1
|
Wen J, Cao X, Zhou B, Yang F, Wang X, Li Y, Zhao X, Mei J, Zhu W, Sun L, Huang F, Wang M. GC-MSCs transcriptionally upregulate SALL4 in gastric cancer through miR-4669/TIMP3/β-catenin signaling. Cell Signal 2025; 130:111668. [PMID: 39965736 DOI: 10.1016/j.cellsig.2025.111668] [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: 08/16/2024] [Revised: 12/07/2024] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUNDS Gastric cancer-associated mesenchymal stem cells (GC-MSCs) as integral components of the tumor microenvironment potentiate gastric cancer growth and metastasis. SALL4 is aberrantly upregulated in gastric cancer and pivotal for malignant progression. Whether GC-MSCs is responsible for SALL4 upregulation and the underlying mechanisms remains elusive. METHODS Cancer growth and metastasis capacities were assessed by cell colony formation assay, transwell assay, and epithelial-mesenchymal transition protein detection in vitro as well as subcutaneous xenograft and peritoneal metastasis models in vivo. SALL4 was measured by qPCR, western blot and immunohistochemistry staining. Gain- and loss-functional analysis were performed for miRNA and target gene. β-catenin signaling was assessed by immunofluorescence staining and Top/FopFlash luciferase assay. Transcriptional regulation was conducted using chemicals, luciferase reporter and ChIP assay. Clinical tissues and TCGA-STAD database were included for expression profile, correlation and clinical relevance analysis. RESULTS GC-MSCs promoted gastric cancer growth and metastasis along with elevation of SALL4 and miR-4669 in cancer cells and tissues. Overexpression of miR-4669 mimicked GC-MSC effects, while miR-4669 knockdown eliminated their oncogenic roles. TIMP3 was identified as a target of miR-4669 and mediated its functions. TIMP3 overexpression counteracted GC-MSC-induced cancer progression and SALL4 expression. GC-MSCs activated SALL4 transcription through the miR-4669/TIMP3/β-catenin pathway. The regulatory axis was aberrantly expressed in gastric cancer tissues, correlated with each other in certain cancer tissues and associated with lymph node metastasis. CONCLUSIONS GC-MSCs transcriptionally upregulate SALL4 to facilitate gastric cancer cell growth and metastasis via miR-4669/TIMP3/β-catenin pathway, highlighting the crucial role of GC-MSCs in the aberrant upregulation of SALL4.
Collapse
Affiliation(s)
- Jing Wen
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xiaoli Cao
- Department of Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province 226321, China
| | - Baocheng Zhou
- Department of Medical Laboratory, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu Province 222000, China
| | - Fang Yang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xiang Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Yuanyuan Li
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xinlan Zhao
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Jingyu Mei
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Wei Zhu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Li Sun
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan 215300, China
| | - Feng Huang
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan 215300, China; Department of Clinical Laboratory, Maternal and Child Health Care Hospital of Kunshan, Suzhou, Jiangsu Province, China
| | - Mei Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China.
| |
Collapse
|
2
|
Zhao C, Yu Y, Xiang P, Liao J, Yu B, Xing Y, Yin G. Association between radiotherapy and the risk of second primary malignancies in breast cancer patients with different estrogen receptor statuses. Eur J Cancer Prev 2025; 34:255-263. [PMID: 39230043 DOI: 10.1097/cej.0000000000000915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
BACKGROUND Breast cancer is the most common cancer among women. Second primary malignancies (SPMs) related to radiotherapy are significant complications. This study aims to investigate the correlation between radiotherapy and the occurrence of SPMs in breast cancer patients with different estrogen receptor statuses. METHODS We used data from the Surveillance, Epidemiology, and End Results (SEER) database, selecting estrogen receptor(+) and estrogen receptor(-) breast cancer patients from 1990 to 2015, with SPMs as the outcome measure. Fine-Gray competing risks regression and Poisson regression were employed to analyze the relationship between radiotherapy and the risk of SPMs in different estrogen receptor status groups. RESULTS Radiotherapy was associated with an increased risk of lung cancer, melanoma, non-Hodgkin lymphoma, and leukemia in estrogen receptor(+) patients. In estrogen receptor(-) patients, radiotherapy was linked to an increased risk of brain cancer and leukemia. The cumulative incidence, standardized incidence ratio, and subgroup analyses showed consistent results. In the dynamic assessment of radiotherapy-related risks, estrogen receptor(+) patients aged 50-70 exhibited a higher risk of leukemia and melanoma. Lung cancer risk was highest during a latency period of 20-30 years, while melanoma, non-Hodgkin lymphoma, and leukemia risks peaked within the first 10 years. For estrogen receptor(-) patients, brain cancer risk was higher between ages 50 and 70, and leukemia risk was elevated between ages 20 and 50. CONCLUSION Postoperative radiotherapy for breast cancer is associated with an increased risk of SPMs, with risks varying by estrogen receptor status and SPM type. Further research into the prevention of radiotherapy-related SPMs in different estrogen receptor status groups is crucial.
Collapse
Affiliation(s)
- Chengshan Zhao
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
3
|
Ma FC, Zhang GL, Chi BT, Tang YL, Peng W, Liu AQ, Chen G, Gao JB, Wei DM, Ge LY. Blood-based machine learning classifiers for early diagnosis of gastric cancer via multiple miRNAs. World J Gastrointest Oncol 2025; 17:103679. [DOI: 10.4251/wjgo.v17.i4.103679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/16/2025] [Accepted: 02/11/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Early screening methods for gastric cancer (GC) are lacking; therefore, the disease often progresses to an advanced stage when patients first start to exhibit typical symptoms. Endoscopy and pathological biopsy remain the primary diagnostic approaches, but they are invasive and not yet widely applicable for early population screening. miRNA is a highly conserved type of RNA that exists stably in plasma. Dysfunction of miRNA is linked to tumorigenesis and progression, indicating that individual miRNAs or combinations of multiple miRNAs may serve as potential biomarkers.
AIM To identify effective plasma miRNA biomarkers and investigate the clinical value of combining multiple miRNAs for early detection of GC.
METHODS Plasma samples from multiple centres were collected. Differentially expressed genes among healthy controls, early-stage GC patients, and advanced-stage GC patients were identified through small RNA sequencing (sRNA-seq) and validated via real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR). A Wilcoxon signed-rank test was used to investigate the differences in miRNAs. Sequencing datasets of GC serum samples were retrieved from the Gene Expression Omnibus (GEO), ArrayExpress, and The Cancer Genome Atlas databases, and a multilayer perceptron-artificial neural network (MLP-ANN) model was constructed for the key risk miRNAs. The pROC package was used to assess the discriminatory efficacy of the model.
RESULTS Plasma samples of 107 normal, 71 early GC and 97 advanced GC patients were obtained from three centres, and serum samples of 8443 normal and 1583 GC patients were obtained from the GEO database. The sRNA-seq and RT-qPCR experiments revealed that miR-452-5p, miR-5010-5p, miR-27b-5p, miR-5189-5p, miR-552-5p and miR-199b-5p were significantly increased in early GC patients compared with healthy controls and in advanced GC patients compared with early GC patients (P < 0.05). An MLP-ANN model was constructed for the six key miRNAs. The area under the curve (AUC) within the training cohort was 0.983 [95% confidence interval (CI): 0.980–0.986]. In the two validation cohorts, the AUCs were 0.995 (95%CI: 0.987 to nearly 1.000) and 0.979 (95%CI: 0.972–0.986), respectively.
CONCLUSION Potential miRNA biomarkers, including miR-452-5p, miR-5010-5p, miR-27b-5p, miR-5189-5p, miR-552-5p and miR-199b-5p, were identified. A GC classifier based on these miRNAs was developed, benefiting early detection and population screening.
Collapse
Affiliation(s)
- Fu-Chao Ma
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Guan-Lan Zhang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Bang-Teng Chi
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yu-Lu Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Wei Peng
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Ai-Qun Liu
- Department of Endoscopy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jin-Biao Gao
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Dan-Ming Wei
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Lian-Ying Ge
- Department of Endoscopy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
4
|
Okasha HH, Tag-Adeen M, Shaaban HE. Role of pancreatic juice cytology in diagnosis of high-grade pancreatic intraepithelial neoplasia. World J Clin Cases 2025; 13:94437. [DOI: 10.12998/wjcc.v13.i10.94437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 11/05/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
High-grade pancreatic intraepithelial neoplasia is a challenging diagnosis and it does not exhibit mass lesions. It is suspected based on changes in the main pancreatic duct in magnetic resonance cholangiopancreatography. Sometimes only an unclear duct shows in magnetic resonance cholangiopancreatography with no focal strictures and upstream dilatation of the main pancreatic duct. Serial pancreatic juice cytology is valuable in diagnosis of those patients.
Collapse
Affiliation(s)
- Hussein Hassan Okasha
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kasr Al-Aini School of Medicine, Cairo University, Cairo 11562, Egypt
| | - Mohammed Tag-Adeen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Qena Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Hossam Eldin Shaaban
- Department of Internal Medicine and Gastroenterology, National Hepatology and Tropical Medicine Research Institute, Cairo 11796, Egypt
| |
Collapse
|
5
|
Lu Q, Li L, Liang W, Xu G, Zhu J, Ma X, Tian W, Gao L, Tian M, Chen Z, Zang H. Rapid screening of esophageal squamous cell carcinoma by near-infrared spectroscopy combined with aquaphotomics. Talanta 2025; 285:127399. [PMID: 39708567 DOI: 10.1016/j.talanta.2024.127399] [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: 08/12/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Esophageal cancer (EC), the fifth most common cause of cancer-related mortality in China, poses a significant threat to public health. Among the pathological types, esophageal squamous cell carcinoma (ESCC) is predominant, comprising approximately 90 % of cases. Screening is crucial for early detection, diagnosis and treatment, thereby reducing ESCC mortality. This study aimed to develop a rapid, accurate, and cost-effective method based on near-infrared (NIR) spectroscopy combined with aquaphotomics for ESCC screening. NIR spectra were obtained from plasma samples of both healthy controls and ESCC patients. Subsequently, principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA) were utilized to identify the water matrix coordinates (WAMACS), thereby delineating the water absorption spectrum pattern (WASP) and constructing an aquagram. The results showed that the PLS-DA screening test model demonstrated high accuracy and precision rates of 95.12 % and 97.10 %, respectively, along with sensitivity and specificity rates of 97.10 % and 84.62 %. The area under the curve (AUC) achieved 0.9064. Aquaphotomic analysis revealed that the WASP of the healthy group predominantly exhibited strong absorption in regions indicative of strong hydrogen bonds (1460 nm, 1480 nm, 1494 nm), while the WASP of the ESCC group showed strong absorption in regions associated with strong hydrogen bonds, weak hydrogen bonds and free water, especially the regions of weak hydrogen bonds (1434 nm) and free water (1390 nm) were significantly different from those of the healthy group. The findings indicated that the rapid screening model for ESCC, integrating NIR spectroscopy with aquaphotomics, is both effective and feasible, with the WASP presenting as a potentially valuable biomarker for ESCC screening.
Collapse
Affiliation(s)
- Qingqing Lu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lian Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Shandong Engineering Research Center for Transdermal Drug Delivery Systems, Jinan, Shandong, 250000, China
| | - Wenyan Liang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoning Xu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jing Zhu
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
| | - Xiaobo Ma
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Weilu Tian
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Lele Gao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Mengyin Tian
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhongjian Chen
- Experimental Research Center, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Hengchang Zang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China; Shandong Engineering Research Center for Transdermal Drug Delivery Systems, Jinan, Shandong, 250000, China; National Glycoengineering Research Center, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
6
|
Chen Y, Zhang D, Li J, Sun Y, Wang J, Xi L. SNS‑032 combined with decitabine induces caspase‑3/gasdermin E‑dependent pyroptosis in breast cancer cells. Oncol Lett 2025; 29:202. [PMID: 40070781 PMCID: PMC11894506 DOI: 10.3892/ol.2025.14948] [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: 09/05/2024] [Accepted: 01/13/2025] [Indexed: 03/14/2025] Open
Abstract
SNS-032 is a synthetic compound that specifically inhibits cyclin-dependent kinases 2, 7 and 9. Its primary anticancer activity involves cell cycle arrest, which prevents tumor cell growth. However, there are limited reports on whether SNS-032 induces pyroptosis, a novel inflammation-mediated programmed cell death pathway in breast cancer (BC). The present study demonstrated that SNS-032 treatment decreased cell viability by inducing pyroptosis in BC cells. Typical morphological indications of pyroptosis were observed, including cell swelling and destruction of cell membrane integrity, leading the release of adenosine 5'-triphosphate and lactate dehydrogenase. Furthermore, the expression of caspase-3, the N terminus of gasdermin E (GSDME) and B-cell lymphoma-2 (BCL-2)-associated X protein increased, whereas expression of BCL-2 decreased. In addition, Z-DEVD-FMK, a specific caspase-3 inhibitor, markedly alleviated pyroptosis triggered by SNS-032. These findings suggested that SNS-032 induced caspase-3/GSDME-dependent pyroptosis. Furthermore, the present study demonstrated that decitabine (DAC), a DNA methyltransferase inhibitor, upregulated the expression of GSDME protein and enhanced SNS-032-induced caspase-3/GSDME-dependent pyroptosis in BC cells. In conclusion, these results suggest that caspase-3/GSDME-induced pyroptosis can be facilitated by SNS-032 treatment in BC cells, and DAC has the potential to enhance SNS-032-induced pyroptosis by increasing GSDME expression. This mechanistic insight indicates that SNS-032 is a promising therapeutic agent for BC treatment.
Collapse
Affiliation(s)
- Yuxin Chen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Danya Zhang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jie Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yue Sun
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jing Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ling Xi
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
7
|
Lan J, Wang H, Huang J, Li W, Ao M, Zhang W, Mu J, Yang L, Ran L. MoLPre: A Machine Learning Model to Predict Metastasis of cT1 Solid Lung Cancer. Clin Transl Sci 2025; 18:e70186. [PMID: 40143527 PMCID: PMC11947056 DOI: 10.1111/cts.70186] [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: 09/04/2024] [Revised: 12/29/2024] [Accepted: 01/10/2025] [Indexed: 03/28/2025] Open
Abstract
Given that more than 20% of patients with cT1 solid NSCLC showed nodal or extrathoracic metastasis, early detection of metastasis is crucial and urgent for improving therapeutic planning and patients' risk stratification in clinical practice. This study collected clinicopathological variables from the pulmonary nodule and lung cancer database of the First Affiliated Hospital of Chongqing Medical University, where patients with early-stage (cT1) solitary lung cancer were evaluated from 2018.11 to 2022.10. The random forest model and Shapley Additive Explanations (SHAP) were used to investigate the importance of clinical features in the feature selection part. Random Forest, Gradient Boosting, and AdaBoost classifiers were applied to build the final model, and the predictive discrimination of each model was compared based on the receiver operating characteristics (ROC) curve and precision and recall curve. With the evaluation of feature importance, 9 features were used to construct the prediction model finally. The Random Forest model yielded an average precision of 0.93 with an area under the curve (AUC) of 0.92 (95% CI: 0.88-0.94) compared with the Gradient Boosting and AdaBoost classifiers in the internal validation dataset, yielding an average precision of 0.87 and 0.91 with AUCs of 0.87 (95% CI: 0.84-0.93) and 0.90 (95% CI: 0.86-0.92), respectively. In addition, the Random Forest classifier performed best in 5 other 5 diagnostic indices. Furthermore, we embedded this model in a web application called MoLPre (https://molpre.cqmu.edu.cn/), a user-friendly tool assisting in the metastasis prediction of cT1 solid lung cancer.
