotted. Results Lower LDH (OR=6.237, 95% CI: 1.625-23.944) were independently associated with disease control rates(DCR). LDH was independently correlated with PFS (HR: 0.227 95% CI: 0.099-0.517). LDH and PLR were independently linked to OS. The C index of the nomogram model is 0.819, indicating good predictive performance. Kaplan-Meier Survival Curve suggested better OS in patients with reduced pretreatment LDH and PLR. The area under the ROC curve showed that the LDH index combined with the PLR index predicts patient survival better than the index alone. Conclusion LDH combined with PLR predicted prognosis in patients with ESCC treated with camrelizumab.
Collapse
Affiliation(s)
- Haotian Shang
- Department of Oncology, Clinical College of Second People’s Hospital of Lianyungang, Bengbu Medical College, Lianyungang, People’s Republic of China
| | - Yanan Chen
- Department of Oncology, Clinical College of Second People’s Hospital of Lianyungang, Bengbu Medical College, Lianyungang, People’s Republic of China
| | - Qiulu Wang
- Department of Oncology, Clinical College of Second People’s Hospital of Lianyungang, Bengbu Medical College, Lianyungang, People’s Republic of China
| | - Yongliang Yang
- Department of Oncology, Clinical College of Second People’s Hospital of Lianyungang, Bengbu Medical College, Lianyungang, People’s Republic of China
| | - Jingyu Zhang
- Department of Oncology, Clinical College of Second People’s Hospital of Lianyungang, Bengbu Medical College, Lianyungang, People’s Republic of China
| |
Collapse
|
43
|
Li P, Yang L, Park SY, Liu F, Li AH, Zhu Y, Sui H, Gao F, Li L, Ye L, Zou Y, Tian Z, Zhao Y, Costa M, Sun H, Zhao X. Stabilization of MOF (KAT8) by USP10 promotes esophageal squamous cell carcinoma proliferation and metastasis through epigenetic activation of ANXA2/Wnt signaling. Oncogene 2024; 43:899-917. [PMID: 38317006 DOI: 10.1038/s41388-024-02955-z] [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: 09/05/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/07/2024]
Abstract
Dysregulation of MOF (also known as MYST1, KAT8), a highly conserved H4K16 acetyltransferase, plays important roles in human cancers. However, its expression and function in esophageal squamous cell carcinoma (ESCC) remain unknown. Here, we report that MOF is highly expressed in ESCC tumors and predicts a worse prognosis. Depletion of MOF in ESCC significantly impedes tumor growth and metastasis both in vitro and in vivo, whereas ectopic expression of MOF but not catalytically inactive mutant (MOF-E350Q) promotes ESCC progression, suggesting that MOF acetyltransferase activity is crucial for its oncogenic activity. Further analysis reveals that USP10, a deubiquitinase highly expressed in ESCC, binds to and deubiquitinates MOF at lysine 410, which protects it from proteosome-dependent protein degradation. MOF stabilization by USP10 promotes H4K16ac enrichment in the ANXA2 promoter to stimulate ANXA2 transcription in a JUN-dependent manner, which subsequently activates Wnt/β-Catenin signaling to facilitate ESCC progression. Our findings highlight a novel USP10/MOF/ANXA2 axis as a promising therapeutic target for ESCC.
Collapse
Affiliation(s)
- Peichao Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Chest Cancer, The Second Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Lingxiao Yang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Sun Young Park
- Division of Environmental Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, 10010, USA
| | - Fanrong Liu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Alex H Li
- Division of Environmental Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, 10010, USA
| | - Yilin Zhu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Huacong Sui
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Fengyuan Gao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lingbing Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lan Ye
- Cancer Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhongxian Tian
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Chest Cancer, The Second Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Yunpeng Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Max Costa
- Division of Environmental Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, 10010, USA
| | - Hong Sun
- Division of Environmental Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, 10010, USA.
