101
|
Mohammed YM, Hammood ZD, Salih AM, Yasseen HA, Tahir SH, Omar SS, Ali RH, Ali RM, Radha BMM, Mustafa AM, Kakamad FH. Ovarian metastasis from lobular breast carcinoma: A case report with review of literature. Radiol Case Rep 2025; 20:3099-3105. [PMID: 40242375 PMCID: PMC12002769 DOI: 10.1016/j.radcr.2025.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/18/2025] Open
Abstract
Although breast cancer can metastasize to various sites, involvement of the ovaries is infrequent. The current study aims to report a case of ovarian cancer originating from the breast along with an in-depth analysis of the literature. A 58-year-old woman with a history of invasive lobular carcinoma underwent surgery followed by adjuvant chemo-radiotherapy. During regular follow-up, a suspicious ovarian mass was detected, later confirmed to be metastasis from the breast cancer. A review of 25 studies on Google Scholar and PubMed identified 7185 ovarian metastasis patients, with 1253 cases originating from breast cancer, accounting for approximately 17.4% of all metastatic ovarian cancers. Bilateral ovarian involvement was noted in 75% of cases, with right-side involvement in 5.7% and left-side in 19.3%. Only 13 studies documented menopausal status, showing 53.6% were premenopausal. Controversies persist in distinguishing primary ovarian cancer from metastasis using clinical signs, serum markers, and imaging. Metastasis of breast cancer to the ovaries is an uncommon event, even after utilizing various therapeutic approaches. Surgery is the treatment of choice for these cases. This case highlights the importance of long-term surveillance in breast cancer patients and emphasizes the role of surgery in managing ovarian metastases.
Collapse
Affiliation(s)
- Yasameen M. Mohammed
- Department of Surgery, Tikrit Teaching Hospital, College of Medicine Street, Saladin, Iraq
| | - Zuhair D. Hammood
- Department of Surgery, Tikrit Teaching Hospital, College of Medicine Street, Saladin, Iraq
| | - Abdulwahid M. Salih
- College of Medicine, University of Sulaimani, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
- Scientific Affairs Department, Smart Health Tower, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
| | - Hadeel A. Yasseen
- College of Medicine, University of Sulaimani, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
- Scientific Affairs Department, Smart Health Tower, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
| | - Soran H. Tahir
- College of Medicine, University of Sulaimani, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
- Scientific Affairs Department, Smart Health Tower, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
| | - Sami S. Omar
- Kscien Organization for Scientific Research, Hamdi Street, Azadi Mall, Sulaymaniyah, Kurdistan, Iraq
- Department of Oncology, Rizgary Oncology Center, Peshawa Qazi Street, Erbil, Kurdistan, Iraq
| | - Rebaz Haji Ali
- Scientific Affairs Department, Smart Health Tower, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
- Department of Oncology, Hiwa Cancer Hospital, Sulaymaniyah, Kurdistan, Iraq
| | - Rawa M. Ali
- Scientific Affairs Department, Smart Health Tower, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
| | - Belan Mikaeil M. Radha
- College of Medicine, University of Sulaimani, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
| | - Ayman M. Mustafa
- Scientific Affairs Department, Smart Health Tower, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
| | - Fahmi H. Kakamad
- College of Medicine, University of Sulaimani, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
- Scientific Affairs Department, Smart Health Tower, Madam Mitterrand Street, Sulaymaniyah, Kurdistan, Iraq
- Kscien Organization for Scientific Research, Hamdi Street, Azadi Mall, Sulaymaniyah, Kurdistan, Iraq
| |
Collapse
|
102
|
Nakauchi C, Masunaga N, Kagara N, Oshiro C, Shimoda M, Shimazu K. Development of a prediction model for ctDNA detection (Cir-Predict) in breast cancer. Breast Cancer Res Treat 2025; 211:331-339. [PMID: 40055250 PMCID: PMC12006266 DOI: 10.1007/s10549-025-07647-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 02/10/2025] [Indexed: 04/18/2025]
Abstract
PURPOSE The detection of circulating tumor DNA (ctDNA) is a valuable method to predict the risk of recurrence and to detect real-time gene changes. The amount of ctDNA is affected by many factors. Moreover, the detection rate of ctDNA varies from report to report. METHODS The present study evaluated differentially expressed genes using a DNA microarray assay for gene expression in tumors with and without detected ctDNA and constructed a prediction model for the detectability of ctDNA in breast tumor tissues. The model, named Cir-Predict, consisted of 126 probe sets (111 genes) and was constructed in a training set of breast cancer patients (n = 35) and validated in a validation set (n = 13). RESULTS The accuracy, sensitivity, and specificity in training and validation sets were over 90%, and Cir-Predict was significantly associated with ctDNA detection independently of the other conventional clinicopathological parameters in training and validation sets (P < 0.001, P = 0.014, respectively). Cir-Predict (+) was significantly associated with worse recurrence-free survival (P = 0.006). Pathway analysis revealed that nine pathways including tight junction and cell cycle tended to be related to ctDNA detectability. CONCLUSION Cir-Predict not only provides information useful for breast cancer treatment, but also helps the understanding of the mechanism by which ctDNA is detected.
Collapse
Affiliation(s)
- Chiaki Nakauchi
- Department of Breast Surgery, ISEIKAI International General Hospital, 4-14 Minamioogimachi, Kita-ku, Osaka City, Osaka, Japan.
| | - Nanae Masunaga
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Naofumi Kagara
- Department of Breast Surgery, Osaka General Medical Center, 3-1-56, Bandai-Higashi, Sumiyoshi-ku, Osaka City, Osaka, 558-8558, Japan
| | - Chiya Oshiro
- Department of Breast Surgery, Kaizuka City Hospital, 3-10-20 Ichibori, Kaizuka, Osaka, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
103
|
Xia Q, Hao B, Liu C, Cao Y. Inhibitory effect and mechanism of purified saponin from Polygonatum cyrtonema Hua on human ovarian cancer cell A2780. Food Chem Toxicol 2025; 200:115368. [PMID: 40049280 DOI: 10.1016/j.fct.2025.115368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/23/2025]
Abstract
Polygonatum cyrtonema Hua (PcH) is a traditional medicinal and edible homologous plant. Here, the anticancer activity of different purified PcH saponins (PcHs) on the ovarian cancer (OC) cell A2780 were investigated. The PcHs were purified by AB-8. Their effects on the apoptosis, migration and invasion of A2780 were explored by CCK8, wound healing assay, Hoechst 33258 staining, Annexin V-FITC/PI and intracellular reactive oxygen species (ROS) measurement. Moreover, western blot and realtime-PCR confirmation were performed for the associated protein and mRNA targets. The results showed that the content of PcHs increased from 0.31 ± 0.02 mg/mL to 0.79 ± 0.02 mg/mL after purification. The inhibition rate of purified PcHs on A2780 could reach 66.76 ± 0.56%. Specifically, it promoted cell apoptosis, disturbed the cell cycle, increased the ROS levels, regulated the expression levels of pro-apoptotic and anti-apoptotic proteins. PcHs can also regulate the expression level of proteins related to the invasion and migration characteristics of A2780, the mRNA expression level related to the P13K-Akt signal transduction process, and the extracellular matrix-receptor interaction pathway, as well as drug resistance. Overall, this study provides a new idea for the treatment of OC.