Collapse
Affiliation(s)
- Jie Lan
- Department of BioinformaticsThe Basic Medical School of Chongqing Medical UniversityChongqingChina
| | - Heng Wang
- Department of BioinformaticsThe Basic Medical School of Chongqing Medical UniversityChongqingChina
| | - Jing Huang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Weiyi Li
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Min Ao
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Wanfeng Zhang
- Department of BioinformaticsThe Basic Medical School of Chongqing Medical UniversityChongqingChina
| | - Junhao Mu
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Li Yang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Longke Ran
- Department of BioinformaticsThe Basic Medical School of Chongqing Medical UniversityChongqingChina
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
8
|
Cárdenas-Garzón K, Jurado D, Coronell K, Florez-Lozano K, Arias-Ortiz N, Bravo LM, Uribe-Perez C, Navarro-Lechuga E, Sanchez GI. Determinants of survival in women diagnosed with breast cancer between 2008 and 2017: An analysis of a cohort using data from four Population-Based Cancer Registries of Colombia. Cancer Epidemiol 2025; 95:102765. [PMID: 40020377 DOI: 10.1016/j.canep.2025.102765] [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: 09/10/2024] [Revised: 12/17/2024] [Accepted: 01/29/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Breast Cancer (BC) is the leading cause of cancer-related mortality in women, especially in low- and middle-income countries. Population-Based Cancer Registries (PBCRs) play a crucial role in monitoring cancer trends and guiding evaluation and planning of cancer control programs. In Colombia, there are no national analyses of BC survival. The aim was to estimate the overall survival up to 5-year of women diagnosed with BC, as well as its determinants, in Colombia using population-based data from four Colombian PBCRs. METHODS We conducted a cohort study with women diagnosed with invasive BC between 2008 and 2017, identified by the corresponding PBCRs as residents of the Colombian cities of Barranquilla, Bucaramanga metropolitan area, Manizales, and Pasto. We performed follow-up up to 5 years after the BC diagnosis, or until death (all-cause). We estimated the overall survival (Kaplan Meier). We evaluated the simultaneous effect of multiple risk factors on death risk using Cox proportional hazards analysis, obtaining adjusted Hazard Ratios (aHR) and Confidence Intervals (CI). RESULTS The analysis cohort included 8020 BC cases. The observed overall survival was 72.5 %. The likelihood of 5-year survival was lowest for women aged 70 or older (aHR 1.61; 95 % CI 1.42-1.83), those living in a middle Socioeconomic Stratum (SES) (aHR 1.32; 95 % CI 1.05-1.66), those affiliated to the subsidized Health Insurance Regime (HIR) (aHR 1.47; 95 % CI 1.32-1.63), and those diagnosed in stages III-IV (aHR 2.29; 95 % CI 2.03-2.57) compared to women with a diagnosis age between 50 and 70 years, residents in high SES, those affiliated to the contributory HIR, and those diagnosed at stages I-II, respectively. CONCLUSION Social disparities are linked to BC survival in Colombia, likely due to limited access to healthcare services. This suggests the importance of strengthening screening and diagnostic care, especially for vulnerable populations.
Collapse
Affiliation(s)
| | - Daniel Jurado
- Group Infection and Cancer, Universidad de Antioquia, Medellín, Colombia
| | - Karen Coronell
- Department of Mathematics and Statistics, Universidad del Norte, Barranquilla, Colombia
| | - Karen Florez-Lozano
- Department of Mathematics and Statistics, Universidad del Norte, Barranquilla, Colombia
| | - Nelson Arias-Ortiz
- Population-Based Cancer Registry of Manizales, Instituto de Investigaciones en Salud, Universidad de Caldas, Manizales, Colombia
| | - Luisa M Bravo
- Population-Based Cancer Registry of Pasto, Grupo de investigación Salud Pública, Universidad de Nariño, Pasto, Colombia
| | - Claudia Uribe-Perez
- Population-Based Cancer Registry of Bucaramanga, Universidad Autónoma de Bucaramanga, Bucaramanga, Colombia
| | - Edgar Navarro-Lechuga
- Population-Based Cancer Registry of Barranquilla, Departamento de Salud Pública, Universidad del Norte, Barranquilla, Colombia.
| | - Gloria I Sanchez
- Group Infection and Cancer, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
9
|
He F, Chen X, Zhu Y, Pang H, Li Z, Liang P, Jin T, Chen Z, Chen Z, Hu J, Yang K. Transforming Growth Factor Beta2 Promotes Migration and Inhibits the Proliferation of Gastric Cancer Cells by Regulating the pSmad2/3-NDRG1 Signaling Pathway. MedComm (Beijing) 2025; 6:e70148. [PMID: 40151835 PMCID: PMC11949502 DOI: 10.1002/mco2.70148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/25/2025] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
Transforming growth factor beta2 (TGFβ2) is upregulated in gastric cancer (GC), playing a crucial role in driving its progression. However, the biological effects of TGFβ2 in GC metastasis and proliferation remain not fully understood. Our study reveals that TGFβ2 enhances N-myc downstream-regulated gene 1 (NDRG1) protein expression by activating the TGFβR/Smad2/3-dependent pathway, accelerating GC progression. TGFβ2 knockdown downregulates NDRG1 by inhibiting the TGFβR/Smad2/3 signaling pathway, which in turn inhibits GC cell migration and epithelial-mesenchymal transition (EMT) but stimulates proliferation. Both TGFβ2 upregulation and NDRG1 upregulation enhance GC cell migration in vitro and promote lung metastasis in mouse models. Interfering with NDRG1 reverses TGFβ2-induced migration, and inhibiting Smad2/3 or TGFβR reverses TGFβ2-induced NDRG1 upregulation and GC cell migration. Clinical sample analysis shows high TGFβ2 and NDRG1 expression in GC, associated with poor prognosis. Our study reveals that TGFβ2 upregulates NDRG1 via the TGFβR/Smad2/3 pathway, driving GC progression and highlighting the potential role of the TGFβ2NDRG1 axis in GC-targeted therapies.
Collapse
Affiliation(s)
- Feng‐Jun He
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
- Department of Thyroid and Breast SurgeryWest China School of Public Health and West China Fourth HospitalChengduChina
| | - Xiao‐Long Chen
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
| | - Yun‐Feng Zhu
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
| | - Hua‐Yang Pang
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Ze‐Dong Li
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
| | - Pan‐Ping Liang
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
| | - Tao Jin
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
- West China School of MedicineWest China HospitalSichuan UniversityChengduChina
| | - Zheng‐Wen Chen
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
| | - Ze‐Hua Chen
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
- West China School of MedicineWest China HospitalSichuan UniversityChengduChina
| | - Jian‐Kun Hu
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
| | - Kun Yang
- Department of General Surgery & Laboratory of Gastric CancerState Key Laboratory of BiotherapyCollaborative Innovation Center of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduChina
- Gastric Cancer CenterWest China Hospital,Sichuan UniversityChengduChina
| |
Collapse
|
10
|
Li J, Zheng T, Xu Y, Tian M, Shi L, Chen J, Li T, Zhang Z. ADAM17 promotes colorectal cancer migration and invasion by regulating the TGF-β/Smad signaling pathway. Tissue Cell 2025; 93:102648. [PMID: 39644618 DOI: 10.1016/j.tice.2024.102648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE Investigate ADAM17 expression in colorectal cancer (CRC) at molecular and cellular levels and its potential mechanism in promoting tumorigenesis by regulating CRC cell migration and invasion. MATERIALS AND METHODS The study measured ADAM17 mRNA and protein levels in colorectal cancer cells and tissues using qPCR and immunohistochemical staining, and assessed the cells' proliferation, migration, and invasion abilities. RESULTS ADAM17 expression was significantly higher in CRC tissues than in non-cancerous tissues and was linked to metastasis and poor prognosis in CRC patients. Silencing ADAM17 reduced cell migration and invasion. Mechanistically, knocking down ADAM17 decreased the expression of TGF-β/Smad pathway-related proteins, which inhibited proteins associated with migration and invasion, thus impairing these cellular processes. CONCLUSION ADAM17 likely promotes the migration and invasion of CRC cells by regulating the TGF-β/Smad signaling pathway. This study aids in understanding the molecular mechanisms of CRC metastasis and development, and supports the development of new therapeutic targets.
Collapse
Affiliation(s)
- Jiaming Li
- Department of Clinical Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Tingjin Zheng
- Department of Clinical Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Yingzhi Xu
- Department of Clinical Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Mengcha Tian
- Department of Clinical Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Liangpan Shi
- Departmenof Gastrointestinal Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Jintu Chen
- Department of Clinical Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Tian Li
- Tianjin Medical University, Tianjin 300102, China
| | - Zhishan Zhang
- Department of Clinical Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China.
| |
Collapse
|
11
|
Pan W, Han Y, Zhang M, Zhu K, Yang Z, Qiu M, Guo Y, Dong Z, Hao J, Zhang X, Gao M, Zhang H. Effects of microplastics on chemo-resistance and tumorigenesis of colorectal cancer. Apoptosis 2025; 30:1005-1020. [PMID: 39924586 DOI: 10.1007/s10495-025-02085-1] [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] [Accepted: 01/24/2025] [Indexed: 02/11/2025]
Abstract
Microplastics (MPs) are widely distributed environmental pollutants around the world. Although studies have demonstrated that MPs have adverse effects on human health, the relationship between MPs and tumors remains unclear. The gut is the main site of microplastics absorption, and the function of MPs in the chemoresistance and progression of colorectal cancer (CRC) needs more investigation. Here, we show that MPs exist in human CRC tissues for the first time by using a laser direct infrared chemical imaging system. MPs can cause an increase in CRC incidence in animal models and promote resistance to oxaliplatin. It is illustrated that the uptake of MPs enhances levels of autophagy by activating the mTOR pathway. MPs can also promote the disorder of intestinal flora and intestinal inflammation, serving as an essential component in the onset and advancement of CRC. These results indicated that microplastic pollutants in colorectal cancer could mediate protective autophagy through the mTOR/ULK1 axis, which is one of the new reasons for chemo-resistance in CRC under the background of increasingly serious microplastics pollution. This study identified the adverse effects of MPs on colorectal cancer progression and chemotherapy prognosis, and attempted to block the intake of MPs to propose a novel approach for clinical precision treatment.
Collapse
Affiliation(s)
- Wen Pan
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China
| | - Yueting Han
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China
| | - Mingqing Zhang
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China
| | - Kegan Zhu
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China
| | - Zhen Yang
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China
| | - Minghan Qiu
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China
| | - Yaoyang Guo
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical ResearchCenter for Cancer, Tianjin, China, 300060
| | - Ziyi Dong
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical ResearchCenter for Cancer, Tianjin, China, 300060
| | - Jie Hao
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China.
| | - Xipeng Zhang
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China.
| | - Ming Gao
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China.
| | - Haiyang Zhang
- Tianjin Institute of Coloproctology,Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300121, China.
| |
Collapse
|
12
|
Jensen‐Marini E, Ayton D, Zalcberg J, Stirling RG. Exploring patient reported quality of life in lung cancer patients: A qualitative study of patient-reported outcome measures. Asia Pac J Clin Oncol 2025; 21:163-173. [PMID: 38520667 PMCID: PMC11880980 DOI: 10.1111/ajco.14056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 03/25/2024]
Abstract
OBJECTIVE Lung cancer is the leading cause of cancer-related death globally and provides a major disease burden likely to substantially impact quality of life (QoL). Patient-reported outcome measures (PROMs) have been identified as effective methods of evaluating patient QoL. Existing lung cancer-specific PROMs however have uncertain utility and minimal patient involvement in their design and development. This qualitative study aimed to evaluate the patient perspective of existing PROMs and to explore their appropriateness for population-based descriptions of lung cancer-related QoL. METHODS A descriptive qualitative study was conducted consisting of semi-structured interviews with 14 patients recruited from the Victorian Lung Cancer Registry and Alfred Hospital using purposive sampling. Interviews first explored the factors most important to lung cancer patients QoL, and second, patient's perspectives on the appropriateness of existing PROMs. Thematic analysis was used to develop themes, and content analysis was conducted to determine PROM acceptability. RESULTS Five novel themes were identified by patients as being important impacts on QoL: Personal attitude toward the disease is important for coping; independence is valued; relationships with family and friends are important; relationships with treating team are meaningful; personal and public awareness of lung cancer is limited. These patient-identified impacts are poorly covered in existing lung cancer-specific PROMs. Patients welcomed and appreciated the opportunity to complete PROMs; however, they identified problems with existing PROMs relevance, tone, and formatting. CONCLUSION Existing lung cancer PROMs poorly reflect the five themes identified in this study as most important to lung cancer patients QoL. This study reaffirms the need to review existing PROMs to ensure utility and construct validity. Future PROM development must engage key patient-generated themes and evolve to reflect the changing management and therapeutic landscape.
Collapse
Affiliation(s)
- Elena Jensen‐Marini
- Department of Epidemiology and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Darshini Ayton
- Department of Epidemiology and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - John Zalcberg
- Department of Epidemiology and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Robert G. Stirling
- Central Clinical SchoolFaculty of Medicine Nursing and Health SciencesMonash UniversityMelbourneAustralia
- Department of Respiratory MedicineAlfred HealthMelbourneVictoriaAustralia
| |
Collapse
|
13
|
Xing H, Bi HN, Yin Q, Zhang J, Zhang X, Li YJ, Gong XM, Shi JF. FSP1 expression as a predictor of platinum resistance and recurrence in epithelial ovarian cancer. Anticancer Drugs 2025; 36:338-346. [PMID: 39927929 DOI: 10.1097/cad.0000000000001676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
The objective of this study is to assess the differential expression of ferroptosis suppressor protein 1 (FSP1) in relation to clinical features, platinum resistance, and recurrence in epithelial ovarian cancer (EOC). In addition, the potential significance of FSP1 in EOC as a predictor of platinum resistance and recurrence in EOC was explored. Patients with pathologically confirmed EOC who underwent surgical treatment were included in this analysis. Immunohistochemistry was employed to evaluate FSP1 expression in ovarian tissues, with quantitative analysis performed on the samples. Clinical data were collected during follow-up, and patients were categorized according to platinum resistance and recurrence criteria. Statistical analysis was conducted using SPSS version 27.0. A total of 40 tissue samples from patients with EOC were analyzed, along with 21 samples from benign ovarian tumors and 20 samples from normal ovarian tissues. The expression of FSP1 was significantly higher in the EOC group compared to both benign and normal tissue groups. Meanwhile, the expressions of FSP1 were higher in groups with clinically advanced stages, high-grade carcinoma, presence of cancerous ascites, lymph node metastasis, and in the clear cell EOC group, compared to those with clinically early stages, low-grade carcinoma, absence of cancerous ascites, no lymph node metastasis, and other pathological subtypes. A positive linear correlation was identified between FSP1 expression in EOC tissues and serum levels of CA125 and human epididymis protein 4 at the time of diagnosis. The elevated expression of FSP1 is positively correlated with serum CA125 and human epididymis protein 4 levels at the time of diagnosis, which is a risk factor for EOC drug resistance and recurrence. These findings suggest that FSP1 may serve as a valuable biomarker for predicting platinum resistance and recurrence in patients with EOC.
Collapse
Affiliation(s)
- Hang Xing
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dali University, Dali
| | - Hai-Ning Bi
- Department of AI and Advanced Computing, Xi'an Jiaotong-Liverpool University, Su'zhou, China
| | - Qi Yin
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dali University, Dali
| | - Ji Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dali University, Dali
| | - Xue Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dali University, Dali
| | - Yao-Jiao Li
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dali University, Dali
| | - Xue-Mei Gong
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dali University, Dali
| | - Ji-Fang Shi
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Dali University, Dali
| |
Collapse
|
14
|
Song A, Yang W, Wang J, Cai Y, Cai L, Pang N, Yu R, Liu Z, Yang C, Jiang F. Application of ATR-FTIR spectroscopy and multivariate statistical analysis in cancer diagnosis. SLAS Technol 2025; 31:100253. [PMID: 39900180 DOI: 10.1016/j.slast.2025.100253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Lung cancer is one of the most prevalent and lethal malignant tumors worldwide. Currently, clinical diagnosis primarily relies on chest X-ray examinations, histopathological analysis, and the detection of tumor markers in blood. However, each of these methods has inherent limitations. The current study aims to explore novel diagnostic approaches for lung cancer by employing attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy in conjunction with multiple machine learning models. Fourier transform infrared spectroscopy can detect subtle differences in the material structures that reflect the carcinogenic process between lung cancer tissues and normal tissues. By applying principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA) to analyze infrared spectral data, these subtle differences can be amplified. The study revealed that the combination of spectral bands within the 3500-3000 cm-1 and 1600-1500 cm-1 ranges is particularly significant for differentiating between the two groups. Three classification models-Support Vector Machine (SVM), k-Nearest Neighbor (kNN), and Linear Discriminant Analysis (LDA)-were constructed for spectral analysis of various band combinations. The results indicated that in detecting lung cancer samples, the combination of the 3500-3000 cm-1 and 1600-1500 cm-1 bands offers significant advantages. The analysis of the receiver operating characteristic (ROC) curve demonstrated that the area under the curve (AUC) exceeded 0.95 for all models, with the LDA model achieving an accuracy rate of 99.4% in identifying lung cancer patients compared to healthy individuals. The findings suggest that the integration of ATR-FTIR spectroscopy with multiple machine learning models represents a promising auxiliary diagnostic method for clinical lung cancer diagnosis, enabling detection at the molecular level.