| | - Xiaogang Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
- Key Laboratory of Chest Cancer, The Second Hospital, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
44
|
Liu XS, Liu ZY, Zeng DB, Hu J, Chen XL, Gu JL, Gao Y, Pei ZJ. Functional enrichment analysis reveals the involvement of DARS2 in multiple biological pathways and its potential as a therapeutic target in esophageal carcinoma. Aging (Albany NY) 2024; 16:3934-3954. [PMID: 38382106 PMCID: PMC10929822 DOI: 10.18632/aging.205569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024]
Abstract
OBJECTIVE The enzyme Aspartyl tRNA synthetase 2 (DARS2) is a crucial enzyme in the mitochondrial tRNA synthesis pathway, playing a critical role in maintaining normal mitochondrial function and protein synthesis. However, the role of DARS2 in ESCA is unclear. MATERIALS AND METHODS Transcriptional data of pan-cancer and ESCA were downloaded from UCSC XENA, TCGA, and GEO databases to analyze the differential expression of DARS2 between tumor samples and normal samples, and its correlation with clinicopathological features of ESCA patients. R was used for GO, KEGG, and GSEA functional enrichment analysis of DARS2 co-expression and to analyze the connection of DARS2 with glycolysis and m6A-related genes. In vitro experiments were performed to assess the effects of interfering with DARS2 expression on ESCA cells. TarBase v.8, mirDIP, miRTarBase, ENCORI, and miRNet databases were used to analyze and construct a ceRNA network containing DARS2. RESULTS DARS2 was overexpressed in various types of tumors. In vitro experiments confirmed that interfering with DARS2 expression significantly affected the proliferation, migration, apoptosis, cell cycle, and glycolysis of ESCA cells. DARS2 may be involved in multiple biological pathways related to tumor development. Furthermore, correlation and differential analysis revealed that DARS2 may regulate ESCA m6A modification through its interaction with METTL3 and YTHDF1. A ceRNA network containing DARS2, DLEU2/has-miR-30a-5p/DARS2, was successfully predicted and constructed. CONCLUSIONS Our findings reveal the upregulation of DARS2 in ESCA and its association with clinical features, glycolysis pathway, m6A modification, and ceRNA network. These discoveries provide valuable insights into the molecular mechanisms underlying ESCA.
Collapse
Affiliation(s)
- Xu-Sheng Liu
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Zi-Yue Liu
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Dao-Bing Zeng
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Jian Hu
- Department of Critical Care Medicine, Danjiangkou First Hospital, Danjiangkou 420381, Hubei, China
| | - Xuan-Long Chen
- Department of Medical Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Jiao-Long Gu
- Department of Obstetrics and Gynecology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yan Gao
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Zhi-Jun Pei
- Department of Nuclear Medicine, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| |
Collapse
|
45
|
Liu Y, Gu Y, Zhou J, Zhang H, Shang Q, Yang Y, Chen L. Mendelian randomization analysis of atopic dermatitis and esophageal cancer in East Asian and European populations. World Allergy Organ J 2024; 17:100868. [PMID: 38293274 PMCID: PMC10825168 DOI: 10.1016/j.waojou.2023.100868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024] Open
Abstract
Background Emerging observational studies showed an association between atopic dermatitis (AD) and gastrointestinal cancers. However, it remains unclear whether this association is causal, particularly in the case of cancers like esophageal cancer, which exhibit ancestral genetic traits. Methods To assess the potential causal relationship between AD and esophageal cancer across diverse ancestral backgrounds, we conducted a 2-sample Mendelian randomization study. Independent genetic instruments for AD from the FinnGen consortium (N case = 7024 and N control = 198 740), BioBank Japan (N case = 2385 and N control = 209 651) and Early Genetics and Lifecourse Epidemiology (EAGLE) eczema consortium (N case = 18 900 and N control = 84 166, without the 23andMe study) were used to investigate the association with esophageal cancer in the UK Biobank study (N case = 740 and N control = 372 016) and BioBank Japan esophageal cancer sample (N case = 1300 and N control = 197 045). Results When esophageal cancer extracted from East Asian ancestry was used as a outcome factor, AD data extracted from BioBank Japan (OR = 0.90, 95% CI: 0.83-0.98), FinnGen consortium (OR = 0.86, 95% CI: 0.77-0.96), and EAGLE consortium (OR = 0.92, 95% CI: 0.81-1.06) were negatively associated with esophageal cancer susceptibility. However, AD as a whole did not show an association with esophageal cancer from European ancestry. Conclusion This study provides support for a causal relationship between AD and esophageal cancer in East Asian populations but not between AD and esophageal cancer from European ancestry. The specific associations between esophageal cancer and AD appear to exhibit significant disparities between the East Asian and European regions.