Collapse
Affiliation(s)
- Qile Xia
- Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Beiqiong Hao
- Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Chenxing Liu
- Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Yan Cao
- Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| |
Collapse
|
104
|
Li F, Du X, Han M, Feng X, Jiang C. Targeting ETHE1 inhibits tumorigenesis in vitro and in vivo by preventing aerobic glycolysis in gastric adenocarcinoma cells. Oncol Lett 2025; 29:286. [PMID: 40264823 PMCID: PMC12012436 DOI: 10.3892/ol.2025.15032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/20/2025] [Indexed: 04/24/2025] Open
Abstract
Gastric adenocarcinoma (GAC) is a prevalent form of cancer that frequently displays abnormal metabolism characterized by increased aerobic glycolysis. Therefore, inhibition of glycolysis may exhibit therapeutic potential for the management of advanced or recurrent gastric cancer. Analysis of ethylmalonic encephalopathy protein 1 (ETHE1) expression levels in 30 pairs of cancerous and paracancerous tissues, and 50 tumor tissue sections collected from patients with GAC revealed that ETHE1 expression was upregulated in cancerous tissues compared with in paracancerous tissues. Advanced tumor stage, lymph node metastasis and Tumor-Node-Metastasis stage were associated with high ETHE1 expression. Knockdown of ETHE1 expression in GAC cells resulted in a significant inhibition of cell proliferation and in cell cycle arrest, accompanied by downregulated levels of cyclin D1 and cyclin-dependent kinase 4. ETHE1 knockdown also resulted in increased apoptosis of GAC cells, and increased caspase-3 and caspase-9 activity. Additionally, the expression levels of proteins associated with aerobic glycolysis were downregulated following ETHE1 knockdown, which may reduce glucose consumption, lactic acid production and ATP levels. In the in vivo experiments, suppressed tumor growth and increased tumor cell apoptosis were observed in the xenograft tumor model in animals injected with ETHE1-knockdown GAC cells. In summary, knockdown of ETHE1 inhibited aerobic glycolysis, promoted apoptosis and inhibited tumor cell proliferation in GAC cells. These results highlight ETHE1 as a promising molecular target for the treatment of GAC potentially using an adjuvant to target it, offering a novel approach in the exploration of targeted therapeutic drugs for GAC.
Collapse
Affiliation(s)
- Fangfei Li
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Xuan Du
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Mei Han
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Xiaoying Feng
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Chunmeng Jiang
- Department of Gastroenterology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| |
Collapse
|
105
|
Ameji PJ, Shtaiwi A, Adnan R. Virtual screening, in silico pharmacokinetic and toxicity profiling of colchicine-based inhibitors of estrogen receptor of breast cancer. Toxicol Rep 2025; 14:101926. [PMID: 39968053 PMCID: PMC11833621 DOI: 10.1016/j.toxrep.2025.101926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
The declining efficacies of existing drugs against estrogen receptor positive (ER+) breast cancer due to multidrug resistance, acute toxicities, and poor pharmacokinetic properties has necessitated the discovery of newer ones. In this study, colchicine analogues with proven in vitro activities against breast cancer cells were screened against estrogen receptor alpha (ERα) via molecular docking simulations to identify some promising drug candidates. The identified ligands were further subjected to MM/GBSA calculations to ascertain their solvation-dependent Gibb's free energy of binding (∆GB). Three most promising ligands (MPLs); 12, 16, and 21 with ∆GB values of - 40.37, - 40.31, and - 40.26 kcal/mol, respectively, were identified. When compared with tamoxifen (standard drug) whose ∆GB value is - 38.66 kcal/mol, the MPLs appear more potent. The kinetic stabilities of 12, 16, and 21 were confirmed by DFT (B3LYP/6-31G*) calculations and the time-dependent thermodynamic stabilities of their complexes with ERα were established by molecular dynamic simulations. In addition, the MPLs display positive pharmacokinetic and toxicity profiles and could be excellent sources of potent and non-toxic drug candidates against ER+ breast carcinoma.
Collapse
Affiliation(s)
- Philip John Ameji
- School of Chemical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
- Department of Chemistry, Federal University Lokoja, P.M.B. 1154, Lokoja, Nigeria
| | - Amneh Shtaiwi
- Chemistry Department, Faculty of Science, Applied Science Private University, Amman 11931, Jordan
| | - Rohana Adnan
- School of Chemical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| |
Collapse
|
106
|
Gou M, Zhang H, Qian N, Zhang Y, Sun Z, Li G, Wang Z, Dai G. Deep learning radiomics analysis for prediction of survival in patients with unresectable gastric cancer receiving immunotherapy. Eur J Radiol Open 2025; 14:100626. [PMID: 39807092 PMCID: PMC11728962 DOI: 10.1016/j.ejro.2024.100626] [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: 12/03/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Immunotherapy has become an option for the first-line therapy of advanced gastric cancer (GC), with improved survival. Our study aimed to investigate unresectable GC from an imaging perspective combined with clinicopathological variables to identify patients who were most likely to benefit from immunotherapy. Method Patients with unresectable GC who were consecutively treated with immunotherapy at two different medical centers of Chinese PLA General Hospital were included and divided into the training and validation cohorts, respectively. A deep learning neural network, using a multimodal ensemble approach based on CT imaging data before immunotherapy, was trained in the training cohort to predict survival, and an internal validation cohort was constructed to select the optimal ensemble model. Data from another cohort were used for external validation. The area under the receiver operating characteristic curve was analyzed to evaluate performance in predicting survival. Detailed clinicopathological data and peripheral blood prior to immunotherapy were collected for each patient. Univariate and multivariable logistic regression analysis of imaging models and clinicopathological variables was also applied to identify the independent predictors of survival. A nomogram based on multivariable logistic regression was constructed. Result A total of 79 GC patients in the training cohort and 97 patients in the external validation cohort were enrolled in this study. A multi-model ensemble approach was applied to train a model to predict the 1-year survival of GC patients. Compared to individual models, the ensemble model showed improvement in performance metrics in both the internal and external validation cohorts. There was a significant difference in overall survival (OS) among patients with different imaging models based on the optimum cutoff score of 0.5 (HR = 0.20, 95 % CI: 0.10-0.37, P < 0.001). Multivariate Cox regression analysis revealed that the imaging models, PD-L1 expression, and lung immune prognostic index were independent prognostic factors for OS. We combined these variables and built a nomogram. The calibration curves showed that the C-index of the nomogram was 0.85 and 0.78 in the training and validation cohorts. Conclusion The deep learning model in combination with several clinical factors showed predictive value for survival in patients with unresectable GC receiving immunotherapy.