Collapse
Affiliation(s)
- Ao Song
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China; Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China
| | - Wanli Yang
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China
| | - Jun Wang
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China
| | - Yisa Cai
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China; Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China
| | - Lizheng Cai
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Nan Pang
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China
| | - Ruihua Yu
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China
| | - Zhikun Liu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China.
| | - Chao Yang
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China.
| | - Feng Jiang
- Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai 202150, China.
| |
Collapse
|
15
|
Barben J, Galvin A, Kamga AM, Bertrand N, Niogret J, Tisserand J, Quipourt V, Bengrine-Lefevre L, Dabakuyo-Yonli TS. The challenge of ovarian cancer care in the oldest old. Cancer Epidemiol 2025; 95:102697. [PMID: 39505669 DOI: 10.1016/j.canep.2024.102697] [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: 06/21/2024] [Revised: 10/02/2024] [Accepted: 10/31/2024] [Indexed: 11/08/2024]
Abstract
INTRODUCTION Ovarian cancer (OC) is the eighth most common cancer in women, with a poor prognosis, particularly in older women. The aim of this study was to describe an octogenarian population with OC and to examine the differences in net survival (NS) according to age. MATERIAL AND METHODS In this retrospective observational population-based study from a gynecological cancer registry, patients aged > 18 years with an identified epithelial ovarian cancer stage IA to IVB diagnosed between 1998 and 2018 were included. Patients with non-available FIGO stage were excluded. Patients were stratified into three age groups: <70, 70-79 and ≥80 years, then by OC stage (FIGO I-II, IIIA-IIIB, IIIC-IV). Sociodemographic and cancer-related variables were compared using univariate test (Khi²). The 5-year NS was calculated using the Pohar-Perme method. RESULTS Among the 721 patients included: 462 (64.1 %) were younger than 70 years, 176 (24.4 %) were aged between 70 and 79 years, and 83 (11.85 %) were aged 80 years or older. Patients ≥80 years had a trend for lower rate of serous carcinoma than the other age subgroups. As age increased, patients were less likely to undergo surgery and chemotherapy. While 73 % of women <70 years received a combination of surgery and chemotherapy, the rate was 62 % among women 70-79 years and 27 % among women ≥80 years (p<.0001). When focusing on FIGO IIIC-IV stages, the 5-year NS rate for women <70 years was 45.1 % (95 % CI 39.1-52.0). For women 70-79 years, it was 25.9 % (95 % CI 18.6-36.1), and for those ≥80 years, it was 19.5 % (95 % CI 10.0-38.0) (p<.005). DISCUSSION The oldest patients had less optimal treatment and a lower NS compared to patients in their seventies or younger. Frailty should be carefully assessed to optimize care in the oldest patients with OC.
Collapse
Affiliation(s)
- Jérémy Barben
- Department of Geriatrics, Hospital of Champmaillot, Dijon University Hospital, Dijon, France; Geriatric Oncology Coordination Unit in Burgundy, Hospital of Champmaillot, Dijon University Hospital, France; Côte d'Or Breast and Gynecological Cancer Registry, George-François Leclerc Cancer Centre, Dijon, France; Epidemiology and Quality of Life Research Unit, INSERM U1231, George-François Leclerc Cancer Centre, Dijon, France; French Society of Geriatric Oncology (SoFOG), France.
| | - Angéline Galvin
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, Epicene Team, UMR 1219, Bordeaux, France; French Society of Geriatric Oncology (SoFOG), France
| | - Ariane Mamguem Kamga
- Côte d'Or Breast and Gynecological Cancer Registry, George-François Leclerc Cancer Centre, Dijon, France; Epidemiology and Quality of Life Research Unit, INSERM U1231, George-François Leclerc Cancer Centre, Dijon, France
| | - Nicolas Bertrand
- Department of Medical Oncology, Eugène Marquis Cancer Centre, Rennes, France; French Society of Geriatric Oncology (SoFOG), France
| | - Julie Niogret
- Geriatric Oncology Coordination Unit in Burgundy, Hospital of Champmaillot, Dijon University Hospital, France; Department of Medical Oncology, George-François Leclerc Cancer Centre, Dijon, France; French Society of Geriatric Oncology (SoFOG), France
| | - Julie Tisserand
- Department of Geriatrics, Poitiers University Hospital, Poitiers, France; University of Poitiers, ProDiCeT, UR 24144, Poitiers, France; French Society of Geriatric Oncology (SoFOG), France
| | - Valérie Quipourt
- Department of Geriatrics, Hospital of Champmaillot, Dijon University Hospital, Dijon, France; Geriatric Oncology Coordination Unit in Burgundy, Hospital of Champmaillot, Dijon University Hospital, France; French Society of Geriatric Oncology (SoFOG), France
| | - Leila Bengrine-Lefevre
- Geriatric Oncology Coordination Unit in Burgundy, Hospital of Champmaillot, Dijon University Hospital, France; Department of Medical Oncology, George-François Leclerc Cancer Centre, Dijon, France; French Society of Geriatric Oncology (SoFOG), France
| | - Tienhan Sandrine Dabakuyo-Yonli
- Côte d'Or Breast and Gynecological Cancer Registry, George-François Leclerc Cancer Centre, Dijon, France; Epidemiology and Quality of Life Research Unit, INSERM U1231, George-François Leclerc Cancer Centre, Dijon, France
| |
Collapse
|
16
|
Bahardoust M, Torabi S, Yarahmadi D, Kakoienejad MM, Abbasi F, Shamohammadi M, Haghmoradi M, Goodarzy B, Tizmaghz A. Impact of negative lymph node removal on survival in esophageal cancer: a systematic review and meta-analysis. BMC Surg 2025; 25:124. [PMID: 40155975 PMCID: PMC11951649 DOI: 10.1186/s12893-025-02858-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Recent studies have reported that a high number of negative lymph nodes (NLNs) removed can be associated with improved survival in esophageal cancer( EC) after surgery; however, the effect size of a high number of removed NLNs on survival rates has been reported to vary, which may be due to the small sample size of early studies. This meta-analysis aimed to evaluate the effect of the high number of NLNs removed on the survival rate of patients with EC after surgery. METHODS We searched PubMed, Embase, Scopus, Web of Science, and Google Scholar databases with relevant Mesh terms to find studies that investigated the effect of the number of NLNs resected on the survival of EC patients after surgery until February 17, 2025. This systematic review was conducted based on the PRISMA 2020 checklist. Cochran's I2 was used to evaluate heterogeneity between studies. Publication bias was evaluated using the Egger test. Heterogeneity between studies was controlled by meta-regression. Finally, eight studies involving 5,521 EC patients were included. RESULTS The survival rate in patients whose number of removed NLNs ≥ 19 was significantly better than those with removed NLNs < 19 (HR: 0.88, 95% CI: 0.81, 0.95, I2 = 84.4). Subgroup analysis of 8 studies showed that the protective effect of the high number of removed NLNs) ≥ 19 (was greater in adenocarcinoma patients than in SCC (Pooled HR: 0.63 vs. 0.88). CONCLUSION The high number of NLNs removed (≥ 19) during surgery was associated with improved survival after surgery, especially in patients with adenocarcinoma. Removing ≥ 19 NLNs significantly improves survival in EC patients, particularly those with adenocarcinoma. This threshold should be incorporated into surgical guidelines.
Collapse
Affiliation(s)
- Mansour Bahardoust
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Danyal Yarahmadi
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Abbasi
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Meisam Haghmoradi
- Department of Orthopedic Surgery, Urmia University of Medical Sciences, Urmia, Iran
| | - Babak Goodarzy
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Adnan Tizmaghz
- Firoozabadi Clinical Research Development Unit (F A CRD U), Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
17
|
Li C, Zhong S, Chen J, Mu X. TsRNA-49-73-Glu-CTC: A promising serum biomarker in non-small cell lung cancer. PLoS One 2025; 20:e0320187. [PMID: 40153423 PMCID: PMC11952254 DOI: 10.1371/journal.pone.0320187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/15/2025] [Indexed: 03/30/2025] Open
Abstract
OBJECTIVE Lung cancer has the highest incidence and mortality rates globally, with the majority of cases classified as non-small cell lung cancer (NSCLC). Due to the absence of specific tumor biomarkers, most lung cancer cases are diagnosed at an advanced stage. Therefore, the identification of novel molecular biomarkers with high sensitivity and specificity for early diagnosis is deemed crucial for enhancing the treatment of NSCLC. Transfer RNA-derived small RNA (tsRNA) is closely associated with malignant tumors and holds promise as a potential biomarker for tumor diagnosis. This study aimed to investigate whether serum tsRNA could serve as a biomarker for NSCLC. METHODS Differentially expressed tsRNAs were identified through high-throughput sequencing of serum samples obtained from patients with NSCLC and healthy individuals. Additional serum samples were collected for validation using Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR). The diagnostic performance of these tsRNAs was assessed through Receiver Operating Characteristic (ROC) Curve Analysis. Furthermore, preliminary functional exploration was undertaken through cell experiments. RESULTS tsRNA-49-73-Glu-CTC is highly expressed in the serum of patients with NSCLC and demonstrates superior diagnostic value compared to commonly used tumor markers in clinical practice, such as Carcinoembryonic Antigen (CEA), Neuron-Specific Enolase (NSE), and Cytokeratin 19 Fragment (CYFRA). A combined diagnostic approach enhances the accuracy of NSCLC detection. Additionally, tsRNA-49-73-Glu-CTC is highly expressed in A549 cells, and transfection with a tsRNA-49-73-Glu-CTC inhibitor significantly reduces both proliferation and migration capabilities. CONCLUSIONS tsRNA-49-73-Glu-CTC has the potential to serve as a novel molecular diagnostic biomarker for NSCLC and plays a significant role in the biological processes associated with NSCLC proliferation and migration.
Collapse
Affiliation(s)
- Chenyu Li
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
- Medical Laboratory, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Shenjie Zhong
- Medical Laboratory, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Juan Chen
- Medical Laboratory, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Xiaofeng Mu
- Medical Laboratory, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| |
Collapse
|
18
|
Sun D, Cui X, Yang W, Wei M, Yan Z, Zhang M, Yu W. Simvastatin inhibits PD-L1 via ILF3 to induce ferroptosis in gastric cancer cells. Cell Death Dis 2025; 16:208. [PMID: 40140647 PMCID: PMC11947124 DOI: 10.1038/s41419-025-07562-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
The treatment of gastric cancer remains challenging, with immunotherapy serving as a critical component of the holistic approach to its treatment. The results of this study indicated that statins could decrease the serum levels of interleukin-enhancing binding factor 3 (ILF3) and programmed cell death ligand 1(PD-L1) in GC patients and improve their prognosis. Functional experiments demonstrated that simvastatin induced ferroptosis by inhibiting ILF3 in GC cells and enhanced the killing effect of activated CD8+ T cells on GC cells. The CUT&Tag assay revealed that, mechanistically, simvastatin inhibited ILF3 expression by reducing the acetylation level at residue site H3K14 in ILF3. Next-generation sequencing and Kyoto Encyclopedia of Genes and Genomes analysis revealed that ILF3 regulated PD-L1 expression through the DEPTOR/mTOR signaling pathway. Overall, simvastatin induced ferroptosis in GC cells by inhibiting ILF3 expression while promoting the activation of CD8+ T cells to augment antitumor immune responses, thereby facilitating synergistic immunotherapy.
Collapse
Affiliation(s)
- Danping Sun
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Xiaohan Cui
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Wenshuo Yang
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Meng Wei
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Zhibo Yan
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Wenbin Yu
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China.
| |
Collapse
|
19
|
Gan C, Yao S, Zhao J, Shi H, Xu J, Zhang M, Cheng H. Expression of inflammatory states in response to psychological distress in breast cancer survivors and its relationship to subjective memory function complaints. BMC Womens Health 2025; 25:140. [PMID: 40133863 PMCID: PMC11934799 DOI: 10.1186/s12905-025-03674-7] [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: 11/05/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Breast cancer (BC) survivors frequently endure psychological distress following chemotherapy, with subjective memory decline being a prevalent aspect of chemotherapy-related cognitive impairment (CRCI). This study aimed to assess the influence of psychological distress on subjective memory decline in BC survivors with CRCI and investigate potential underlying mechanisms. METHODS A total of 104 BC survivors who had completed chemotherapy were categorized based on the distress thermometer (DT) score into a no-psychological distress group (NPD group, n = 51) and a psychological distress group (PD group, n = 53). The groups were compared using the Mini-Mental State Examination (MMSE), the Prospective and Retrospective Memory (PM and RM) Questionnaire (PRMQ), cytokine levels (of interleukin-1β [IL-1β], tumor necrosis factor-alpha [TNF-α], and IL-4), and inflammatory markers (neutrophil-to- lymphocyte ratio [NLR], platelet-to-lymphocyte ratio [PLR], monocyte count-to-lymphocyte ratio [MLR], granulocyte-to-lymphocyte ratio [GLR], and systemic immune-inflammation index [SII]). Mediation analysis was performed to explore whether cytokine and inflammatory marker levels mediate the effect of psychological distress on subjective memory function complaints. RESULTS The NPD group performed significantly better in the PD group both RM (z = -3.370, p = 0.001) and PM (z = -1.967, p = 0.049). The IL-1β levels were substantially higher in the PD group than in the NPD group (z = -2.920, p = 0.004). Similarly, NLR (z = -2.585, p = 0.010), GLR (z = -2.858, p = 0.004), and SII (z = -2.747, p = 0.006) were higher in the PD group. Mediation analysis revealed that IL-1β partially mediated the relationship between DT and RM (β = 0.019, p = 0.007), while SII fully mediated the relationship between DT and PM (β = 0.003, p = 0.017). CONCLUSION BC survivors experiencing psychological distress exhibited worse subjective memory and elevated levels of IL-1β, NLR, GLR, and SII. These findings suggest that inflammation may be a cause of subjective memory function complaints in BC survivors with psychological distress.
Collapse
Affiliation(s)
- Chen Gan
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Medical University, Hefei, Anhui, China
- Shenzhen Clinical Medical School of Southern Medical University, Shenzhen, Guangdong, China
| | - Senbang Yao
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Medical University, Hefei, Anhui, China
| | - Jingjing Zhao
- Department of Oncology, Hangzhou Mingzhou Hospital, Hangzhou, Zhejiang, China
| | - Huangyuxin Shi
- Music College, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Jian Xu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Medical University, Hefei, Anhui, China
| | - Mingjun Zhang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
- Anhui Medical University, Hefei, Anhui, China.
| | - Huaidong Cheng
- Shenzhen Clinical Medical School of Southern Medical University, Shenzhen, Guangdong, China.
- Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China.
| |
Collapse
|
20
|
Teodózio CGC, Gomes TVFDSC, Fernandes LAPP, de Lucena RN, da Silva JMP, Aguiar SSD, Thuler LCS, Bergmann A. Lower physical activity and functional capacity after breast cancer neoadjuvant chemotherapy. BMJ Support Palliat Care 2025:spcare-2024-005358. [PMID: 40121022 DOI: 10.1136/spcare-2024-005358] [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: 12/26/2024] [Accepted: 02/12/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE This study aimed to assess the levels of physical activity (PA) and functional capacity of breast cancer patients before and after neoadjuvant chemotherapy. METHODS The investigation comprises a prospective cohort study including women 18 years or older who underwent neoadjuvant chemotherapy for breast cancer at a single oncology centre between 4 April 2016 and 31 October 2018. Patients were evaluated in terms of self-reported PA levels and physical capacity using the following physical tests: handgrip strength, sitting and standing up in 30 s and stationary walking for 2 min. Variables were compared before and after neoadjuvant chemotherapy using the Wilcoxon test. RESULTS A total of 440 women were analysed, 88.2% of whom presented at an advanced clinical stage. The mean age of the patients was 51.0 years (±11.0), 68.0% were non-white and 53.9% were not working at the time of diagnosis. Decreased PA levels and handgrip strength (p<0.002) were observed following chemotherapy, although no changes in aerobic capacity and lower limb resistance (LLR) were noted (p=0.595 and p=0.163, respectively). CONCLUSION Women with breast cancer exhibit decreased PA levels and handgrip strength following neoadjuvant chemotherapy, although no alterations in aerobic capacity and LLR were observed at the end of treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anke Bergmann
- Instituto Nacional do Câncer, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Kelly SA, Ronan KE, Zameer M, Brown J, Johnston G, Adams R, Murphy D, Kelly D, Darwish W, McCaffery J, Coyne GO, Harrold E, Iqbal S, Cowzer D, Duffy AG. An analysis of compassionate access programmes for novel oncology drugs. Ir J Med Sci 2025:10.1007/s11845-025-03930-7. [PMID: 40117035 DOI: 10.1007/s11845-025-03930-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Despite the rise in the number of approved novel oncology drugs, just over half of all new cancer medicines approved by the EMA between 2017 and 2021 were granted reimbursement in Ireland by the HSE. Compassionate access programmes (CAPs) are a means of providing managed access to drugs which are of proven benefit but have not yet received full approval for reimbursement by the state, or where the requested indication has not been yet been authorised/licensed. METHODS A retrospective review was performed of patients attending The Mater hospital for treatment of advanced malignancy who availed of a CAP between August 2012 and July 2022. Clinical data collected included disease type, treatment received, duration of treatment received, and best response to treatment. To categorize outcome "Clinical Benefit" was defined as a radiological complete, partial, or stable disease response to treatment. RESULTS One hundred and thirteen patients were included in the study. Ninety-three received at least one dose of CAP treatment. Treatment duration ranged from 0 to 112 months, with 12 patients on treatment for ≥ 2 years. N = 47 (42%) experienced a Clinical Benefit. Of these, N = 7 experienced a complete response [CR]. Thirty patients (27%) did not receive a planned treatment or died within 3 months of treatment. CONCLUSIONS In this review of a decade of CAPs at our institution we observed that a significant proportion of patients derived a clinical benefit from CAP treatment. Unfortunately, however, a significant proportion of patients did not receive a planned treatment due to disease progression or died within 3 months of treatment suggesting availability came too late. While CAPs can provide meaningful benefit, they are not a substitute for timely approval of novel agents.
Collapse
Affiliation(s)
- Sarah A Kelly
- School of Medicine, University College Dublin, Dublin, Ireland.
| | - Karine E Ronan
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Mohammed Zameer
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Jennifer Brown
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Grainne Johnston
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Ruth Adams
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Dearbhla Murphy
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Deirdre Kelly
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Waseem Darwish
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - John McCaffery
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | | | - Emily Harrold
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Shahid Iqbal
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Darren Cowzer
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Austin G Duffy
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland
- University College Dublin Centre in Translational Oncology, Dublin, Ireland
| |
Collapse
|
22
|
Chen Z, Fang Y, Zhong S, Lin S, Yang X, Chen S. ITGB5 is a prognostic factor in colorectal cancer and promotes cancer progression and metastasis through the Wnt signaling pathway. Sci Rep 2025; 15:9225. [PMID: 40097546 PMCID: PMC11914080 DOI: 10.1038/s41598-025-93081-7] [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: 04/15/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Integrin beta5 (ITGB5) expression levels are dysregulated in a variety of cancers. However, the mechanism and clinical value of ITGB5 in colorectal cancer (CRC) remain unclear. The Gene Expression Omnibus (GEO) database, real-time PCR, Western blotting and immunohistochemistry were utilized to evaluate ITGB5 expression levels in CRC tissue. Clinical data from the GEO database were obtained to further explore the associations of ITGB5 with clinical features and patient survival. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and gene set enrichment analysis (GSEA) were performed to explore the functions and signaling pathways of ITGB5. In addition, ITGB5 expression was inhibited by siRNA, and the roles of ITGB5 in SW480 and RKO cell growth, migration and invasion, as well as in the Wnt/β-catenin signaling pathway, were investigated. Pancancer studies have shown that ITGB5 is highly expressed in a variety of cancers. Moreover, ITGB5 expression is significantly increased in CRC tissues and is correlated with TNM stage, invasion depth, lymph node metastasis and distant metastasis stage. Kaplan-Meier analysis and meta-analysis of the GSE39582 and GSE17538 datasets indicated that a high level of ITGB5 is a high risk factor for overall survival (OS) and disease-free survival (DFS). In addition, receiver operating characteristic (ROC) curve analysis revealed the value of ITGB5 in predicting DFS, and univariate and multivariate analyses showed that ITGB5 may be an independent prognostic factor for DFS. GO and KEGG analyses indicated that many GO terms related to the extracellular matrix (ECM), focal adhesion and ECM-receptor interaction pathways were enriched. GSEA revealed focal adhesion, cancer pathways, ECM-receptor interactions and Wnt signaling pathways in the samples with high ITGB5 expression. Correlation analysis revealed that high ITGB5 expression is significantly correlated with the TGF-β/EMT pathway and WNT targets. Silencing of ITGB5 inhibited SW480 and RKO cell proliferation, invasion and migration. Mechanistically, downregulated ITGB5 expression blocked the Wnt/β-catenin signaling pathway and epithelial-mesenchymal transition (EMT) in CRC cells. Moreover, ITGB5 expression was related to M0 macrophages, M2 macrophages, neutrophils and plasma cell fractions. ITGB5 may be associated with poor prognosis and metastasis in patients with CRC. ITGB5 may hold promise as a prognostic biomarker and a new potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Gastrointestinal Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, No. 66, Jintang Road, Jianxin Town, Cangshan District, Fuzhou, 350002, Fujian, China
| | - Yuan Fang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
| | - Shuwu Zhong
- Intensive Care Unit (ICU), The Second Affiliated Hospital of University of South China, No. 35 Jiefang Avenue, Zhengxiang District, Hengyang, 421001, Hunan, China
| | - Suyong Lin
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Gastrointestinal Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, No. 66, Jintang Road, Jianxin Town, Cangshan District, Fuzhou, 350002, Fujian, China
| | - Xiaoyu Yang
- School of Basic Medicine Sciences, Fujian Medical University, No. 1, Xuefu North Road, Minhou County, Fuzhou, 350122, China.
| | - Shaoqin Chen
- Department of Gastrointestinal Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, No. 66, Jintang Road, Jianxin Town, Cangshan District, Fuzhou, 350002, Fujian, China.
| |
Collapse
|
23
|
Ryding HG, Rigby RR, Johnston EA, Kruger R, Mitchell LJ. Dietitians' practices and perspectives of the delivery of nutritional care to cancer survivors in the primary care setting. Support Care Cancer 2025; 33:290. [PMID: 40095197 PMCID: PMC11913905 DOI: 10.1007/s00520-025-09330-y] [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: 10/22/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE The number of people living longer after a cancer diagnosis is increasing. Guidelines for cancer survivorship recommend a healthy diet and maintaining a healthy weight post-treatment. While cancer survivors often express the need for professional support for nutrition management, few report seeing a dietitian. This study aimed to explore primary care dietitians' experiences, practices, and perspectives in providing nutritional care to cancer survivors in Australia. METHODS This qualitative study used in-depth, semi-structured interviews with primary care dietitians working in private practice and community care. Interviews were recorded and transcribed. A qualitative descriptive methodological approach integrated with a working analytical framework was utilized for coding and data analysis. RESULTS Twenty-four dietitians working in primary care participated. Four themes and 13 sub-themes were identified: (1) diversity in dietetic practice and cancer-related care interactions; (2) accessing referral pathways and funding sources in a complex healthcare system; (3) the application of nutrition education, and upskilling in cancer care; (4) client barriers and dietitians' challenges and factors influencing confidence in cancer care. CONCLUSION Dietitians in this study highlighted the need for clear referral pathways to primary care particularly as a continuation of cancer-related care following the acute setting. There is a need for tailored support for dietitians supporting people diagnosed with cancer in the primary care setting, including opportunities to upskill in cancer care.
Collapse
Affiliation(s)
- Henriette G Ryding
- School of Health Sciences and Social Work, Griffith University, Gold Coast, QLD, Australia
| | - Roshan R Rigby
- School of Health Sciences and Social Work, Griffith University, Gold Coast, QLD, Australia
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Elizabeth A Johnston
- Viertel Cancer Research Centre, Cancer Council Queensland, Fortitude Valley, QLD, Australia
- School of Exercise and Nutrition Sciences, Queensland University of Technology, Kelvin Grove, QLD, Australia
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Rozanne Kruger
- School of Health Sciences and Social Work, Griffith University, Gold Coast, QLD, Australia
- School of Sport, Exercise and Nutrition, Massey University, Auckland, New Zealand
| | - Lana J Mitchell
- School of Health Sciences and Social Work, Griffith University, Gold Coast, QLD, Australia.
| |
Collapse
|
24
|
Demarchis L, Chiloiro S, Giampietro A, De Marinis L, Bianchi A, Fleseriu M, Pontecorvi A. Cancer screening in patients with acromegaly: a plea for a personalized approach and international registries. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09957-6. [PMID: 40088375 DOI: 10.1007/s11154-025-09957-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/17/2025]
Abstract
Acromegaly is a rare condition, and often diagnosis is delayed by several years, for most patients. Acromegaly is characterized by short and long-term respiratory, cardiovascular and metabolic comorbidities, with possible impact on mortality. In the last two decades, life expectancy has progressively increased in part due to a reduction in biochemically active disease, multidisciplinary treatment approaches and a reduction in complications, and the availability of new drugs. Of note, a leading cause of mortality, cardiovascular comorbidity, has been replaced by cancer(s). As such, neoplasms more frequently observed (colon, thyroid, breast, prostate, and stomach) in patients with acromegaly are receiving increased attention. Chronic exposure to increased growth hormone serum levels may contribute to an increase in the occurrence and progression of cancers. Various efforts have been made to determine the pathogenetic mechanisms involved. However, there are no clear medical-related societal agreement(s) in relation to screening methods or timing regarding neoplasm(s) diagnosis in patients with acromegaly. Additionally, independent and dependent risk factor data in patients with acromegaly is lacking. International/national registries could help lay the groundwork to better study the impact of cancer(s) in patients with acromegaly and subsequently lead to and validate the most appropriate diagnostic and therapeutic path forward.
Collapse
Affiliation(s)
- Luigi Demarchis
- Dipartimento Di Medicina Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Medicina Interna, Endocrinologia E Diabetologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sabrina Chiloiro
- Dipartimento Di Medicina Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
- Dipartimento Di Medicina Interna, Endocrinologia E Diabetologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Antonella Giampietro
- Dipartimento Di Medicina Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Medicina Interna, Endocrinologia E Diabetologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Laura De Marinis
- Dipartimento Di Medicina Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Medicina Interna, Endocrinologia E Diabetologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Bianchi
- Dipartimento Di Medicina Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Medicina Interna, Endocrinologia E Diabetologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Fleseriu
- Pituitary Center, and Departments of Medicine, and Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Alfredo Pontecorvi
- Dipartimento Di Medicina Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Di Medicina Interna, Endocrinologia E Diabetologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
25
|
Han YD, Bae SU, Kim WR, Lim DR, Kim CW. The COVID-19 pandemic and clinical characteristics of colorectal cancer: a multicenter retrospective study. Sci Rep 2025; 15:8903. [PMID: 40087486 PMCID: PMC11909218 DOI: 10.1038/s41598-025-93349-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
The spread of COVID-19 has led to numerous hospitals prioritizing case management and to delays in diagnosis and treatment. Consequently, many cancer patients have developed life-threatening complications during the COVID-19 pandemic. The aim of this study was to investigate the impact of COVID-19 pandemic on colorectal cancer (CRC), including its clinical and pathologic characteristics. This multicenter cohort study was performed at six institutions in Korea and included a total of 3871 patients with CRC treated between March 2019 and February 2021. After exclusion of 211 patients who did not undergo surgery, the data of 3660 patients were compared 1 year before and after the COVID-19 pandemic. The patients' baseline characteristics, CRC-related complications, perioperative outcomes including emergency surgery, R0 resection rates, stoma formations, postoperative complications, and pathologic outcomes were assessed. The number of patients decreased during the pandemic (- 18.0%, from 2127 to 1744), but the baseline characteristics did not differ. The pandemic group had greater disease severity given the presence of bleeding, perforation, and obstruction as complications (9.8% vs. 12.7%, P = 0.033). The proportion of patients who had open surgery (15.9% vs. 17.6%, P = 0.049), stoma formation (11.9% vs. 15.4%, P < 0.001), early postoperative complications (13.5% vs. 17.5%, P = 0.001), and adjuvant chemotherapy increased in the pandemic group (45.5% vs. 50.1%, P = 0.003). The clinical and pathologic features of CRC partly worsened during the pandemic. Healthcare providers and governments should prepare to encounter patients with CRC having poor clinical features for years and encourage people to participate in cancer screening programs. The Clinical Research Information Service (No. KCT0008063), January 2, 2023, retrospectively registered.
Collapse
Affiliation(s)
- Yoon Dae Han
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Uk Bae
- Department of Surgery, School of Medicine, Keimyung University and Dongsan Medical Center, Daegu, Korea
| | - Woo Ram Kim
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Dae Ro Lim
- Department of Surgery, Suncheonhyang University Bucheon Hospital, Suncheonhyang University College of Medicine, Bucheon, Korea
| | - Chang Woo Kim
- Department of Surgery, Ajou University Hospital, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, 16499, Korea.
| |
Collapse
|
26
|
Cai F, Guo Z, Wang G, Luo F, Yang Y, Lv M, He J, Xiu Z, Tang D, Bao X, Zhang X, Yang Z, Chen Z. Integration of intratumoral and peritumoral CT radiomic features with machine learning algorithms for predicting induction therapy response in locally advanced non-small cell lung cancer. BMC Cancer 2025; 25:461. [PMID: 40082786 PMCID: PMC11907900 DOI: 10.1186/s12885-025-13804-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVES To extract intratumoral, peritumoral, and integrated intratumoral-peritumoral CT radiomic features, develop multi-source radiomic models using various machine learning algorithms to identify the optimal model, and integrate clinical factors to establish a nomogram for predicting the therapeutic response to induction therapy(IT) in locally advanced non-small cell lung cancer. METHODS This study included 209 patients with locally advanced non-small cell lung cancer (LA-NSCLC) who received IT as the training cohort, and an external validation cohort comprising 50 patients from another center. Radiomic features were extracted from intratumoral, peritumoral, and integrated intratumoral-peritumoral regions by manually delineating the gross tumor volume (GTV) and an additional 3 mm surrounding area. Three machine learning algorithms-Support Vector Machine (SVM), XGBoost, and Gradient Boosting-were employed to construct radiomic models for each region. Model performance was evaluated in the external validation cohort using metrics such as Area Under the Curve (AUC), confusion matrix, accuracy, precision, recall, and F1 score. Finally, a comprehensive nomogram integrating the optimal radiomic model with independent clinical predictors was developed. RESULTS Through a comparison of optimal machine learning algorithms, INTRAPERI, INTRA, and PERI achieved the best performance with Gradient Boosting, SVM, and XGBoost, respectively. Compared to the INTRA_SVM and PERI_XGBoost INTRA models, the fusion model that integrates INTRA and peritumoral regions within a 3 mm margin around the tumor (INTRAPERI_GradientBoosting) showed better predictive performance in the training set, with AUCs of 93.7%, 82.5%, and 89.4%, respectively. In the clinical model, the PS score was identified as an independent predictive factor. The nomogram combining clinical factors with the INTRAPERI_GradientBoosting score demonstrated clinical predictive value. CONCLUSION The INTRAPERI_GradientBoosting model, which integrates intra-tumoral and peritumoral features, performs better than the INTRA intra-tumoral and PERI peritumoral radiomics models in predicting the efficacy of IT therapy in LA-NSCLC. Additionally, the nomogram based on INTRAPERI intra-tumoral and peritumoral features combined with independent clinical predictors has clinical predictive value.