Collapse
Affiliation(s)
| | | | | | - Hanlu Zhang
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qixin Shang
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yushang Yang
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Su X, Fu C, Liu F, Bian R, Jing P. T-cell exhaustion prediction algorithm in tumor microenvironment for evaluating prognostic stratification and immunotherapy effect of esophageal cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:592-611. [PMID: 37493251 DOI: 10.1002/tox.23887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/01/2023] [Accepted: 06/29/2023] [Indexed: 07/27/2023]
Abstract
Esophageal cancer (EC) is a common digestive malignancy that ranks sixth in cancer deaths, with a 5-year survival rate of 15%-25%. As a result, reliable prognostic biomarkers are required to accurately predict the prognosis of EC. T-cell exhaustion (TEX) is associated with poorer prognosis and immune infiltration in EC. In this study, nine risk genes were finally screened to constitute the prognostic model using least absolute shrinkage and selection operator analysis. Patients were divided into two groups based on the expression of the TEX-related genes: high-risk group and low-risk group. The expression of TEX-related genes differed significantly between the two groups. The findings revealed that the risk model developed was highly related to the clinical prognosis and amount of immune cell infiltration in EC patients. It was also significantly correlated with the therapeutic sensitivity of multiple chemotherapeutic agents in EC patients. Subsequently, we successfully constructed drug-resistant cell lines KYSE480/CDDP-R and KYSE180/CDDP-R to verify the correlation between PD-1 and drug resistance in EC. Then, we examined the mRNA and protein expression levels of PD-1 in parental and drug-resistant cells using qPCR and WB. It was found that the expression level of PD-1 was significantly increased in the plasma red of drug-resistant cells. Next, we knocked down PD-1 in drug-resistant cells and found that the resistance of EC cells to CDDP was significantly reduced. And the proportion of apoptotic cells in cells treated with 6 μM CDDP for 24 h was significantly in increase. The TEX-based risk model achieved good prediction results for prognosis prediction in EC patients. And it was also significantly associated with the level of immune cell infiltration and drug therapy sensitivity of EC patients. Additionally, the downregulation of PD-1 may be associated with increased drug sensitivity in EC and enhanced T-cell infiltration. The high-risk group had lower TIDE scores, indicating that the high-risk group benefits more after receiving immunotherapy. Thus, the TEX-based risk model can be used as a novel tumor prognostic biomarker.