Collapse
Affiliation(s)
- Miaomiao Gou
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Hongtao Zhang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Yong Zhang
- Department of Medical Oncology, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Zeyu Sun
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Guang Li
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Zhikuan Wang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Guanghai Dai
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| |
Collapse
|
107
|
Alizade-Harakiyan M, Khodaei A, Yousefi A, Zamani H, Mesbahi A. Decision tree-based machine learning algorithm for prediction of acute radiation esophagitis. Biochem Biophys Rep 2025; 42:101991. [PMID: 40230494 PMCID: PMC11995095 DOI: 10.1016/j.bbrep.2025.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Background Radiation-induced esophagitis remains a significant challenge in thoracic and neck cancer treatment, impacting patient quality of life and potentially limiting therapeutic efficacy. This study aimed to develop and validate a decision tree-based model for predicting acute esophagitis grades in patients undergoing chemoradiotherapy. Methods Data from 100 patients receiving thoracic and neck radiotherapy were analyzed. The dataset comprised 33 features, including demographic, clinical, and dosimetric parameters. A decision tree classifier was implemented for both binary (Grade ≥2 vs. <2) and multi-class (Grades 1, 2, and 3) classification. Model performance was evaluated using standard metrics including accuracy, precision, recall, and F1-score. Results The binary classification model achieved 97 % accuracy in distinguishing acute esophagitis. The multi-class model demonstrated 98 % accuracy in predicting specific grades. Key predictive features included V40 (volume receiving 40 Gy), V60, and average esophageal dose. The model generated interpretable decision rules, with V60 ≥ 2.3 strongly indicating Grade 3 esophagitis. Conclusions The decision tree model demonstrates high accuracy in predicting radiation-induced esophagitis grades while maintaining clinical interpretability. This approach offers potential for treatment optimization and personalized risk assessment in radiotherapy planning. The model's transparency and reliability make it a promising tool for clinical decision support in radiation oncology.
Collapse
Affiliation(s)
- Mostafa Alizade-Harakiyan
- Department of Radiation Oncology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Khodaei
- Faculty of Electrical and Computer Engineering, University of Tabriz, Tabriz, Iran
| | - Ali Yousefi
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Zamani
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asghar Mesbahi
- Medical Physics Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
108
|
Xu J, Pei Z, Wang Y, Jiang N, Gong Y, Gong F, Ni C, Cheng L. Bioactive microspheres to enhance sonodynamic-embolization-metalloimmune therapy for orthotopic liver cancer. Biomaterials 2025; 317:123063. [PMID: 39753085 DOI: 10.1016/j.biomaterials.2024.123063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/07/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025]
Abstract
The development of novel microspheres for the combination of sonodynamic therapy (SDT) with transarterial embolization (TAE) therapy to amplify their efficacy has received increasing attention. Herein, a novel strategy for encapsulating sonosensitizers (e.g., oxygen-deficient manganese tungstate (MnWOX) nanodots) with gelatin microspheres was proposed. The obtained MnWOX-encapsulated microspheres (abbr. Mn-GMSs) facilitated efficient sonodynamic-embolization-metalloimmune therapy via the immune effects of metal ions on orthotopic liver cancer tumor after transarterial embolization (TAE). Due to the strong cavitation effect caused by the porous structure, Mn-GMSs exhibited a greater reactive oxygen species (ROS) generation rate than the free MnWOX nanodots under US irradiation. Efficient SDT revealed robust cell-killing effects and triggered strong immunogenic cell death (ICD). Moreover, the Mn ions released from the bioactive Mn-GMSs further stimulated the dendritic cells (DCs) maturation and triggered the activation of the cGAS/STING pathway to enhance the immunological effect. Thus, Mn-GMSs achieved significant SDT therapeutic outcomes in H22 tumors in mice, and the combination of the Mn-GMSs triggered SDT with programmed cell death ligand 1 (PD-L1) antibodies could further enhance therapeutic outcomes. The Mn-GMSs exhibited high ROS generation efficacy under US irradiation, significant immune activation, good efficacy in combination with immune checkpoint inhibitor, and great potential for artery embolization-assisted drug delivery, thus enabling effective destruction of liver tumors in rats and rabbits. Therefore, this work provides a strategy for applying SDT in deep tumors and highlights a promising sonodynamic-embolization therapy for combating liver cancers.
Collapse
Affiliation(s)
- Jiachen Xu
- Department of Vascular Surgery and Interventional Radiology, The Forth Affiliated Hospital of Soochow University, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215125, China; Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yuanjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Nan Jiang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yuehan Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Fei Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China.
| | - Caifang Ni
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
109
|
Lee KJ, Ahn JH, Kim JH, Lee YS, Lee JS, Lee JH, Kim TJ, Choi JH. Non-coding RNA RMRP governs RAB31-dependent MMP secretion, enhancing ovarian cancer invasion. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167781. [PMID: 40057205 DOI: 10.1016/j.bbadis.2025.167781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Non-coding RNAs (ncRNAs) are frequently dysregulated in various cancers and have been implicated in the etiology and progression of cancer. Ovarian cancer, the most fatal gynecological cancer, has a poor prognosis and a high patient fatality rate due to metastases. In this study, we classified patients with ovarian cancer into three groups based on their ncRNA expression levels. Notably, an ncRNA transcribed by RNA polymerase III, RNA component of mitochondrial RNA processing endoribonuclease (RMRP), is highly expressed in a group with a poor prognosis. Functional assays using SKOV3 and HeyA8 human ovarian cancer cell lines revealed that while RMRP modulation had no significant effect on cell viability, it markedly enhanced cell invasion. Knockdown and ectopic expression experiments demonstrated that RMRP promotes the secretion of matrix metalloproteinase (MMP)-2 and -9, thereby facilitating ovarian cancer cell invasiveness. Transcriptomic analysis further revealed a positive correlation between RMRP expression and genes involved in cellular localization, including RAB31, a member of the Ras-related protein family. Notably, RAB31 knockdown abrogated the pro-invasive effects of RMRP, identifying it as a key downstream effector in SKOV3 and HeyA8 cells. In addition, MechRNA analysis identified RAB31 as a putative RMRP-interacting transcript. These findings establish RMRP as a critical regulator of RAB31-dependent MMP secretion and ovarian cancer cell invasion. Moreover, our results suggest that RMRP could serve as a promising prognostic biomarker for ovarian cancer.