Collapse
Affiliation(s)
- FangHao Cai
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, No. 288 Tianwen Road, Nan'an District, Chongqing, 400010, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400010, China
| | - Zhengjun Guo
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, No. 288 Tianwen Road, Nan'an District, Chongqing, 400010, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400010, China.
| | - GuoYu Wang
- Department of Oncology and Hematology, The First People's Hospital of Longquanyi District, No. 669, Donglang Road, Chengdu, 610100, China
| | - FuPing Luo
- Department of Oncology and Hematology, The First People's Hospital of Longquanyi District, No. 669, Donglang Road, Chengdu, 610100, China
| | - Yang Yang
- Department of Oncology and Hematology, The First People's Hospital of Longquanyi District, No. 669, Donglang Road, Chengdu, 610100, China
| | - Min Lv
- Department of Oncology and Hematology, The First People's Hospital of Longquanyi District, No. 669, Donglang Road, Chengdu, 610100, China
| | - JiMin He
- Department of Oncology and Hematology, The First People's Hospital of Longquanyi District, No. 669, Donglang Road, Chengdu, 610100, China
| | - ZhiGang Xiu
- Department of Oncology and Hematology, The First People's Hospital of Longquanyi District, No. 669, Donglang Road, Chengdu, 610100, China
| | - Dan Tang
- Department of Oncology and Hematology, The First People's Hospital of Longquanyi District, No. 669, Donglang Road, Chengdu, 610100, China
| | - XiaoHui Bao
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, No. 288 Tianwen Road, Nan'an District, Chongqing, 400010, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400010, China
| | - XiaoYue Zhang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, No. 288 Tianwen Road, Nan'an District, Chongqing, 400010, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400010, China
| | - ZhenZhou Yang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, No. 288 Tianwen Road, Nan'an District, Chongqing, 400010, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400010, China.
| | - Zhi Chen
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, No. 288 Tianwen Road, Nan'an District, Chongqing, 400010, China.
- Department of Oncology and Hematology, The First People's Hospital of Longquanyi District, No. 669, Donglang Road, Chengdu, 610100, China.
| |
Collapse
|
27
|
Markozannes G, Cividini S, Aune D, Becerra-Tomás N, Kiss S, Balducci K, Vieira R, Cariolou M, Jayedi A, Greenwood DC, Brockton NT, Croker H, Mitrou P, Copson E, Renehan AG, Bours M, Demark-Wahnefried W, Hudson MM, May AM, Odedina FT, Skinner R, Steindorf K, Tjønneland A, Velikova G, Baskin ML, Chowdhury R, Hill L, Lewis SJ, Seidell J, Weijenberg MP, Krebs J, Cross AJ, Tsilidis KK, Chan DSM. The role of physical activity, sedentary behaviour, diet, adiposity and body composition on health-related quality of life and cancer-related fatigue after diagnosis of colorectal cancer: a Global Cancer Update Programme (CUP Global) systematic literature review and meta-analysis. ESMO Open 2025; 10:104301. [PMID: 40086399 PMCID: PMC11952013 DOI: 10.1016/j.esmoop.2025.104301] [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: 10/30/2024] [Accepted: 01/23/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The impact of physical activity, sedentary behaviour, diet, adiposity, and body composition on health-related quality of life (HRQoL) and cancer-related fatigue among colorectal cancer survivors remains uncertain. METHODS PubMed, Embase, and CENTRAL were systematically searched until April 2023 for relevant randomised controlled trials (RCTs) and cohort studies. Random-effects meta-analyses or descriptive syntheses were conducted depending on the number of studies. The evidence was interpreted and graded by an independent World Cancer Research Fund Expert Committee and Expert Panel. RESULTS We included 31 RCTs (18 exercise, 14 diet) and 30 cohort studies (8 physical activity, 3 sedentary behaviour, 13 diet, 9 adiposity and body composition). Meta-analyses were possible for exercise RCTs that showed non-significant effects but indicative of improved HRQoL (overall four trials for global HRQoL, physical and emotional well-being) and fatigue (five trials). These studies were rated at a high risk of bias (RoB), and evidence was graded as 'very low certainty of an effect'. Descriptive synthesis of interventions to improve diet quality suggested small improvements in global HRQoL and physical well-being, but with a high RoB rating leading to a 'low certainty' grading. Evidence from RCTs on probiotics and supplements and evidence from observational studies on sedentary behaviour, and various dietary and body composition factors was generally inconsistent and too scarce to draw conclusions. CONCLUSIONS Exercise and diet quality interventions might improve HRQoL and fatigue outcomes in colorectal cancer survivors. The evidence overall was limited and should be strengthened by larger, well-designed RCTs across the cancer continuum.
Collapse
Affiliation(s)
- G Markozannes
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - S Cividini
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - D Aune
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK; Department of Nutrition, Oslo New University College, Oslo, Norway; Department of Research, The Cancer Registry of Norway, Oslo, Norway
| | - N Becerra-Tomás
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK; Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Rovira i Virgili University, Reus, Spain
| | - S Kiss
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - K Balducci
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - R Vieira
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - M Cariolou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - A Jayedi
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - D C Greenwood
- Leeds Institute for Data Analytics, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - N T Brockton
- American Institute for Cancer Research, Washington, USA
| | - H Croker
- World Cancer Research Fund International, London, UK
| | - P Mitrou
- World Cancer Research Fund International, London, UK
| | - E Copson
- Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - A G Renehan
- The Christie NHS Foundation Trust, Manchester Cancer Research Centre, NIHR Manchester Biomedical Research Centre, Manchester, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - M Bours
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - W Demark-Wahnefried
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Alabama, USA
| | - M M Hudson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, USA
| | - A M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - F T Odedina
- Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA
| | - R Skinner
- Department of Paediatric and Adolescent Haematology and Oncology, Great North Children's Hospital, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, and Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - K Steindorf
- Division of Physical Activity, Prevention and Cancer, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - A Tjønneland
- Danish Cancer Institute, Diet, Cancer and Health, Copenhagen, Denmark; Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - G Velikova
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Leeds Cancer Centre, St James's University Hospital, Leeds, UK
| | - M L Baskin
- UPMC Hillman Cancer Center, Pittsburgh, USA
| | - R Chowdhury
- Department of Global Health, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, USA
| | - L Hill
- American Institute for Cancer Research, Washington, USA
| | - S J Lewis
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - J Seidell
- Department of Health Sciences, Faculty of Science, Vrije Universiteit Amsterdam, The Netherlands
| | - M P Weijenberg
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - J Krebs
- Department of Zoology, University of Oxford, Oxford, UK
| | - A J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - K K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - D S M Chan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
| |
Collapse
|
28
|
Dai L, Yin J, Xin X, Yao C, Tang Y, Xia X, Chen Y, Lai S, Lu G, Huang J, Zhang P, Li J, Chen X, Zhong X. An interpretable machine learning model based on computed tomography radiomics for predicting programmed death ligand 1 expression status in gastric cancer. Cancer Imaging 2025; 25:31. [PMID: 40075494 PMCID: PMC11905525 DOI: 10.1186/s40644-025-00855-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Programmed death ligand 1 (PD-L1) expression status, closely related to immunotherapy outcomes, is a reliable biomarker for screening patients who may benefit from immunotherapy. Here, we developed and validated an interpretable machine learning (ML) model based on contrast-enhanced computed tomography (CECT) radiomics for preoperatively predicting PD-L1 expression status in patients with gastric cancer (GC). METHODS We retrospectively recruited 285 GC patients who underwent CECT and PD-L1 detection from two medical centers. A PD-L1 combined positive score (CPS) of ≥ 5 was considered to indicate a high PD-L1 expression status. Patients from center 1 were divided into training (n = 143) and validation sets (n = 62), and patients from center 2 were considered a test set (n = 80). Radiomics features were extracted from venous-phase CT images. After feature reduction and selection, 11 ML algorithms were employed to develop predictive models, and their performance in predicting PD-L1 expression status was evaluated using areas under receiver operating characteristic curves (AUCs). SHapley Additive exPlanations (SHAP) were used to interpret the optimal model and visualize the decision-making process for a single individual. RESULTS Nine features significantly associated with PD-L1 expression status were ultimately selected to construct the predictive model. The light gradient-boosting machine (LGBM) model demonstrated the best performance for PD-L1 high expression status prediction in the training, validation, and test sets, with AUCs of 0.841(95% CI: 0.773, 0.908), 0.834 (95% CI:0.729, 0.939), and 0.822 (95% CI: 0.718, 0.926), respectively. The SHAP summary and bar plots illustrated that a feature's value affected the feature's impact attributed to the model. The SHAP waterfall plots were used to visualize the decision-making process for a single individual. CONCLUSION Our CT radiomics-based LGBM model may aid in preoperatively predicting PD-L1 expression status in GC patients, and the SHAP method may improve the interpretability of this model.
Collapse
Affiliation(s)
- Lihuan Dai
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Jinxue Yin
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Xin Xin
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Chun Yao
- Department of Radiology, Meizhou People's Hospital, Mei Zhou, 514031, China
| | - Yongfang Tang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Xiaohong Xia
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Yuanlin Chen
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Shuying Lai
- Department of Radiology, Meizhou People's Hospital, Mei Zhou, 514031, China
| | - Guoliang Lu
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Jie Huang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Purong Zhang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Jiansheng Li
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| | - Xiangguang Chen
- Department of Radiology, Meizhou People's Hospital, Mei Zhou, 514031, China.
| | - Xi Zhong
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| |
Collapse
|
29
|
Canale MG, Muñoz FL, Muñoz SE, Diaz MDP. Favorable trends in lung cancer incidence with unfavorable survival prognosis: A spatiotemporal analysis by histology in Córdoba, Argentina. Cancer Epidemiol 2025; 96:102796. [PMID: 40081021 DOI: 10.1016/j.canep.2025.102796] [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: 11/08/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Lung cancer (LC) represents the leading cause of cancer-related death and the third in incidence in Argentina. Survival rates are low. OBJECTIVE To analyze the spatial distribution of LC incidence in Córdoba-Argentina (2004-2014), explore trends in histological types, and estimate the probability of survival. METHODS A longitudinal ecological study was conducted using data from the Provincial Cancer Registry. Age-specific and standardized incidence rates for LC (ICD-10: C33-34) were calculated, truncated (35-84 years), and stratified by sex, year (2004-2014), and histology (small cell carcinoma and non-small cell: adenocarcinoma, squamous cells, large cells, and other carcinomas). Temporal analysis employed Joinpoint regression models, estimating annual percentage changes (APC). Median times estimated survival curves and semiparametric Cox regression models were employed for survival. Statistical significance: log-rank tests and proportional hazards tests. Software: Joinpoint-Regression-Program and Stata17. RESULTS From 2004-2014, 8246 LC cases were diagnosed in individuals aged 35-84. The highest incidence occurred in males aged 75-79 and females aged 80-84. The Age-standardized incidence rates for males and females were 57.9 and 23.6 cases per 100,000 person-years, respectively. In both sexes, the temporal incidence trend was decreasing (APC -3.21 %; p = 0.001), more pronounced in males (APC -3.99 %, p = 0.011), with negative APCs in all histological subtypes. The probability of survival decreased to 32 % (95 %CI: 31 %-34 %) within just 12 months (38 % in females, 30 % in males). The risk of death increased proportionally with age (males HR: 1.007, (95 %CI: 1.004-1.01, p = 0.000); females HR: 1.005, (95 %CI: 1.00-1.01, p = 0.031)) and across all histological types, with lower proportional risks in females and disparities based on histology: in males, the highest risk was in large cells (p = 0.008) and SMCC, while in females, it was SCLC (p = 0.055). CONCLUSIONS Despite estimating a favorable trend in LC incidence since 2004, the survival prognosis remains unfavorable one-year post-diagnosis, dependent on sex, age, and histological type.
Collapse
Affiliation(s)
- Marcela Guadalupe Canale
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina; Escuela de Nutrición, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Fabian Leonardo Muñoz
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina; Escuela de Nutrición, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sonia Edith Muñoz
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Del Pilar Diaz
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
30
|
Langenberg KPS, van Hooff SR, Koopmans B, Strijker JGM, Kholosy WM, Ober K, Zwijnenburg DA, van der Hoek JJF, Keller KM, Vernooij L, Schild LG, Looze EJ, Ebus ME, Essing AHW, Vree PD, Tas ML, Matser YAH, Wienke J, Volckmann R, Tops BBJ, Kester LA, Badloe S, Hehir-Kwa JY, Kemmeren P, Goemans BF, Zwaan CM, Oehme I, Jäger N, Witt O, van Eijkelenburg NKA, Dierselhuis MP, Tytgat GAM, Wijnen MHW, van Noesel MM, de Krijger RR, Eising S, Koster J, Dolman EM, Molenaar JJ. Exploring high-throughput drug sensitivity testing in neuroblastoma cell lines and patient-derived tumor organoids in the era of precision medicine. Eur J Cancer 2025; 218:115275. [PMID: 39954414 PMCID: PMC11884408 DOI: 10.1016/j.ejca.2025.115275] [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: 03/15/2023] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Despite druggable events to be present in 80 % of neuroblastomapatients within the Princess Máxima Center precision medicine program 'iTHER', clinical uptake of treatment recommendations has been low, and the clinical impact for individual patients remains hard to predict. This stresses the need for a method integrating genomics and transcriptomics with functional approaches into therapeutic decision making. METHODS We aimed to launch an online repository integrating genomics and transcriptomics with high-throughput drug screening (HTS) of nineteen commonly used neuroblastoma cell lines and fifteen neuroblastoma patient-derived organoids (NBL-PDOs). Cell lines, NBL-PDOs and their parental tumors were characterized utilizing (lc)WGS, WES and RNAseq. Cells were exposed to ∼200 compounds. Results were transferred to the R2 visualization platform. RESULTS A powerful reference set of cell lines is available, reflecting distinct known pharmacologic vulnerabilities. HTS identified additional therapeutic vulnerabilities, such as a striking correlation between a positive mesenchymal signature and sensitivity to BCL2-inhibitor venetoclax. Finally, we explored personalized drug sensitivities within iTHER, demonstrating HTS can support genomic and transcriptomic results, thereby strengthening the rationale for clinical uptake. CONCLUSION We established a dynamic publicly available dataset with detailed genomic, transcriptomic, and pharmacological annotation of classical neuroblastoma cell lines as well as novel sharable NBL-PDOs, representing the heterogeneous landscape of neuroblastoma. We anticipate that in vitro drug screening will be complementary to genomic-guided precision medicine by supporting clinical decision making, thereby improving prognosis for all neuroblastoma patients in the future.
Collapse
Affiliation(s)
- Karin P S Langenberg
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Sander R van Hooff
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Bianca Koopmans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Josephine G M Strijker
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Waleed M Kholosy
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Kimberley Ober
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Danny A Zwijnenburg
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Jessica J F van der Hoek
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Kaylee M Keller
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Lindy Vernooij
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Linda G Schild
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Eleonora J Looze
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Marli E Ebus
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Anke H W Essing
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Paula de Vree
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Michelle L Tas
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands
| | - Yvette A H Matser
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Judith Wienke
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Richard Volckmann
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Bastiaan B J Tops
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Lennart A Kester
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Shashi Badloe
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jayne Y Hehir-Kwa
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Patrick Kemmeren
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Bianca F Goemans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 430, Heidelberg 69120, Germany.
| | - Nathalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands.
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg 69120 , the Netherlands; Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Im Neuenheimer Feld 430, Heidelberg 69120, Germany; Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital,National Center for Tumor Diseases (NCT) Network, Heidelberg, Germany.
| | | | - Miranda P Dierselhuis
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Marc H W Wijnen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Division Imaging & Cancer, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| | - Selma Eising
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jan Koster
- Department of Oncogenomics, Cancer Center Amsterdam, Amsterdam UMC, the Netherlands.
| | - Emmy M Dolman
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands.