Collapse
Affiliation(s)
- Xiangyu Su
- School of Medicine, Southeast University, Nanjing, China
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chenchun Fu
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Fei Liu
- Department of Oncology, Luhe People's Hospital of Nanjing, Nanjing, China
| | - Rongrong Bian
- Department of Oncology, Luhe People's Hospital of Nanjing, Nanjing, China
| | - Ping Jing
- Department of Gastroenterology, Luhe People's Hospital of Nanjing, Nanjing, China
| |
Collapse
|
47
|
Nopour R. Design of risk prediction model for esophageal cancer based on machine learning approach. Heliyon 2024; 10:e24797. [PMID: 38312629 PMCID: PMC10835323 DOI: 10.1016/j.heliyon.2024.e24797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Background and aim Esophageal cancer (EC) is a highly prevalent and progressive disease. Early prediction of EC risk in the population is crucial in preventing this disease and enhancing the overall health of individuals. So far, few studies have been conducted on predicting the EC risk based on the prediction models, and most of them focused on statistical methods. The ML approach obtained efficient predictive insights into the clinical domain. Therefore, this study aims to develop a risk prediction model for EC based on risk factors and by leveraging the ML approach to stratify the high-risk EC people and obtain efficient preventive purposes at the community level. Material and methods The current retrospective study was performed from 2018 to 2022 in Sari City based on 3256 EC and non-EC cases. The six selected algorithms, including Random Forest (RF), eXtreme Gradient Boosting (XG-Boost), Bagging, K-Nearest Neighbor (K-NN), Support Vector Machine (SVM), and Artificial Neural Networks (ANNs), were used to develop the risk prediction model for EC and achieve the preventive purposes. Results Comparing the performance efficiency of algorithms revealed that the XG-Boost model gained the best predictability for EC risk with AU-ROC = 0.92 and AU-ROC-test = 0.889 for internal and validation states, respectively. Based on the XG-Boost, the factors, including sex, drinking hot liquids, fruit consumption, achalasia, and vegetable consumption, were considered the five top predictors of EC risk. Conclusion This study showed that the XG-Boost could provide insight into the early prediction of the EC risk for people and clinical providers to stratify the high-risk group of EC and achieve preventive measures based on modifying the risk factors associated with EC and other clinical solutions.
Collapse
Affiliation(s)
- Raoof Nopour
- Department of Health Information Management, Student Research Committee, School of Health Management and Information Sciences Branch, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Guo W, Li B, Xu W, Cheng C, Qiu C, Sam SK, Zhang J, Teng X, Meng L, Zheng X, Wang Y, Lou Z, Mao R, Lei H, Zhang Y, Zhou T, Li A, Cai J, Ge H. Multi-omics and Multi-VOIs to predict esophageal fistula in esophageal cancer patients treated with radiotherapy. J Cancer Res Clin Oncol 2024; 150:39. [PMID: 38280037 PMCID: PMC10821966 DOI: 10.1007/s00432-023-05520-5] [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: 08/10/2023] [Accepted: 11/20/2023] [Indexed: 01/29/2024]
Abstract
OBJECTIVE This study aimed to develop a prediction model for esophageal fistula (EF) in esophageal cancer (EC) patients treated with intensity-modulated radiation therapy (IMRT), by integrating multi-omics features from multiple volumes of interest (VOIs). METHODS We retrospectively analyzed pretreatment planning computed tomographic (CT) images, three-dimensional dose distributions, and clinical factors of 287 EC patients. Nine groups of features from different combination of omics [Radiomics (R), Dosiomics (D), and RD (the combination of R and D)], and VOIs [esophagus (ESO), gross tumor volume (GTV), and EG (the combination of ESO and GTV)] were extracted and separately selected by unsupervised (analysis of variance (ANOVA) and Pearson correlation test) and supervised (Student T test) approaches. The final model performance was evaluated using five metrics: average area under the receiver-operator-characteristics curve (AUC), accuracy, precision, recall, and F1 score. RESULTS For multi-omics using RD features, the model performance in EG model shows: AUC, 0.817 ± 0.031; 95% CI 0.805, 0.825; p < 0.001, which is better than single VOI (ESO or GTV). CONCLUSION Integrating multi-omics features from multi-VOIs enables better prediction of EF in EC patients treated with IMRT. The incorporation of dosiomics features can enhance the model performance of the prediction.
Collapse
Affiliation(s)
- Wei Guo
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Bing Li
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Wencai Xu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Chen Cheng
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Chengyu Qiu
- Department of Medical Informatics, Nantong University, Nantong, China
| | - Sai-Kit Sam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Lingguang Meng
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Xiaoli Zheng
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Yuan Wang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Zhaoyang Lou
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Ronghu Mao
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Hongchang Lei
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China
| | - Yuanpeng Zhang
- Department of Medical Informatics, Nantong University, Nantong, China
| | - Ta Zhou
- School of Electrical and Information Engineering, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Aijia Li
- Zhengzhou University School of Medicine, Zhengzhou, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Hong Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dong Ming Rd, Zhengzhou, Henan Province, China.