Collapse
Affiliation(s)
- Ki Jun Lee
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, South Korea; College of Pharmacy, Kyung Hee University, South Korea
| | - Ji-Hye Ahn
- Department of Korean Pharmacy, College of Pharmacy, Woosuk University, South Korea
| | - Jin-Hyung Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, South Korea
| | - Yong Sun Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, South Korea
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Jae-Hyung Lee
- Department of Oral Microbiology, College of Dentistry, Kyung Hee University, South Korea
| | - Tae Jin Kim
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, South Korea
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, South Korea; College of Pharmacy, Kyung Hee University, South Korea.
| |
Collapse
|
110
|
Zhang Y, Liu H, Zhen W, Jiang T, Cui J. Advancement of drugs conjugated with GalNAc in the targeted delivery to hepatocytes based on asialoglycoprotein receptor. Carbohydr Res 2025; 552:109426. [PMID: 40068307 DOI: 10.1016/j.carres.2025.109426] [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: 12/03/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 04/22/2025]
Abstract
The asialoglycoprotein receptor (ASGPR) is specifically expressed in hepatocytes. Sugar molecules, such as asialoglycoprotein, galactose, galactosamine, and N-acetyl galactosamine (GalNAc), have a high affinity for ASGPR. This review summarizes the structure of ASGPR, the distribution of this molecule in different cells, and the factors influencing the binding of GalNAc to ASGPR. We introduce the application of GalNAc in targeted delivery into hepatocytes by forming conjugated compounds with RNAs and small molecules, and the standard methods for synthesizing GalNAc are also briefly presented. This is to provide an overview of the current research on GalNAc and to shed light on the design of the new GalNAc.
Collapse
Affiliation(s)
- Yafang Zhang
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Hongliang Liu
- Pharmaron Beijing Co., Ltd. (China), Beijing, 100176, China
| | - Weina Zhen
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Tingting Jiang
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Jingxuan Cui
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
111
|
Colciago RR, Ferrario F, Chissotti C, Rossano G, De Sanctis L, Faccenda V, Panizza D, Trivellato S, Arcangeli S. Long-term outcomes of volume de-escalation for breast nodal irradiation. Breast Cancer Res Treat 2025; 211:375-384. [PMID: 39992611 PMCID: PMC12006277 DOI: 10.1007/s10549-025-07652-3] [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: 10/01/2024] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION NCCN recommendations suggest irradiating chest wall/breast only + regional node irradiation (RNI) of the undissected axillary levels for node-positive breast cancer (BC) patients. We retrospectively analyzed a cohort of node-positive BC patients who received adjuvant radiotherapy (RT) with a volume de-escalation at the level of axillary nodes. MATERIAL AND METHODS We conducted a retrospective analysis of node-positive BC patients treated with adjuvant RT administered following a conventional fractionation schedule using a 3D-conformal technique to the chest wall or breast and only the IV axillary level. The primary endpoint of the study was disease free survival (DFS). Secondary endpoints included loco-regional control (LRC), and Overall Survival (OS). Toxicity was documented according to the Radiation Therapy Oncology Group (RTOG) criteria. RESULTS A total cohort of 343 patients was analyzed. Loco-regional recurrence occurred in 100 (29.1%). The 5- and 10-year Kaplan-Meyer curves for DFS were 81.4% (95% CI: 79.3%-83.5%) and 60.9% (95% CI: 57.6%-64.5%), respectively. Multivariate Cox analysis confirmed that lymph node ratio (HR = 9.76, 95% CI: 3.12-30.53, p = 0.0001), Luminal B subtype (HR = 2.03, 95% CI: 1.26-3.29, p = 0.004), and triple-negative subtype (HR = 2.70, 95% CI: 1.22-5.99, p = 0.01) were significant predictors of poor DFS. Lymphedema in the ipsilateral arm was reported in 32 (9.3%) patients, primarily Grade 1 or 2. CONCLUSIONS Improved patients' selection and a broader use of systemic therapy could make de-escalation a feasible option. However, this approach should be avoided in patients with extensive nodal involvement, specific molecular subtypes, or comorbidities that prevent the use of chemotherapy.
Collapse
Affiliation(s)
| | - Federica Ferrario
- Medicine and Surgery Department, University of Milan Bicocca, 20126, Milano, Italy
- Radiation Oncology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
| | - Chiara Chissotti
- Medicine and Surgery Department, University of Milan Bicocca, 20126, Milano, Italy.
- Radiation Oncology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Via Pergolesi 33, 20900, Monza, Italy.
| | - Giulia Rossano
- Medicine and Surgery Department, University of Milan Bicocca, 20126, Milano, Italy
- Radiation Oncology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
| | - Lorenzo De Sanctis
- Medicine and Surgery Department, University of Milan Bicocca, 20126, Milano, Italy
- Radiation Oncology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
| | - Valeria Faccenda
- Radiation Oncology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
- Physics Unit, Fondazione IRCCS San Gerardo Dei Tintori, 20900, Monza, Italy
| | - Denis Panizza
- Radiation Oncology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
- Physics Unit, Fondazione IRCCS San Gerardo Dei Tintori, 20900, Monza, Italy
| | - Sara Trivellato
- Radiation Oncology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
- Physics Unit, Fondazione IRCCS San Gerardo Dei Tintori, 20900, Monza, Italy
| | - Stefano Arcangeli
- Medicine and Surgery Department, University of Milan Bicocca, 20126, Milano, Italy
- Radiation Oncology Unit, Fondazione IRCCS San Gerardo Dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
| |
Collapse
|
112
|
Yan B, Dong X, Wu Z, Chen D, Jiang W, Cheng J, Chen G, Yan J. Association of proteomics with lymph node metastasis in early gastric cancer patients. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167773. [PMID: 40048938 DOI: 10.1016/j.bbadis.2025.167773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/13/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025]
Abstract
Surgical decision making for early gastric cancer (EGC) is heavily influenced by its metastasis into the lymph nodes. Currently, the clinicopathological features of EGC cannot be used to accurately distinguish between EGC patients with and without lymph node metastasis. Our retrospective case-matching study included a total of 132 samples from 66 pairs of EGC patients with or without lymph node metastasis and conducted proteomic assays. By comparing the lymph node metastasis group and the nonmetastasis group, we found that two proteins, GABARAPL2 and NAV1, were significantly associated with lymph node metastasis in EGC patients. Our prediction model using protein biomarkers had good prediction accuracy, with an area under the curve (AUC) of 0.87, a sensitivity of 0.78, a specificity of 0.89, and an accuracy of 0.84, which can help distinguish between EGC patients with and without lymph node metastasis and guide the decision-making process for performing tailored surgery.