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, the Netherlands; Department of Pharmaceutical Sciences, Utrecht University, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands.
| |
Collapse
|
31
|
Lu Z, Sun G, Li J, Zhao J, Wang Z, Qian D, Yang Z, Li N, Wang J, Yuan S, Wang Y, Li S, Yang Z, Ran F, Ji Y, Zhu S, Zhang Y, Wang C, Wan L, Zheng R, Deng W, Cheng F, Shen L. Effectiveness, safety, and patterns of use of camrelizumab in advanced esophageal cancer: an individual patient data pooled analysis of 987 patients from three prospective cohort studies. Cancer Immunol Immunother 2025; 74:138. [PMID: 40056201 PMCID: PMC11890476 DOI: 10.1007/s00262-025-03970-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: 12/02/2024] [Accepted: 02/06/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND AND AIMS This individual patient data pooled analysis aimed to evaluate the effectiveness, safety, and patterns of use of camrelizumab in a large cohort of advanced esophageal cancer (AEC) patients. APPROACH AND RESULTS Adult patients (≥ 18 years) who had received camrelizumab as part of AEC treatment were pooled from three independent, prospective observational cohort studies (NCT04616040, ChiCTR1900027275, and ChiCTR2000039499). The main outcomes were patterns of camrelizumab use, progression-free survival (PFS), overall survival (OS), and safety in the overall population and specific subgroups of underrepresented patients. Among 987 patients, 450 (45.6%) received camrelizumab in the first line, 398 (40.3%) in the second line, and 139 (14.1%) in the third line or later. Most (69.7%) patients received camrelizumab plus chemotherapy regardless of treatment lines. The median PFS was 9.9 (95% CI 7.4, 14.4), 6.6 (95% CI 5.1, 8.8), and 5.7 (95% CI 3.1, 9.6) months in the first line, second line, and third line or later, respectively. The corresponding median OS was 15.5 (95% CI 12.6, 18.4), 12.1 (95% CI 10.0, 14.7), and 10.9 (95% CI 8.1, 14.5) months. Patients with poor performance status (ECOG PS ≥ 2) and with camrelizumab in the second line or later, but not patients with older age (≥ 75 years), were associated with poor survival. Adverse events occurred in 721 (73.0%) patients, with no new safety signals. CONCLUSIONS This study provides an overview of camrelizumab use in unselected AEC patients. The real-world effectiveness and safety of camrelizumab are generally consistent with those observed in pivotal trials.
Collapse
Affiliation(s)
- Zhihao Lu
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Beijing, China.
| | - Guoping Sun
- Department of Medical Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiancheng Li
- Department of Radiation Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jun Zhao
- Department of Oncology, Changzhi People's Hospital of Changzhi Medical College, Changzhi, China
| | - Zishu Wang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Hefei, China
| | - Zhe Yang
- Cancer Research and Treatment Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Na Li
- Department of Oncology, Suining Central Hospital, Suining, China
| | - Junsheng Wang
- Department of Internal Medicine, Anyang Cancer Hospital, Anyang, China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Yusheng Wang
- Department of Gastroenterology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Suyi Li
- Department of Oncology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Hefei, China
| | - Zhen Yang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fengming Ran
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinghua Ji
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shaojin Zhu
- Department of Thoracic Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Yanqiao Zhang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chen Wang
- Department of Gastroenterology, Ganzhou People's Hospital - North Hospital, Ganzhou, China
| | - Lixin Wan
- Department of Medial Oncology, Nanyang Central Hospital, Nanyang, China
| | - Rongrong Zheng
- Department of Medical Affairs, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, China
| | - Wenjie Deng
- Department of Medical Affairs, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, China
| | - Fengzhuo Cheng
- Department of Medical Affairs, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Beijing, China.
| |
Collapse
|
32
|
Yari M, Eidi M, Omrani MA, Fazeli Z, Rahmanian M, Ghafouri-Fard S. Comprehensive identification of hub mRNAs and lncRNAs in colorectal cancer using galaxy: an in silico transcriptome analysis. Discov Oncol 2025; 16:282. [PMID: 40056245 DOI: 10.1007/s12672-025-02026-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/03/2025] [Indexed: 03/10/2025] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related mortality. Using the Galaxy platform, the present study aimed to assess the differentially expressed genes (DEGs) in CRC patients. The expression data was obtained from the Gene Expression Omnibus database (GSE137327). DEGs were analyzed using Gene Ontology (GO) and GeneMANIA databases to detect the most critical biological pathways and processes. Protein-Protein Interaction Studies (PPIS) identified four hub genes (CCN1, CCL2, FLNC, MYH11). This article presents findings on three mRNAs (CEMIP, MMP7, and DPEP1) and also two notable lncRNAs, EVADR and DLX6-AS1, that have an impact on CRC pathogenesis and play a role in the epithelial-mesenchymal transition in tumor cells. The identified genes and lncRNAs are putative therapeutic targets and diagnostic markers. For instance, CRISPR/Cas9 editing systems can be designed in order to modulate expression of these genes, or edit them for the purpose of inducing sensitivity to conventional therapies. Besides, these genes can be incorporated into clinical prognostic models, offering panels of genes to choose appropriate personalized methods of treatment. Together, these genes represent novel markers and possible therapeutic targets for CRC.
Collapse
Affiliation(s)
- Mohsen Yari
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Milad Eidi
- The Endocrine Genetics Laboratory, Child Health and Human Development Program and Department of Pediatrics, Mcgill University Health Centre Research Institute, Montreal, QC, Canada
| | - Mohammad-Amin Omrani
- Urology and Nephrology Research Center (UNRC), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Fazeli
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rahmanian
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Liu L, Wang L, Ding Y, Zhang Q, Shu Y. Cost-effectiveness of atezolizumab plus bevacizumab versus sorafenib as first-line therapy in unresectable hepatocellular carcinoma in the US and Chinese setting: a modelling comparison study. BMJ Open 2025; 15:e094804. [PMID: 40050065 PMCID: PMC11887288 DOI: 10.1136/bmjopen-2024-094804] [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: 10/09/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
OBJECTIVE Atezolizumab plus bevacizumab demonstrates a significant improvement in overall survival and progression-free survival compared with sorafenib in patients with unresectable hepatocellular carcinoma (HCC). The combined usage of these two medications could result in substantial consumption of resources, primarily due to their exceptionally high costs. The current study aims to evaluate the cost-effectiveness of atezolizumab plus bevacizumab as a first-line treatment for advanced HCC from the perspective of payers in developed and developing countries. DESIGN A partitioned survival model was constructed to evaluate the cost-effectiveness of atezolizumab plus bevacizumab versus sorafenib as a first-line treatment for advanced HCC. The efficacy and safety data incorporated within the model were derived from the IMbrave150 trial. Costs and utilities were extracted from published sources. INTERVENTIONS Atezolizumab plus bevacizumab versus sorafenib. OUTCOME MEASURES Estimates were calculated for costs, life-years, quality-adjusted life-years (QALYs), incremental cost-effectiveness ratio (ICER) for both treatment strategies. One-way sensitivity, probabilistic sensitivity, expected value of perfect information (EVPI), subgroup and scenario analyses were conducted. RESULTS The combination therapy of atezolizumab and bevacizumab results in an additional 0.72 life-years/0.57 QALYs in the USA and 0.64 life-years/0.47 QALYs in China compared with standard sorafenib treatment, although with a significant increase in costs, yielding an average ICER of US$253 247.07/QALY in the USA and US$181 552.71/QALY in China. The probability sensitivity analysis indicated that atezolizumab plus bevacizumab demonstrated a 13.60% likelihood of cost-effectiveness in the USA, whereas this likelihood is negligible (0%) in China. The expected value of uncertainty, as quantified by the EVPI, was estimated at approximately US$3658.41/patient in the USA and US$0/patient in China. The ICER was most sensitive to the cost of subsequent treatment in the USA, and most sensitive to the cost of atezolizumab in China. In scenario analyses, the atezolizumab plus bevacizumab treatment becomes favourable when the cost of atezolizumab decreases to 67.85% and 18.45% of its original price in the USA and China, respectively. CONCLUSIONS The atezolizumab plus bevacizumab is unlikely to be cost-effective compared with sorafenib for patients with unresectable HCC in the context of the USA and China. The implementation of significant reductions in drug prices may render the treatment economically viable.
Collapse
Affiliation(s)
- Lulu Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Lei Wang
- Outpatient Department Office, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiling Ding
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qilin Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yamin Shu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Wang KX, Shi DM, Shi XL, Wang JY, Ai XH. Obesity promotes immunotherapy efficacy by up-regulating the glycolytic-mediated histone lactacylation modification of CD8+ T cells. Front Pharmacol 2025; 16:1533464. [PMID: 40110127 PMCID: PMC11920648 DOI: 10.3389/fphar.2025.1533464] [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: 11/24/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
The response rate of immune checkpoint blockade (ICB) therapy for non-small-cell lung cancer (NSCLC) remains limited. Recent evidence suggests that obese cancer patients are more likely to benefit from ICB therapy, however, the specific mechanism needs further research. In this study, we found that anti-PD-1 therapy was more effective in obese NSCLC patients compared to normal weight patients and this was verified in mouse NSCLC model. Further bioinformatics analysis indicated that the glycolytic metabolism was markedly elevated in obese NSCLC patients. In vitro co-culture experiment showed that both increased glycolysis of tumor cells and external addition of lactate promoted T cell PD-1 expression. And, PD-1 upregulation was related to monocarboxylate transporter 1 (MCT1)-mediated lactate transport and subsequent lysine lactylation of histones in T cells. Based on the aforementioned data, our study contributes to better application of anti-PD-1 therapy in NSCLC.
Collapse
Affiliation(s)
- Kai-Xuan Wang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dong-Min Shi
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Li Shi
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jing-Yuan Wang
- Department of Medical Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xing-Hao Ai
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
35
|
Jia G, Li X. Survival trends of gastrointestinal stromal tumor in real-world settings: a population-based retrospective study. Pathol Oncol Res 2025; 31:1611896. [PMID: 40103620 PMCID: PMC11913614 DOI: 10.3389/pore.2025.1611896] [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: 07/03/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
Purpose This study aims to evaluate whether survival outcomes for GIST patients have improved over the past decades and to identify the specific patient subgroups that have benefited from advances in treatment. Patients and methods A total of 4,127 GIST patients diagnosed between January 1980, and December 2019, were included in this study using data from the Surveillance, Epidemiology, and End Results (SEER)-9 Registries. Survival differences among GIST patients were analyzed across five time periods (1980-1999, 2000-2004, 2005-2009, 2010-2014, and 2015-2019) and within demographic, neoplastic, temporal, economic, and geographic categories using the log-rank test. Multivariable Cox regression models were employed to identify risk factors associated with GIST-specific survival. Associations between time periods and GIST-specific mortality (TSM) were examined using a multivariable Cox regression model. Results Survival outcomes for GIST patients significantly improved in the 2000-2009 period but showed no substantial improvement in the 2010-2019 period. After adjusting for age, gender, tumor location, ethnicity, tumor stage, median household income, and geographic area, the multivariable Cox regression models revealed that older age (≥65 years) (HR = 1.977, 95% CI = 1.470-2.657), tumors located outside the gastrointestinal tract (HR = 1.505, 95% CI = 1.267-1.786), regional lesions (HR = 2.225, 95% CI = 1.828-2.708), and distant lesions (HR = 5.177, 95% CI = 4.417-6.069) were independent risk factors for TSM (p < 0.05). After adjusting for time periods and age, gender, tumor location, tumor stage, median household income, patients in 2000-2004 (HR = 0.662, 95% CI = 0.523-0.839), 2005-2009 (HR = 0.431, 95% CI = 0.339-0.549), 2010-2014 (HR = 0.437, 95% CI = 0.341-0.561), and 2015-2019 (HR = 0.365, 95% CI = 0.273-0.489) had a significantly lower risk of TSM than patients in 1980-1999 (p < 0.05). Similarly, patients in 2005-2009 (HR = 0.661, 95% CI = 0.555-0.788), 2010-2014 (HR = 0.696, 95% CI = 0.578-0.838), and 2015-2019 (HR = 0.607, 95% CI = 0.476-0.773) also had a significantly lower risk of TSM than patients in 2000-2004 (p < 0.05). However, patients in 2010-2014 (HR = 1.042, 5% CI = 0.863-1.258) and 2015-2019 (HR = 0.945, 95% CI = 0.734-1.216) did not have a significantly lower risk of TSM compared to patients in 2005-2009 (p > 0.05). Conclusion GIST survival has significantly improved during the period 2000-2009 but showed no substantial improvement in 2010-2019, with the turning point for lower risk of TSM being 2005. Innovative strategies are needed to further improve survival outcomes for GIST patients, particularly for older patients and those with tumors originating outside the gastrointestinal tract.
Collapse
Affiliation(s)
- Guohua Jia
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiangpan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
36
|
Dickerson LD, Gittens J, Brunning C, Jackson R, Schmid MC, Mielgo A, Palmer D, Halloran CM, Ghaneh P. Neoadjuvant treatment versus upfront surgery in borderline resectable and resectable pancreatic ductal adenocarcinoma: meta-analysis. BJS Open 2025; 9:zrae172. [PMID: 40126570 PMCID: PMC11932015 DOI: 10.1093/bjsopen/zrae172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/25/2024] [Accepted: 12/25/2024] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Pancreatic cancer prognosis remains poor despite advances in adjuvant treatment. Neoadjuvant treatment may improve survival and disease-free survival. This meta-analysis evaluates the outcomes for patients with borderline-resectable (borderline-resectable pancreatic cancer) or resectable disease (resectable pancreatic cancer) in randomized trials of neoadjuvant therapy versus upfront surgery. METHODS The review was performed according to PRISMA guidance. Articles were included from the start of the database until 1 May 2024. The primary outcome was overall survival. Secondary outcomes were progression-free survival, resection rate, R0 rate, N0 rate, vascular resection rate, surgical complications, significant adverse events and rates of adjuvant therapy. Data was collected from study manuscripts or through individual patient-level data extraction. Meta-analysis was performed using a random-effects model with subgroup comparisons for resectability status (resectable pancreatic cancer versus borderline-resectable pancreatic cancer) and treatment modality (chemotherapy versus chemoradiotherapy). RESULTS Nine trials were included representing 1194 patients. Four trials recruited borderline-resectable pancreatic cancer, four resectable pancreatic cancer and one both. Four trials reported chemotherapy, four chemoradiotherapy and one both treatments. Neoadjuvant treatment improved overall survival (HR 0.73, 95% c.i. 0.55 to 0.98; P = 0.001) and progression-free survival (HR 0.80, 95% c.i. 0.65 to 0.99; P = 0.041). Subgroup analysis demonstrated neoadjuvant treatment improved overall survival for borderline-resectable pancreatic cancer (HR 0.60, 95% c.i. 0.38 to 0.96) but not resectable pancreatic cancer (HR 0.90, 95% c.i. 0.63 to 1.28). The overall resection rate was lower in neoadjuvant treatment (72.6% versus 80.6%, RR 0.94, 95% c.i. 0.89 to 0.99; P = 0.020). R0 rate (43.8% versus 23.0%, RR 1.35, 95% c.i. 1.16 to 1.57; P = 0.002) and N0 rate (30.9% versus 15.0%, RR 2.03, 95% c.i. 1.50 to 2.74; P = 0.001) was improved in neoadjuvant treatment. Significant adverse events occurred more frequently in neoadjuvant treatment (56.1% versus 27.0%, RR 1.92, 95% c.i. 1.28 to 1.89; P = 0.007). CONCLUSION Neoadjuvant treatment significantly improves survival in borderline-resectable pancreatic cancer but not resectable pancreatic cancer. It should be regarded as standard of care for these patients. Further work is needed to identify the optimum neoadjuvant regimen and a possible role in the treatment of resectable pancreatic cancer.
Collapse
Affiliation(s)
- Luke D Dickerson
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Pancreatobiliary Surgery Unit, Liverpool University Foundation Trust, Liverpool, UK
| | - Jayden Gittens
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Chris Brunning
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Richard Jackson
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Michael C Schmid
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Ainhoa Mielgo
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Daniel Palmer
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Department of Medical Oncology, Clatterbridge Cancer Centre, Liverpool, UK
| | - Christopher M Halloran
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Pancreatobiliary Surgery Unit, Liverpool University Foundation Trust, Liverpool, UK
| | - Paula Ghaneh
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Pancreatobiliary Surgery Unit, Liverpool University Foundation Trust, Liverpool, UK
| |
Collapse
|
37
|
Oliveira JCDS, Galvão ND, Andrade ACDS, da Silva AMC. Five-year overall and specific survival of breast cancer in great Cuiaba (MT), Brazil. REVISTA BRASILEIRA DE EPIDEMIOLOGIA 2025; 28:e250010. [PMID: 40053007 PMCID: PMC11884821 DOI: 10.1590/1980-549720250010] [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: 05/28/2024] [Revised: 11/30/2024] [Accepted: 12/12/2024] [Indexed: 03/10/2025] Open
Abstract
OBJECTIVE To analyze the overall and cancer-specific five-year survival rates for female breast cancer in Greater Cuiabá, Mato Grosso, Brazil. METHODS A non-concurrent, population-based cohort study using the Population-Based Cancer Registry of Greater Cuiabá (Cuiabá and Varzea Grande), including women diagnosed with breast cancer from 2008 to 2013, followed through 2018 in the regional mortality database. The sample consisted of a total of 1,220 women. Five-year survival analysis was performed using Kaplan-Meier curves and the Cox proportional hazards regression model, computing hazard ratios for variable estimation. Survival curves were compared using the log-rank test (p<0.05). Probabilistic linkage technique by the RecLink III software and survival analysis were conducted using STATA software version 12.0. RESULTS There was no statistical difference between the overall (OS) and cancer-specific survival (SS) rates (OS 78.0%, 95%CI 75.6-80.2; SS 81.0%, 95%CI 78.7-83.2). Women with lower educational levels (OS=58.33%; SS=64.89%) and those without a partner (OS 64.81%; SS 70.41%) exhibited poorer survival. CONCLUSION This study demonstrates that educational level and marital status significantly impact both overall and cancer-specific survival rates for female breast cancer. There is a need to propose policies that address the profile of women with lower survival rates.