| |
Collapse
|
49
|
Wang L, Peng B, Yan Y, Liu G, Yang D, Wang Q, Li Y, Mao Q, Chen Q. The tRF-3024b hijacks miR-192-5p to increase BCL-2-mediated resistance to cytotoxic T lymphocytes in Esophageal Squamous Cell Carcinoma. Int Immunopharmacol 2024; 126:111135. [PMID: 37977065 DOI: 10.1016/j.intimp.2023.111135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
The limited efficacy of immune checkpoint inhibitors (ICIs) in the treatment of advanced Esophageal Squamous Cell Carcinoma (ESCC) poses a challenge. Recent evidence suggests that tumor cells' insensitivity to cytotoxic T lymphocytes (CTLs) contributes to drug resistance against ICIs. Here, a particular tRNA-derived fragment called tRF-3024b has been identified as playing a significant role in tumor cell resistance to CTLs. Through tRF sequencing (tRF-seq), we observed a high expression of tRF-3024b in ESCC cells that survived co-culture with CTLs. Further in vitro studies demonstrated that tRF-3024b reduced the apoptosis of tumor cells when co-cultured with CTLs. The mechanism behind this resistance involves tRF-3024b promoting the expression of B-cell lymphoma-2 (BCL-2) by sequestering miR-192-5p, a microRNA that would normally inhibit BCL-2 expression. This means that tRF-3024b indirectly enhances the protective effects of BCL-2, reducing apoptosis in tumor cells. Rescue assays confirmed that the suppressive function of tRF-3024b relies on BCL-2. In summary, the tRF-3024b/miR-192-5p/BCL-2 axis sheds light on the crucial role of tRF-3024b in regulating BCL-2 expression. These findings offer valuable insights into strategies to enhance the response of ESCC to CTLs and improve the effectiveness of immunotherapy approaches in treating ESCC.
Collapse
Affiliation(s)
- Lin Wang
- Department of Oncology, Department of Geriatric Lung Cancer Laboratory, The Affiliated Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Bo Peng
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan Yan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guangjun Liu
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dunpeng Yang
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qibin Wang
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongcheng Li
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Qixing Mao
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Qiang Chen
- Department of Thoracic Surgery, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
50
|
Xu LB, Wu CY, Wang Y, Zhou JY. Effect of Raltitrexed on ECA109 Cellular Radiosensitivity and its Mechanism in Esophageal Cancer. Curr Pharm Des 2024; 30:1519-1529. [PMID: 38716546 DOI: 10.2174/0113816128286282240405064210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/07/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND To investigate the effect of raltitrexed + X-ray irradiation on esophageal cancer ECA109 cells and analyze the potential action mechanism. METHODS The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to analyze the inhibitory effect of raltitrexed on cell proliferation. The effect of raltitrexed on radiosensitivity was studied through a clone-forming experiment. The scratch assay and invasion test were performed to understand the cell migration and invasion abilities. The apoptosis rate change was measured using a flow cytometer, and Western Blotting was used to determine the expression of B cell lymphoma-2 (Bcl-2) and Bcl2-associated X protein (Bax) in each group. RESULTS Raltitrexed significantly inhibited ECA109 proliferation in a time-dose-dependent manner; there were significant differences among different concentrations and times of action. The results of the clone-forming experiment showed a sensitization enhancement ratio of 1.65, and this demonstrated a radiosensitization effect. After the combination of raltitrexed with X-ray, the cell migration distance was shortened, and the number of cells penetrating the membrane was reduced. CONCLUSION Raltitrexed can inhibit the growth of esophageal cancer ECA109 cells and has a radiosensitization effect.
Collapse
Affiliation(s)
- Li-Ben Xu
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Chao-Yang Wu
- Department of Radiotherapy, Jiangsu University Affiliated People's Hospital, Zhenjiang 212000, Jiangsu, China
| | - Yan Wang
- Department of Radiotherapy, Jiangsu University Affiliated People's Hospital, Zhenjiang 212000, Jiangsu, China
| | - Ju-Ying Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| |
Collapse
|