Collapse
Affiliation(s)
- Botao Yan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Xiaoyu Dong
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China; Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, PR China
| | - Zaizeng Wu
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University, Fuzhou, Fujian 350001, PR China
| | - Dexin Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China; Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, PR China
| | - Wei Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China; Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, PR China
| | - Jiaxin Cheng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China; Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, PR China
| | - Gang Chen
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University, Fuzhou, Fujian 350001, PR China.
| | - Jun Yan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China; Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, PR China.
| |
Collapse
|
113
|
Zhang L, Yuan J, Yao S, Wen G, An J, Jin H, Tuo B. Role of m5C methylation in digestive system tumors (Review). Mol Med Rep 2025; 31:142. [PMID: 40183387 PMCID: PMC11979572 DOI: 10.3892/mmr.2025.13507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Currently, the incidence of digestive system tumors has been increasing annually, thus becoming a prevalent cause of cancer‑related mortalities. Although significant strides have been made in targeting the molecular mechanisms that underpin the development of these tumors, their treatment and prognosis still pose substantial challenges. This is primarily due to the ambiguity of early diagnostic indicators and the fact that most digestive system tumors are detected at an advanced stage. However, epigenetic modifications are capable of altering the expression of oncogenes and regulating biological processes in cancer. In recent years, the study of methylation in relation to tumor pathogenesis has become a focus of prominent research. Among the various types of methylation, 5‑methylcytosine (m5C) methylation plays a crucial role in the development of digestive system tumors and is anticipated to serve as a novel therapeutic target. However, to date, a comprehensive and systematic review concerning the role of m5C methylation in digestive system tumors is lacking. Consequently, the present study reviewed the role of m5C methylation in digestive system tumors such as esophageal cancer, gastric cancer and hepatocellular carcinoma, with the aim of providing a valuable reference for future research endeavors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianbo Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
114
|
Chan WC, Millwood I, Kartsonaki C, Du H, Schmidt D, Stevens R, Chen J, Pei P, Yu C, Sun D, Lv J, Han X, Li L, Chen Z, Yang L. Adiposity and risks of gastrointestinal cancers: A 10-year prospective study of 0.5 million Chinese adults. Int J Cancer 2025; 156:2094-2106. [PMID: 39737804 PMCID: PMC11970548 DOI: 10.1002/ijc.35303] [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: 04/20/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 01/01/2025]
Abstract
Associations of adiposity with risks of oesophageal squamous cell carcinoma (ESCC) and non-cardia stomach cancer, both prevalent in China, are still inconclusive. While adiposity is an established risk factor for colorectal cancer, the relevance of fat-free mass and early-adulthood adiposity remains to be explored. The prospective China Kadoorie Biobank study included 0.5 million adults (aged 30-79 years) from 10 areas in China. Participants' body size and composition were measured at baseline and at resurveys (amongst a subset). After >10 years of follow-up, 2350, 3345 and 3059 incident cases of oesophageal (EC), stomach (SC) and colorectal (CRC) cancers were recorded, respectively. Cox regression was used to estimate hazard ratios (HRs) for these cancers in relation to different adiposity traits. General and central adiposity were inversely associated with EC (primarily ESCC) risk, with HRs of 0.81 (95% CI 0.77-0.85), 0.76 (0.72-0.81) and 0.87 (0.83-0.92) per SD increase in usual levels of BMI, body fat percentage (BF%) and waist circumference (WC), respectively. Adiposity was also inversely associated with SC risk [HR = 0.79 (0.75-0.83) and 0.88 (0.84-0.92) per SD increase in usual BF% and WC], with heterogeneity by cardia and non-cardia subsites, and positively associated with CRC [HR = 1.09 (1.03-1.15) and 1.17 (1.12-1.22) per SD higher usual BF% and WC]. Fat-free mass was inversely associated with EC [HR = 0.93 (0.89-0.98) per SD increase] but positively associated with CRC [1.09 (1.04-1.14)], while BMI at age 25 was positively associated with all three cancers. After mutual adjustment, general adiposity remained inversely associated with EC and SC, while central adiposity remained positively associated with CRC.
Collapse
Affiliation(s)
- Wing Ching Chan
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Iona Millwood
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Huaidong Du
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Daniel Schmidt
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Rebecca Stevens
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Junshi Chen
- China National Center For Food Safety Risk AssessmentBeijingChina
| | - Pei Pei
- Peking University Center for Public Health and Epidemic Preparedness & ResponseBeijingChina
| | - Canqing Yu
- Peking University Center for Public Health and Epidemic Preparedness & ResponseBeijingChina
- Department of Epidemiology and Biostatistics, School of Public HealthPeking University Health Science CenterBeijingChina
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of EducationBeijingChina
| | - Dianjianyi Sun
- Peking University Center for Public Health and Epidemic Preparedness & ResponseBeijingChina
- Department of Epidemiology and Biostatistics, School of Public HealthPeking University Health Science CenterBeijingChina
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of EducationBeijingChina
| | - Jun Lv
- Peking University Center for Public Health and Epidemic Preparedness & ResponseBeijingChina
- Department of Epidemiology and Biostatistics, School of Public HealthPeking University Health Science CenterBeijingChina
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of EducationBeijingChina
| | | | - Liming Li
- Peking University Center for Public Health and Epidemic Preparedness & ResponseBeijingChina
- Department of Epidemiology and Biostatistics, School of Public HealthPeking University Health Science CenterBeijingChina
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of EducationBeijingChina
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Ling Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | | |
Collapse
|
115
|
Jovanovic Ristivojevic J, Jovanovic Korda N, Vujanac V, Nikitovic M, Arsenijevic T. Non‑endemic non‑keratinizing nasopharyngeal carcinoma: Long‑term toxicity following chemoradiation. Oncol Lett 2025; 29:283. [PMID: 40247988 PMCID: PMC12004034 DOI: 10.3892/ol.2025.15029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Chemoradiotherapy (CRT) is considered the standard of care for non-keratinizing nasopharyngeal carcinoma (NK-NPC) worldwide, with improved overall survival, local recurrence-free survival and distant metastasis-free survival rates compared with radiotherapy alone. However, CRT is associated with late toxicities that can diminish a patient's quality of life and increase morbidity and mortality rates. Following the geographical distribution of NK-NPC, research has predominantly been performed on the endemic Asian population of patients. To extrapolate these results, more investigations in non-Asian populations are needed. The present study aimed to analyze the occurrence and severity of late toxicities following CRT strictly in patients with non-endemic NK-NPC. The clinical retrospective study included 36 patients >18 years of age with pathohistologically confirmed NK-NPC who were treated in the Institute of Oncology and Radiology of Serbia (Begrade, Serbia) with CRT during a 5-year period (January 2015 to December 2020). After completing combined treatment with a mean tumor dose of 68.64Gy and a median of 4 cycles of weekly cisplatin (40 mg/m2), late sequelae were routinely assessed during regular follow-ups and graded according to the Radiation Therapy Oncology Group/European Organization for Research and Treatment of Cancer 'Late Radiation Morbidity Scoring Schema'. Overall late toxicities were registered in 83.3% of the patients, mostly at grade ≤2. Neck fibrosis was observed in 69.44% and xerostomia in 58.33% of patients. Late dysphagia was experienced by 2 patients, secondary hypothyroidism by 4 patients and neuropathy by 3 patients. In conclusion, based on the results of the present study, late toxicities can be expected in the majority of patients with non-endemic NK-NPC following CRT. However, late sequelae are of lower grade, with neck fibrosis and xerostomia being the most predominant.