Collapse
Affiliation(s)
| | - Noemi Dreyer Galvão
- Universidade Federal de Mato Grosso, Institute of Public Health – Cuiabá (MT), Brazil
- Mato Grosso State Health Department – Cuiabá (MT), Brazil
| | | | | |
Collapse
|
38
|
Gao X, Zhang G, Wang F, Ruan W, Sun S, Zhang Q, Liu X. Emerging roles of EGFL family members in neoplastic diseases: Molecular mechanisms and targeted therapies. Biochem Pharmacol 2025; 236:116847. [PMID: 40044051 DOI: 10.1016/j.bcp.2025.116847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
Epidermal growth factor-like proteins (EGFLs) contain more than a single EGF/EGF-like domain within their protein structure. To date, ten EGFL family members (EGFL1-10) have been characterized across diverse tissues and developmental stages under different conditions. In this review, we conclude that EGFLs are instrumental in regulating biological activities and pathological processes. Under physiological conditions, EGFLs participate in angiogenesis, neurogenesis, osteogenesis, and other processes. Under pathological conditions, EGFLs are linked with different diseases, particularly cancers. Furthermore, we highlight recent advancements in the study of EGFLs in biological conditions and cancers. In addition, the regulatory role and key underlying mechanism of EGFLs in mediating tumorigenesis are discussed. This paper also examines potential antagonists that target EGFL family members in cancer therapeutics. In summary, this comprehensive review elucidates the critical role of EGFLs in neoplastic diseases and highlights their potential as therapeutic targets.
Collapse
Affiliation(s)
- Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Guopeng Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Feitong Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Wenhui Ruan
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China
| | - Shishuo Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China; Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China.
| |
Collapse
|
39
|
Maher E, Kamal M, El-Ayadi M, Refaat A, Enayet A, El-Beltagy M, Eldebawy E, Taha H, Awad M, Zaghloul MS. Epidemiology and Clinical Outcomes of Childhood Central Nervous System Cancers in a Large Low/Middle-Income Country Pediatric Oncology Center: A Report on 5,051 Kids. Cancer Epidemiol Biomarkers Prev 2025; 34:420-427. [PMID: 39688610 DOI: 10.1158/1055-9965.epi-24-1188] [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: 08/10/2024] [Revised: 10/26/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Central nervous system (CNS) tumors are the leading cause of cancer-related deaths in children. Although most cases come from low- and middle-income countries (LMIC) where their prognosis is worse, few epidemiologic studies are conducted in these regions. METHODS We conducted a registry-based cohort study for childhood CNS tumors at Children's Cancer Hospital, Egypt, over 15 years. Unified treatment protocols were implemented. Survival analyses were conducted using the Kaplan-Meier function. Cases were additionally annotated using the International Classification of Childhood Cancer-3 classification. RESULTS In total, 5,051 children ≤18 years of age were identified, accounting for 20% of all childhood cancers treated at Children's Cancer Hospital, Egypt. The most common tumor sites were the posterior fossa (36.8%) and brainstem (17.7%). Pathologies were predominantly astrocytic (n = 1,360; 26.9%) and embryonal (n = 1,003; 19.9%) in origin. The 5-year overall survival (OS) and event-free survival for all cases were 64.6% and 51.8%, respectively. More specifically, 1,421 low-grade gliomas were identified, with a 5-year OS of 91.1%. Medulloblastoma (n = 801) recorded a 5-year OS of 66%. The entity with the worst prognosis was diffuse intrinsic pontine glioma (n = 633), with a 5-year OS of 3.2%. CONCLUSIONS We report on a large number of childhood CNS tumors from an LMIC. This study underscores the need to understand the burden of childhood brain tumors and its outcomes in resource-constrained settings. IMPACT This study reports on the epidemiology and clinical outcomes of 5,000+ children with CNS tumors from a specialized LMIC center. Despite the lack of many sophisticated and advanced facilities, LMICs can improve the clinical end-results with experience and augmented efforts.
Collapse
Affiliation(s)
- Eslam Maher
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Mohamed Kamal
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Moatasem El-Ayadi
- Department of Pediatric Oncology, National Cancer Institute, Cairo University, Giza, Egypt
- Department of Pediatric Oncology, Children's Cancer Hospital, Cairo, Egypt
| | - Amal Refaat
- Department of Radiology, Children's Cancer Hospital, Cairo, Egypt
- Department of Radiology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Abdelrahman Enayet
- Department of Neurosurgery, Children's Cancer Hospital, Cairo, Egypt
- Department of Neurosurgery, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed El-Beltagy
- Department of Neurosurgery, Children's Cancer Hospital, Cairo, Egypt
- Department of Neurosurgery, Kasr Al-Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Eman Eldebawy
- Department of Radiation Oncology, Children's Cancer Hospital, Cairo, Egypt
- Department of Radiation Oncology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Hala Taha
- Department of Pathology, Children's Cancer Hospital, Cairo, Egypt
- Department of Pathology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Madiha Awad
- Department of Pediatric Oncology, Children's Cancer Hospital, Cairo, Egypt
| | - Mohamed S Zaghloul
- Department of Radiation Oncology, Children's Cancer Hospital, Cairo, Egypt
- Department of Radiation Oncology, National Cancer Institute, Cairo University, Giza, Egypt
| |
Collapse
|
40
|
Santos TDS, Gonçalves CDA, da Cunha CP, Milhomem JP, da Silva KM, da Costa BT, Piantolo RG, Fernandes RJC, da Silva YM, Guimarães RM. Temporal trend of breast cancer burden among younger and older Brazilian women, 1990-2019. REVISTA BRASILEIRA DE EPIDEMIOLOGIA 2025; 28:e250006. [PMID: 40053004 PMCID: PMC11884820 DOI: 10.1590/1980-549720250006] [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: 06/12/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 03/10/2025] Open
Abstract
OBJECTIVE To analyze the temporal trend of the burden of breast cancer in Brazilian women under 40 years of age compared to the age group over 40 years of age, between 1996 and 2019. METHODS An ecological time trend study was conducted in Brazil between 1996 and 2019 using data from the Global Burden of Disease (GBD) study. The segmented regression method (Joinpoint Regression) was applied to analyze rates among women under and over 40 years of age. To capture differences in the level and trend of mortality and DALYs, the rate ratio was calculated for the two groups on a year-by-year basis. RESULTS Regarding DALY, an average annual decline of 0.7% (95%CI -0.8 to -0.5, p<0.01) was observed among women over 40 years old, while an annual increase of 1.0% (95%CI 0.9 to 1.1, p<0.001) was noted for women up to 40 years old. For mortality, the decline among older women was 0.3% per year (95%CI -0.4 to -0.2, p<0.001), and the increase among young women was 0.8% per year (95%CI 0.7 to 1.0, p<0.001). The average rate ratio for DAILY was 5.2, while for mortality, the average rate ratio was 8.1. CONCLUSION the analysis reinforces the idea that the magnitude and trend of breast cancer mortality among young women is a health issue requiring attention from health decision-makers. This diagnosis underscores the importance of initiating discussions on the need to review population screening criteria, incorporating clinical prediction rules.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Raphael Mendonça Guimarães
- Universidade Estácio de Sá, School of Medicine – Rio de Janeiro (RJ), Brazil
- Oswaldo Cruz Foundation, National School of Public Health – Rio de Janeiro (RJ), Brazil
| |
Collapse
|
41
|
Li J, Cao X, Zhang L, Liu A, Liu S, Chen F, Li Y, Ma H, Sun W, Ouyang S, Dai L, Liu J. Anti-FDX1 Autoantibody as a Potential Biomarker for Non-Small Cell Lung Cancer Detection. Cancer Epidemiol Biomarkers Prev 2025; 34:439-447. [PMID: 39699293 DOI: 10.1158/1055-9965.epi-24-1096] [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: 07/28/2024] [Revised: 10/07/2024] [Accepted: 12/17/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Autoantibodies can be readily identified prior to biopsy and may serve as valuable biomarkers for cancer detection. Ferredoxin 1 (FDX1) is a key regulator in the process of cuproptosis and affects the prognosis of lung cancer. In this study, we investigated whether the anti-FDX1 autoantibody could serve as a novel biomarker for the detection of non-small cell lung cancer (NSCLC). METHODS A total of 1,155 plasma samples were divided into the verification and validation groups. The expression levels of the anti-FDX1 autoantibody in 414 patients with NSCLC, 327 patients with benign pulmonary nodules (BPN), and 414 normal controls (NC) were detected using ELISA. Western blotting and immunofluorescence analyses were performed to confirm the ELISA results. RESULTS Plasma anti-FDX1 autoantibody levels were significantly higher in patients with NSCLC than in patients with BPN and NCs in the verification and validation groups. The ELISA results were confirmed by Western blotting and immunofluorescence. The anti-FDX1 autoantibody distinguished NSCLC from NC and BPN with an AUC (95% confidence interval) of 0.806 (0.772-0.839) and 0.627 (0.584-0.670), respectively. CONCLUSIONS Our study demonstrated the potential benefits of the anti-FDX1 autoantibody as a novel biomarker for NSCLC detection. IMPACT These findings suggested that the anti-FDX1 autoantibody may facilitate the detection of NSCLC.
Collapse
Affiliation(s)
- Jing Li
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Xiaobin Cao
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- BGI College, Zhengzhou University, Zhengzhou, China
| | - Lulu Zhang
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Aichen Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Siyu Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Fengqi Chen
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Yutong Li
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- BGI College, Zhengzhou University, Zhengzhou, China
| | - Hanke Ma
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Wenke Sun
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Songyun Ouyang
- Department of Respiratory and Sleep Medicine in the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Jingjing Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
Li R, Liu Y, Xue R, Wang Y, Zhao F, Chen L, Liu JE. Effectiveness of Nonpharmacologic Interventions for Chemotherapy-Induced Peripheral Neuropathy in Patients With Breast Cancer: A Systematic Review and Network Meta-analysis. Cancer Nurs 2025; 48:E98-E110. [PMID: 37851424 DOI: 10.1097/ncc.0000000000001278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy (CIPN) is a common adverse effect in patients with breast cancer (BC) during treatment. Patients experiencing CIPN develop neuropathic symptoms, which could lead to the modification or discontinuation of chemotherapy. Nonpharmacological interventions can be simple and safe, but evidence of their effectiveness in patients with BC experiencing CIPN is currently insufficient. OBJECTIVE To compare and rank the effectiveness of nonpharmacologic interventions for CIPN in patients with BC. METHODS We conducted a systematic search of randomized controlled trials registered from database inception until October 2022 in 7 databases. We assessed studies that met the inclusion and exclusion criteria and evaluated the risk of bias. Network meta-analysis was conducted using Stata SE 17.0 (StataCorp, College Station, Texas). RESULTS A total of 13 studies involving 9 nonpharmacologic interventions and comprising 571 participants were included. The results of the network meta-analysis showed that cryotherapy (standard mean difference, -1.22; 95% confidence interval, -2.26 to -0.17) exerted significant effects versus usual care. Cryotherapy (surface under the cumulative ranking area [SUCRA]: 0.74) was associated with the highest likelihood of effectively alleviating CIPN in patients with BC, followed by exercise (SUCRA: 0.62) and self-acupressure (SUCRA: 0.59). CONCLUSIONS Cryotherapy was the most effective nonpharmacologic intervention for alleviating CIPN in patients with BC. Large-scale studies are required to verify the present findings. IMPLICATIONS FOR PRACTICE This study provides evidence regarding the effectiveness of nonpharmacologic interventions for CIPN. Physicians and nurses could incorporate cryotherapy into clinical practice to alleviate CIPN in patients with BC.
Collapse
Affiliation(s)
- Ruolin Li
- Authors' Affiliation: School of Nursing, Capital Medical University, Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
43
|
Fuentes-García A, Flores-Figueroa C, Castillo-Delgado A. Sociodemographic, political, and policy contexts of cancer care: A comparative analysis of countries with the highest survival rates. J Cancer Policy 2025; 43:100559. [PMID: 39894214 DOI: 10.1016/j.jcpo.2025.100559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Cancer remains a leading cause of mortality, with 20 million new cases and 10 million deaths in 2022 (WHO). Despite advances in detection and treatment, structural inequalities affect exposure to risk factors and healthcare access. This study compares the cancer care policy contexts of five countries with the highest five-year survival rates. METHODS This qualitative review examines cancer care policies in Australia, Canada, Costa Rica, Belgium, and Japan countries through a critical comparative approach. Data was gathered from official and international documents, focusing on four domains: socio-demographic characteristics, socio-political traditions, health systems, and cancer policies. RESULTS The countries share high life expectancy, and education, while face similar population challenges. Australia and Canada have implemented telemedicine and mobile services to address the needs of dispersed rural populations, while Belgium and Japan ensure equitable access in dense areas. All countries integrate public-private partnerships, and adapt governance structures to contexts, under a strong welfare state with universal health coverage. Cancer policies are characterised by participatory processes that emphasise equity, accessibility, and innovation POLICY SUMMARY: The study identifies consistent patterns in cancer care policies, highlighting contributing factors to high survival rates. Participatory and bottom-up policy design enables responses to complex contexts. Strategies focus on financial sustainability, equity, cultural relevance, and territorial adaptation. An innovative framework for assessing cancer care policy contexts is introduced.
Collapse
Affiliation(s)
- Alejandra Fuentes-García
- School of Public Health, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Center for cancer prevention and control (CECAN) ANID FONDAP 152220002, Chile.
| | - Carla Flores-Figueroa
- Center for cancer prevention and control (CECAN) ANID FONDAP 152220002, Chile; School of Nursing, Faculty of Health, Universidad Santo Tomás, Chile.
| | - Alondra Castillo-Delgado
- Center for cancer prevention and control (CECAN) ANID FONDAP 152220002, Chile; School of Speech Therapy, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
44
|
Dong Y, Xu H, Yu W, Liu Z, Zhao G, Zhang Z, Xia Y, Xiao S, Yi Q, Lin Z. Prevention strategies of esophageal stenosis after endoscopic resection for superficial esophageal cancer: a Bayesian network meta-analysis. Int J Surg 2025; 111:2651-2661. [PMID: 39869370 DOI: 10.1097/js9.0000000000002241] [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: 08/28/2024] [Accepted: 11/29/2024] [Indexed: 01/28/2025]
Abstract
INTRODUCTION What interventions effectively prevent postoperative stenosis following endoscopic resection (ER) of superficial esophageal cancer? This study aimed to identify effective interventions or combinations through a systematic review and network meta-analysis. METHODS Six databases were systematically searched for eligible studies up to 30 April 2023, on interventions to prevent esophageal stenosis post-ER. Odds ratios (ORs) evaluated stenosis rate (primary outcome) and complications (secondary outcome), while mean differences (MD) evaluated endoscopic balloon dilatation (EBD) sessions post-stenosis. RESULTS Twenty-three studies involving 1271 patients and 11 different interventions were included. Eight interventions were effective in preventing post-ER stenosis: oral hydrocortisone sodium succinate and aluminum phosphate gel (OHA) (OR: 0.02, 95% credible interval [CrI]: 0.00-0.11), polyglycolic acid (PGA) + ST (OR: 0.02, 95% CrI: 0.00-0.23), oral tranilast (OT) + preemptive endoscopic balloon dilatation (PEBD) (OR: 0.08, 95% CrI: 0.01-0.77), botulinum toxin (BT) (OR: 0.10, 95% CrI: 0.03-0.32), ST (OR: 0.08, 95% CrI: 0.01-0.67), oral steroid (OS) (OR: 0.11, 95% CrI: 0.05-0.28), endoscopic triamcinolone injection (ETI) + OS (OR: 0.17, 95% CrI: 0.07-0.42), and ETI (OR: 0.18, 95% CrI: 0.11-0.30). Five interventions significantly reduced EBD sessions: PGA + ST (MD: -5.78, 95% CrI: -11.04 to -1.21), ETI + OS (MD: -3.27, 95% CrI: -5.37 to -0.72), OS (MD: -6.18, 95% CrI: -9.43 to -3.38), ETI (MD: -3.81, 95% CrI: -5.74 to -1.99), and BT (MD: -2.16, 95% CrI: -4.12 to -0.40). None of the interventions significantly increased complications. CONCLUSIONS This study confirmed the efficacy of OS, ETI, and ETI + OS and verified five other interventions (OHA, PGA + ST, OT + PEBD, BT, and ST) in preventing stenosis. Notably, PGA + ST and BT also reduced the number of EBD sessions.
Collapse
Affiliation(s)
- Yongqi Dong
- Department of Gastroenterology, Wushan County People's Hospital of Chongqing, Chongqing, People's Republic of China
| | - Hongyan Xu
- Department of Infectious Disease, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Wanli Yu
- Department of Neurosurgery, The Chongqing General Hospital of Chongqing University, Chongqing, People's Republic of China
| | - Zijing Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Gang Zhao
- Department of Gastroenterology, Wushan County People's Hospital of Chongqing, Chongqing, People's Republic of China
| | - Zhihuan Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yuan Xia
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, People's Republic of China
| | - Shiyong Xiao
- Department of Clinical Nutrition, Wushan County People's Hospital of Chongqing, Chongqing, People's Republic of China
| | - Qianzhang Yi
- Department of Radiology, Wushan County People's Hospital of Chongqing, Chongqing, People's Republic of China
| | - Zebin Lin
- Department of Geriatrics, Zhongshan Hospital Xiamen University, Fujian, People's Republic of China
| |
Collapse
|
45
|
Wang L, Miao Y, Gu Y, Kawaguchi T, Mizutani M, Nakai T, Watanabe T, Asai K, Zhang H, Cai W, Tani Y, Kaneda H. Differences in Lung Cancer-related Clinical Practice and Basic Research Background between Japan and China: A Narrative Review. Intern Med 2025:4808-24. [PMID: 40024741 DOI: 10.2169/internalmedicine.4808-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2025] Open
Abstract
With its increasing incidence, lung cancer has become one of the leading causes of cancer-related deaths worldwide, posing a great threat to the health and lives of patients. Due to varying economic and cultural backgrounds, there are significant differences in clinical treatment practices and related basic research on lung cancer between China and Japan. These differences are mainly reflected in many aspects, such as cancer prevention, cancer treatment, provision of medical insurance, patient compliance, medical education system, and sources of research funding. By understanding these differences, China and Japan can learn from each other, make progress together, and strengthen further exchanges and cooperation, which will help improve the long-term efficacy of lung cancer treatment and improve patients' clinical outcomes.
Collapse
Affiliation(s)
- Lan Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, China
| | - Yiyan Miao
- The Jiangyin Clinical College of Xuzhou Medical University, China
| | - Yihang Gu
- Department of Geriatrics, The Jiangyin Clinical College of Xuzhou Medical University, China
| | - Tomoya Kawaguchi
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka Metropolitan University, Japan
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, Japan
| | - Megumi Mizutani
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka Metropolitan University, Japan
| | - Toshiyuki Nakai
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka Metropolitan University, Japan
| | - Tetsuya Watanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka Metropolitan University, Japan
| | - Kazuhisa Asai
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka Metropolitan University, Japan
| | - Hui Zhang
- The Jiangyin Clinical College of Xuzhou Medical University, China
| | | | - Yoko Tani
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, Japan
| | - Hiroyasu Kaneda
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, Japan
| |
Collapse
|
46
|
Wang G, Pan S. Factor analysis of postsurgical gastroparesis syndrome after right hemicolectomy for colon cancer. Oncol Lett 2025; 29:154. [PMID: 39898286 PMCID: PMC11782927 DOI: 10.3892/ol.2025.14900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
The present study aimed to investigate factors influencing postsurgical gastroparesis syndrome (PGS) in patients with right-sided colon cancer. In total, 260 patients who underwent complete mesocolic excision for right-sided colon cancer were included in the present analysis. Among the included patients, 69 underwent open radical right-sided colon resection, 175 underwent laparoscopic radical right-sided colon resection and 16 underwent robot-assisted radical right-sided colon resection. The occurrence of PGS was observed, and both the χ2 test and multivariate regression analysis were conducted to identify influencing factors. Among the 260 patients, 32 experienced PGS, with an incidence rate of 12.3%. Univariate analysis demonstrated that age, perioperative blood glucose levels, self-rated anxiety scale scores and surgical approach were significantly associated with PGS (P<0.05), whereas sex, surgical duration, diabetes and perioperative albumin levels were not significant factors (P>0.05). Multivariate logistic regression analysis showed that age >70 years, perioperative blood glucose ≥11.1 mmol/l, a self-rating anxiety scale score ≥50 and radical extended right-sided colon resection were risk factors for PGS occurrence. In conclusion, the occurrence of PGS in patients with right-sided colon cancer was revealed to be associated with age, perioperative blood glucose levels, self-rated anxiety scale scores and surgical approach.
Collapse
Affiliation(s)
- Gang Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Shengjie Pan
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
47
|
Majewska O, Pach R, Brzewski P, Kulig J, Kulig P. Impact of Clinicopathological Features on Gastric Cancer Stage According to TNM Classification. In Vivo 2025; 39:1112-1121. [PMID: 40010943 PMCID: PMC11884451 DOI: 10.21873/invivo.13916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 02/28/2025]
Abstract
BACKGROUND/AIM TNM stage is crucial for patients with gastric cancer because curative resection and treatment are only possible in early TNM stages. Therefore, our objective was to assess the association of clinicopathological features with TNM stage in such patients. PATIENTS AND METHODS The association of age, sex, tumor location and Lauren type with TNM stage was analyzed in 910 patients with gastric cancer. RESULTS Age, sex, and tumor location did not have any association with TNM stage in univariate nor multivariate analyses (p>0.05). However, compared to the diffuse and mixed types, the intestinal type (as defined by the Lauren classification) presented lower T stage of gastric cancer in the chi-squared test (p<0.001) and this association was confirmed in the multinominal log normal model (p=0.001). CONCLUSION The histological Lauren type of gastric cancer is associated with lower TNM T stage.
Collapse
Affiliation(s)
- Oliwia Majewska
- Andrzej Frycz Modrzewski Krakow University Faculty of Medicine and Health Sciences, Krakow, Poland
| | - Radosław Pach
- Jagiellonian University Medical College, Krakow, Poland
| | - Paweł Brzewski
- Andrzej Frycz Modrzewski Krakow University Faculty of Medicine and Health Sciences, Krakow, Poland
- Jagiellonian University Medical College, Krakow, Poland
| | - Jan Kulig
- Jagiellonian University Medical College, Krakow, Poland
| | - Piotr Kulig
- Andrzej Frycz Modrzewski Krakow University Faculty of Medicine and Health Sciences, Krakow, Poland;
| |
Collapse
|
48
|
He Q, Sun M, Sun N, Han Q, Shen Y, Li L. Polysocial risk score, lifestyle, genetic factors and risk of incident lung cancer. Public Health 2025; 242:50-57. [PMID: 40024208 DOI: 10.1016/j.puhe.2025.02.024] [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: 10/28/2024] [Revised: 01/24/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVES Lung cancer (LC) is the most frequently diagnosed cancer globally; however, the role of social risk factors in its development is not well understood. While previous studies have identified various lifestyle and genetic factors contributing to LC, the interplay between these elements and broader social determinants of health is still unclear. In this study, we aimed to construct a polysocial risk score (PsRS) that captures the multifaceted nature of social environment risk exposure and its relationship with incident LC, and to explore whether the effect of PsRS is influenced by lifestyle behaviours and heritable risk. STUDY DESIGN Cohort study. METHODS In the UK Biobank cohort, 349,553 participants without previous a cancer diagnosis were recruited. For PsRS construction, 12 social determinants of health were calculated across three domains consistently associated with incident LC. Cox models were used to estimate the association between PsRS and incident LC. Healthy lifestyle and LC genetic risk scores were constructed to evaluate whether lifestyle behaviours and genetic susceptibility modified the effect of PsRS on LC incidence. Mediation analysis was used to estimate whether a healthy lifestyle mediates the effect of PsRS' on LC incidence. RESULTS Compared with participants with low PsRS (≤3), the fully adjusted hazard ratio (HR) (95 % CI) for high PsRS (≥7) in developing LC was 2.75 (2.43-3.12). We observed an additive interaction between PsRS and lifestyle. The proportion of mediation effect of lifestyle in the association between PsRS and LC was 6.41 % (95 % CI: 5.74-7.08 %). Individuals with high PsRS and genetic risk had a 4.63-fold higher risk of incident LC. CONCLUSION A high PsRS is associated with a higher risk of LC, and lifestyle influences this association. High heritable susceptibility and unfavourable social vulnerability may synergistically contribute to higher LC incidence.
Collapse
Affiliation(s)
- Qida He
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong SAR, 999077, China; Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou City, Jiangsu Province, China
| | - Mengtong Sun
- Department of Data Science, City University of Hong Kong, Hong Kong SAR, 999077, China; Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou City, Jiangsu Province, China
| | - Na Sun
- Department of Health Statistics, School of Public Health, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Qiang Han
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou City, Jiangsu Province, China
| | - Yueping Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou City, Jiangsu Province, China.
| | - Linyan Li
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong SAR, 999077, China; Department of Data Science, City University of Hong Kong, Hong Kong SAR, 999077, China.
| |
Collapse
|
49
|
Vincenzi MM, Cicchetti A, Castriconi R, Mangili P, Ubeira-Gabellini MG, Chiara A, Deantoni C, Mori M, Pasetti M, Palazzo G, Tummineri R, Rancati T, Di Muzio NG, Vecchio AD, Fodor A, Fiorino C. Training and temporally validating an NTCP model of acute toxicity after whole breast radiotherapy, including the impact of advanced delivery techniques. Radiother Oncol 2025; 204:110700. [PMID: 39725068 DOI: 10.1016/j.radonc.2024.110700] [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: 06/13/2024] [Revised: 11/21/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE The aim is to train and validate a multivariable Normal Tissue Complication Probability (NTCP) model predicting acute skin reactions in patients with breast cancer receiving adjuvant Radiotherapy (RT). METHODS AND MATERIALS We retrospectively reviewed 1570 single-institute patients with breast cancer treated with whole breast irradiation (40 Gy/15fr). The patients were divided into training (n = 878, treated with 3d-CRT, from 2009 to 2017) and validation cohorts (n = 692, treated from 2017 to 2021, including advanced RT techniques). In the validation cohort, patients were classified according to the delivery techniques into static (n = 404) and arc techniques (n = 288). Several clinical/technical information and DVHs of the "skin" (5 mm inner expansion from the body contour) were available. Skin toxicity was assessed during follow-up using the RTOG scale criteria. A multivariable logistic regression model was generated combining skin DVH and clinical parameters, using cross-validation methods that ensured high internal consistency and robustness. The performance of the model was tested in the validation cohort. RESULTS 14.0 %/17.4 % of patients developed ≥ G2 toxicity, in the training/validation cohorts, respectively. The resulting multivariable logistic model included axillary lymph node dissection (OR = 1.58, 95 %CI = 1.01-2.48, p = 0.045), hypertension (OR = 1.54, 95 %CI = 1.04-2.27, p = 0.030) and skin V20Gy (OR = 1.008, 95 %CI = 1.004-1.013, p < 0.0001). The AUC of the model was 0.64/0.59 in training/validation, with better performance in the validation cohort if considering only V20Gy (0.62). The model showed satisfactory agreement between predicted and observed toxicity rates: in the validation group, the slope of the calibration plot was 0.96 (R2 = 0.6) with excellent goodness-of-fit (Hosmer-Lemeshow p-value = 0.99). Looking at each of the three predictors individually, only the role of V20Gy was confirmed in the validation group. Results were similar when considering patients treated with static or arc techniques. CONCLUSION An NTCP model for acute toxicity after moderately hypofractionated breast RT was trained. The model underwent temporal validation even for patients treated with advanced delivery techniques. Despite clinical differences and techniques, the confirmation of the dosimetry parameter in the validation cohort highlights its robustness and corroborates the hypothesis that skin DVH may assess the risk with the potential for improving plan optimisation.
Collapse
Affiliation(s)
| | - Alessandro Cicchetti
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Data Science Unit, Milan, Italy
| | - Roberta Castriconi
- IRCCS San Raffaele Scientific Institute, Medical Physics Dept., Milan, Italy
| | - Paola Mangili
- IRCCS San Raffaele Scientific Institute, Medical Physics Dept., Milan, Italy
| | | | - Anna Chiara
- IRCCS San Raffaele Scientific Institute, Radiotherapy Dept., Milan, Italy
| | - Chiara Deantoni
- IRCCS San Raffaele Scientific Institute, Radiotherapy Dept., Milan, Italy
| | - Martina Mori
- IRCCS San Raffaele Scientific Institute, Medical Physics Dept., Milan, Italy
| | - Marcella Pasetti
- IRCCS San Raffaele Scientific Institute, Radiotherapy Dept., Milan, Italy
| | - Gabriele Palazzo
- IRCCS San Raffaele Scientific Institute, Medical Physics Dept., Milan, Italy
| | - Roberta Tummineri
- IRCCS San Raffaele Scientific Institute, Radiotherapy Dept., Milan, Italy
| | - Tiziana Rancati
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Data Science Unit, Milan, Italy
| | - Nadia Gisella Di Muzio
- IRCCS San Raffaele Scientific Institute, Radiotherapy Dept., Milan, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | | | - Andrei Fodor
- IRCCS San Raffaele Scientific Institute, Radiotherapy Dept., Milan, Italy
| | - Claudio Fiorino
- IRCCS San Raffaele Scientific Institute, Medical Physics Dept., Milan, Italy.
| |
Collapse
|
50
|
Xi M, Luo X, Chen F, Wang Z, Xiao X, Luo B, Chen M, Gan T, Yang J, Deng K. Iodine Staining With Distance Countdown Improving the Safety for Reduction of Adverse Events: A Randomized Controlled Trial. Clin Transl Gastroenterol 2025; 16:e00822. [PMID: 39836052 PMCID: PMC11932585 DOI: 10.14309/ctg.0000000000000822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Lugol chromoendoscopy (LCE) is valuable, cost-effective, and widely used in early esophageal cancer screening, yet it suffers from low compliance because of adverse events after LCE. In addition, the reflux of iodine during iodine staining in the upper esophagus brings the risk of bucking and aspiration. We introduced a new model called distance countdown (DC) aimed to reduce reflux during iodine staining in upper esophageal LCE. METHODS In this randomized controlled trial, 204 patients were randomized into the DC and No-DC groups. The primary end point was the difference in the incidence of positive starch reagent reaction (iodine solution reflux) between the 2 groups. The secondary end points were the comparisons of the incidence of other adverse events after LCE between the 2 groups. RESULTS The rate of iodine solution reflux was 1.0% in the DC group and 26.5% in the No-DC group ( P < 0.001). Furthermore, the incidences of bucking between the 2 groups were 1.0% and 9.8% ( P = 0.005). LCE satisfaction rates were 78.4% and 76.5% in the DC and No-DC groups ( P = 0.363), respectively. Concerning symptoms after LCE, incidences of sore throat, pharyngeal discomfort or odor, bitter taste, and heartburn were also reduced in the DC group (all P < 0.05). DISCUSSION Adding DC as an auxiliary effect during LCE would reduce the risk of iodine solution reflux, as well as other adverse events after LCE. Implementing this measure could be beneficial in improving the safety of LCE in early esophageal cancer screening.
Collapse
Affiliation(s)
- Mingjia Xi
- Department of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xinyue Luo
- Department of Gastroenterology & Hepatology, The First People's Hospital of Longquanyi District, Chengdu, Sichuan, China
| | - Feifan Chen
- Department of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhu Wang
- Department of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xue Xiao
- Department of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Binyang Luo
- Department of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mo Chen
- Department of Gerontology, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan, China
- Department of Gerontology, Tibetan Chengdu Branch Hospital of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Gan
- Department of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinlin Yang
- Department of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kai Deng
- Department of Gastroenterology & Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|