Collapse
Affiliation(s)
- Julija Jovanovic Ristivojevic
- Department of Radiotherapy for Solid Tumors and Hematological Malignancies, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
| | - Natasa Jovanovic Korda
- Department of Radiotherapy for Solid Tumors and Hematological Malignancies, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
| | - Vukac Vujanac
- Department of Radiotherapy for Solid Tumors and Hematological Malignancies, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
| | - Marina Nikitovic
- Department of Radiotherapy for Solid Tumors and Hematological Malignancies, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
- Unit for Clinical Oncology and Radiotherapy, University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia
| | - Tatjana Arsenijevic
- Department of Radiotherapy for Solid Tumors and Hematological Malignancies, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
- Unit for Clinical Oncology and Radiotherapy, University of Belgrade, Faculty of Medicine, 11000 Belgrade, Serbia
| |
Collapse
|
116
|
Ren S, Zhang M, Cai C, Zhang N, Wang Z, Li G, Liu Q, Zhu H, An H, Chen Y. A carrier-free ultrasound-responsive polyphenol nanonetworks with enhanced sonodynamic-immunotherapy for synergistic therapy of breast cancer. Biomaterials 2025; 317:123109. [PMID: 39826335 DOI: 10.1016/j.biomaterials.2025.123109] [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: 09/24/2024] [Revised: 12/29/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Sonodynamic therapy (SDT) is an efficient non-invasive strategy for treating breast cancer. However, the therapeutic efficacy of SDT is greatly limited by various defense mechanisms in the tumor microenvironment, particularly the overexpression of B-cell lymphoma-2 (Bcl-2). In this study, based on drug self-delivery systems, a carrier-free ultrasound-responsive polyphenol nanonetwork (GTC) was developed to enhance SDT by inhibiting Bcl-2. A one-pot method, involving the interaction of the polyphenolic Bcl-2 inhibitor gossypol (GOS), transferrin, and the sonosensitizer chlorin e6 (Ce6), was used to synthesize the GTC. The GTC was efficiently internalized by MDA-MB-231 and 4T1 cells through specific binding to transferrin receptors, and no external carriers were needed. After cellular internalization, GOS increased the lethality of Ce6-mediated SDT by reducing the expression of the Bcl-2 protein, which caused multiple toxic effects. RNA-seq analysis confirmed the transcriptomic alterations in oxidative stress and apoptotic pathways induced by the GTC nanosystem. In vivo studies revealed that GOS-assisted SDT not only eliminated tumors through sonodynamic effects and triggered immunogenic cell death but also enhanced sono-immunotherapy, thus effectively suppressing distant tumors and metastasis. This study might provide insights into carrier-free nanomedicines for SDT-based synergistic tumor therapy.
Collapse
Affiliation(s)
- Shenzhen Ren
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, PR China
| | - Mingzhe Zhang
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Electrical Engineering, Hebei University of Technology, Tianjin, 300401, PR China
| | - Chunxiu Cai
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, PR China
| | - Ning Zhang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, PR China
| | - Zijia Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, PR China
| | - Gen Li
- School of Sciences, Hebei University of Technology, Tianjin, 300401, PR China
| | - Quan Liu
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, PR China
| | - Hailiang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China.
| | - Hailong An
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, PR China.
| | - Yafei Chen
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, PR China
| |
Collapse
|
117
|
Deng Y, Li Y, Cao H. BRD9 promotes the malignant phenotype of thyroid cancer by activating the MAPK/ERK pathway. Anticancer Drugs 2025; 36:359-373. [PMID: 39903580 PMCID: PMC11969370 DOI: 10.1097/cad.0000000000001694] [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: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 02/06/2025]
Abstract
Thyroid cancer is one of the most common endocrine gland malignancies in China. During gene transcription, the bromodomain and extraterminal domain (BET) proteins perform epigenome interpretation tasks. Bromodomain-containing protein 9 (BRD9) is one of the BET family members. Increasing evidence has implicated that BRD9 plays significant roles in multiple malignancies. However, its role in thyroid cancer is still not fully understood. In this research, our results demonstrated that high expression of BRD9 can facilitate the malignant phenotype of thyroid cancer cell lines, while low expression of BRD9 can impede the malignant phenotype of thyroid cancer cell lines. Pharmacologically, I-BRD9 treatment inhibits the proliferation and promotes the rate of apoptosis in thyroid cancer cell lines. Moreover, our results also revealed that BRD9 promoted xenograft tumor growth. In addition, our study showed that the expression of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated protein kinase (ERK) pathway-related proteins was decreased in BRD9 knockdown thyroid cancer cells, such as Raf, ERK, p-ERK, c-Fos, and c-Myc, which could be significantly reversed by overexpressing the BRD9 in different thyroid cancer cells. After the specific inhibitor of ERK (SCH772984) was applied to thyroid cancer cells (BCPAP cells) overexpressing the BRD9 gene, the results suggested that SCH772984 reverses the high expression of MAPK/ERK pathway-associated protein in BCPAP cells (over-expression BRD9 cells). In conclusion, this study demonstrated that BRD9 was highly expressed in serum and malignant tumor tissues of thyroid cancer patients and further promoted the development of the malignant phenotype of thyroid cancer by activating the MAPK/ERK signaling pathway.
Collapse
Affiliation(s)
- Yingcheng Deng
- Department of Anatomy, Hunan Traditional Chinese Medical College, Zhuzhou
| | - Yilin Li
- Department of Breast and Thyroid Surgery, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hong Cao
- Department of Breast and Thyroid Surgery, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
118
|
Mondaca JM, Muñoz JMF, Barraza GA, Vanderhoeven F, Redondo AL, Flamini MI, Sanchez AM. Therapeutic potential of GNRHR analogs and SRC/FAK inhibitors to counteract tumor growth and metastasis in breast cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167826. [PMID: 40189112 DOI: 10.1016/j.bbadis.2025.167826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Breast cancer (BC) is the leading cause of cancer death in women, with hormone-dependent BC accounting for about 80 % of cases, primarily affecting postmenopausal women with gonadotropins, luteinizing hormone (LH), and follicle-stimulating hormone (FSH) elevated. Treatments targeting the gonadotropin-releasing hormone receptor (GnRHR), such as the agonist leuprorelin (LEU) and antagonist degarelix (DEGA), are used for hormone-dependent tumors. While the functional role of gonadotropin receptors in extragonadal tissues remains uncertain, recent studies suggest LH contributes to tumor development and progression. Tumor progression involves reorganization in the actin cytoskeleton, induction of adhesion, and cell migration, driven by proteins such as Src and the focal adhesion kinase (FAK), which are related to invasive behaviors. The overexpression of both protein kinases generates an invasive and metastatic phenotype, then inhibitors targeting Src (PP2) and FAK (FAKi) have been developed to counteract this effect. This study combined GnRH analogs with Src and FAK inhibitors to target BC progression. We found that LH treatment influenced gene expression linked to tumor development. Examining the GnRHR-LEU and GnRHR-DEGA complexes revealed structural differences affecting ligand binding. In an orthotopic tumor model, DEGA reduced tumor growth, while LEU had the opposite effect. Combining DEGA with PP2 or FAKi enhanced tumor inhibition, improving mice survival. These findings provide valuable insights into the essential regulatory role of gonadotropins in genes involved in tumorigenic processes, highlighting the potential of GnRHR antagonists combined with Src or FAK inhibitors as a promising strategy to develop new drugs that interfere with the ability of breast tumor progression.
Collapse
Affiliation(s)
- Joselina Magali Mondaca
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Juan Manuel Fernandez Muñoz
- Departamento de Laboratorio de Salud Pública, Ministerio de Salud y Deportes, Gobierno de Mendoza, Mendoza, Argentina
| | - Gustavo Adolfo Barraza
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Fiorella Vanderhoeven
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Analía Lourdes Redondo
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Marina Inés Flamini
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Angel Matias Sanchez
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
119
|
Abdeljawaad KAA, Abdelrahman AHM, Sidhom PA, Tallima H, Shoeib T, Mekhemer GAH, Sayed SRM, El-Tayeb MA, Hegazy MEF, Ibrahim MAA. Potential P-glycoprotein (P-gp) inhibitors from SuperDRUG2 database toward reversing multidrug resistance in cancer treatment: Database mining, molecular dynamics, and binding energy estimations. J Mol Graph Model 2025; 137:108997. [PMID: 40043638 DOI: 10.1016/j.jmgm.2025.108997] [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: 05/25/2024] [Revised: 12/06/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
P-glycoprotein (P-gp) transporter is included in the failure of various carcinoma chemotherapeutics because of the multidrug resistance (MDR) phenomenon, in which the chemotherapeutic drugs are eliminated from target cells. Consequently, inhibiting P-gp transporter function is a prospective strategy for cancer treatment. In the current study, the SuperDRUG2 database containing >4600 pharmaceutical compounds was virtually screened toward the P-gp transporter utilizing the docking predictions. For inhibitors with a docking score lower than -10.5 kcal/mol, molecular dynamics (MD) simulations were performed, accompanied by binding energy evaluations using the MM-GBSA approach. In accordance with the MM-GBSA//100 ns MD, angiotensin amide (SD003508), terlipressin (SD002603), argipressin (SD002535), and lanreotide (SD001365) exhibited potential binding affinities against the P-gp transporter with ΔGbinding < -120.0 kcal/mol. The outstanding consistency of the investigated inhibitors inside the P-gp binding pocket was shown by the post-dynamics analyses. Additionally, MD simulations of the inhibitor-P-gp complexes in a POPC membrane environment were conducted to mimic the physiological conditions. These results demonstrated that angiotensin amide, terlipressin, argipressin, and lanreotide are promising P-gp inhibitors and deserve additional in-vitro/in-vivo studies.
Collapse
Affiliation(s)
- Khlood A A Abdeljawaad
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt; Academy of Scientific Research & Technology (ASRT), 101 El-Kasr Alaini Street, Cairo, 4262104, Egypt; Frank Laboratory of Neutron Physics, Department of Raman Spectroscopy, Joint Institute for Nuclear Research, Dubna, 141980, Russia.
| | - Alaa H M Abdelrahman
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt.
| | - Peter A Sidhom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Hatem Tallima
- Department of Chemistry, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Tamer Shoeib
- Department of Chemistry, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Gamal A H Mekhemer
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt.
| | - Shaban R M Sayed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.
| | - Mohamed A El-Tayeb
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.
| | - Mohamed-Elamir F Hegazy
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, Mainz, 55128, Germany.
| | - Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, 61519, Egypt; Department of Engineering, College of Engineering and Technology, University of Technology and Applied Sciences, Nizwa 611, Oman; School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa.
| |
Collapse
|
120
|
Wei J, Qin Y, Zhang L, Gong X. Combined protein and transcriptomics identifies DCTPP1 as a putative biomarkers for predicting immunotherapy responsiveness in gastric cancer patients. Anticancer Drugs 2025; 36:415-426. [PMID: 39908235 DOI: 10.1097/cad.0000000000001704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
This study aimed to screen the changes after overexpression of dCTP pyrophosphatase 1 (DCTPP1) in human gastric adenocarcinoma cells (AGS) cells by proteome and transcriptome sequencing. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed to explore the functional significance of the differentially expressed DCTPP1 in gastric cancer (GC). Cell Counting Kit-8 (CCK-8) assay was used to detect the proliferation of cells. Western blot was used to detect the expression of proteins. A total of 28 genes that were significantly associated with DCTPP1 overexpression and had prognostic value were screened by Cox regression analysis. The results of gene set enrichment analysis showed that the genomes of patients with subtype A exhibited significant enrichment in pathways such as DNA repair, pyrimidine synthesis, and glucose metabolism. The tumor immune dysfunction and exclusion and The Cancer Immunome Atlas databases showed that patients with type A GC were better candidates for immunotherapy than patients with type B GC. Furthermore, the CCK-8 assay indicated significantly enhanced proliferative activity after overexpressing DCTPP1 in AGS cells, corroborating the findings from the bioinformatic analysis. The data suggest a potential association between DCTPP1 expression and both the prognosis of GC patients and the efficacy of immunotherapy. These findings offer valuable insights for the potential optimization of therapeutic strategies in gastric cancer.
Collapse
Affiliation(s)
- Jun Wei
- Department of Gastroenterology, General Practice, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical University
| | | | - Luwen Zhang
- School of Clinic Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Xiaobing Gong
- Department of Gastroenterology, General Practice, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province
| |
Collapse
|
121
|
Guo Y, Hu Z, Bai L, Tang Y, Hu J, Zhang Q, Liu J, Feng S. Increased glucose utilization is a targetable vulnerability to overcome drug resistance associated with neddylation blockade. Biochem Pharmacol 2025; 236:116905. [PMID: 40158819 DOI: 10.1016/j.bcp.2025.116905] [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: 12/10/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Gastric cancer, a leading cause of cancer-related mortality, has a median survival of just 15 months in advanced stages and currently lacks effective treatment options. Neddylation blockade is a promising therapeutic strategy, yet its clinical application faces challenge with the emergence of drug resistance. Currently, the underlying mechanisms behind the drug resistance are not fully understood. Our study uncovers the link between MLN4924-induced metabolic reprogramming and its antitumor efficacy in gastric cancer cells. We first demonstrated that MLN4924, a neddylation blocker, has multiple effects on gastric cancer cell growth, notably inducing mitochondrial damage. Untargeted metabolomic analysis revealed that MLN4924 enhances glucose utilization in gastric cancer cells in a concentration-dependent manner. Mechanistically, MLN4924 reduces the neddylation of cullin2, thereby inhibiting the degradation of HIF-1α. This leads to the accumulation of HIF-1α, which upregulates GLUT1 levels and facilitates increased glucose uptake. This metabolic adaptation allows gastric cancer cells to maintain their energy supply despite mitochondrial impairment. Based on the increased glucose dependency following neddylation inhibition by MLN4924, we propose a co-targeting strategy with GLUT1 inhibition, which significantly improves therapeutic efficacy in vitro and in vivo models without safety risks. This dual-targeting approach represents a potent new strategy for gastric cancer treatment.
Collapse
Affiliation(s)
- Yueyang Guo
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Zhuang Hu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Linyue Bai
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Yanjun Tang
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Jingyi Hu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Qianqian Zhang
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Jiali Liu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Siqi Feng
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
122
|
Munusamy MA, Bharathi M, Alarfaj AA, Hussein-Al-Ali SH, Nagaiya R, Subbarayan S. Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles (GNANPs) induce antimicrobial properties and apoptosis in gastric cancer cells. Toxicol In Vitro 2025; 106:106037. [PMID: 40037499 DOI: 10.1016/j.tiv.2025.106037] [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: 08/18/2024] [Revised: 02/08/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
An assessment of the anticancer activity of Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles (GNANPs) against gastric cancer cells was the purpose of this study. The increasing prevalence of gastric cancer and the limitations of conventional therapies necessitate novel approaches that combine targeted drug delivery with therapeutic efficacy. Several techniques were used to characterize the synthesized GNANPs, including UV-visible spectroscopy, X-ray diffractometer (XRD), scanning electron microscope (SEM), transmission electron microscope (TEM), Fourier transform infrared (FT-IR), dynamic light scattering (DLS), and photoluminescence (PL). They were evaluated for their antimicrobial properties, cytotoxicity, ROS accumulation, apoptotic activity, and oxidative stress markers against AGS cells. The characterization analyses indicated the existence of Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles with an oval-shaped morphology and an average particle size of 127.80 nm. The existence of several elements and functional groups in the GNANPs was also detected using EDX and FT-IR analyses, respectively. The synthesized GNANPs have shown exceptional antibacterial activities by effectively inhibiting the growth of several infections. The treatment of GNANPs efficiently inhibited the growth of AGS cells. Fluorescence staining studies showed increased apoptosis and oxidative stress markers in AGS cells treated with synthesized Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles, indicating their potential as a viable cancer treatment option.
Collapse
Affiliation(s)
- Murugan Alwarkurichi Munusamy
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 602 105, India
| | - Muruganantham Bharathi
- Centre for Bioinformatics, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore-641021
| | - Abdullah A Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box.2455, Riyadh 11451, Saudi Arabia
| | | | - Ravichandran Nagaiya
- Department of Life Science and Environmental Biochemistry, Pusan National University, Busan 46241, Republic of Korea
| | - Sarathbabu Subbarayan
- Muthayammal Centre for Advanced Research, Muthayammal College of Arts and Science, Rasipuram, Namakkal, Tamil Nadu 637408, India.
| |
Collapse
|
123
|
Liu H, Li G, Shen C, Qi X, Liu Y, Hua D, Mao Y, Zhang T. B7-H3-mediated deubiquitination stabilizing CYP1B1 expression promotes chemotherapy resistance in colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167771. [PMID: 40057208 DOI: 10.1016/j.bbadis.2025.167771] [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: 01/18/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025]
Abstract
Colorectal cancer (CRC) is the second‑leading cause of cancer-related mortality worldwide. It is frequently characterized by chemotherapy resistance,which is a predominant factor contributing to unfavorable patient prognosis. B7-H3 is a novel tumor marker and a potential immunotherapy target. High B7-H3 expression in colorectal cancer is associated with adverse prognosis. In this study, we noted increased B7-H3 expression in colorectal cancer tumor tissues. Both in vivo and in vitro experiments demonstrated that increased B7-H3 expression promotes resistance to chemotherapy in CRC. Furthermore, our findings suggest that B7-H3 mediates CRC resistance by modulating CYP1B1 expression. Mechanistic investigations indicated that B7-H3 inhibited the ubiquitination of CYP1B1, stabilized its expression,and consequently enhanced chemotherapeutic resistance in CRC. In summary, our results underscore the significance of the B7-H3-CYP1B1 interaction as a crucial therapeutic target for overcoming chemotherapy resistance in CRC.
Collapse
Affiliation(s)
- Huan Liu
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu Province, China
| | - Guifang Li
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu Province, China
| | - Chenjie Shen
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu Province, China
| | - Xiaowei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu Province, China
| | - Yankui Liu
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu Province, China
| | - Dong Hua
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu Province, China
| | - Ting Zhang
- Institute of Cancer, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu Province, China; Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth PL6 8BU, Devon, UK.
| |
Collapse
|
124
|
Dong S, Wang L, Liu X, Hou D, Liu Q, Zheng J, Wang H. CELF2 inhibits bladder cancer progression by decreasing the stability of CXCL5. Life Sci 2025; 370:123585. [PMID: 40154776 DOI: 10.1016/j.lfs.2025.123585] [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: 01/01/2025] [Revised: 03/12/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
AIMS CUGBP Elav-like family (CELF), an RNA-binding protein group, has been implicated in numerous diseases, including cancer. The role of CELF2 in bladder cancer is still not well understood. This study aims to investigate the role of CELF2 in bladder cancer in vitro and in vivo using bioinformatics, biochemical, and functional methods. MATERIALS AND METHODS We explored CELF2 and CELFs expression patterns and their association with bladder cancer by analyzing The Cancer Genome Atlas, University of California, Santa Cruz XENA, and Cancer Cell Line Encyclopedia databases using various computational and statistical analyses, including unsupervised clustering, Kaplan-Meier analysis, and correlation assessments. We utilized the bladder cancer cell lines T24 and J82 for functional analyses. We performed in vitro and in vivo experiments to investigate the impact of CELF2 expression levels on bladder cancer cell proliferation and migration. KEY FINDINGS CELF2 expression was downregulated in bladder cancer and positively correlated with the progression-free interval in patients. Increased CELF2 expression suppressed the proliferation and migration of bladder cancer cells. Furthermore, CELF2 was bound to AU-rich motifs in the 3'-UTR of CXCL5, reducing its stability, inhibiting CXCL5/CXCR2/AKT signaling, and repressing bladder cancer progression. Finally, we developed a prognostic model that revealed CELF2 and CXCL5 as independent prognostic factors for progression-free intervals in patients with bladder cancer. SIGNIFICANCE CELF2 reduced the stability of CXCL5 and suppressed the proliferation and migration of bladder cancer cells by inhibiting p-AKT expression. The findings of this study highlight CELF2 as a potential therapeutic target for bladder cancer treatment.
Collapse
Affiliation(s)
- Shiqiang Dong
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China; Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Key Laboratory of Precision Medicine for Sex Hormones and Diseases, Tianjin 300211, China
| | - Lili Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Key Laboratory of Precision Medicine for Sex Hormones and Diseases, Tianjin 300211, China; Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300211, China
| | - Xinyu Liu
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Dingkun Hou
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Key Laboratory of Precision Medicine for Sex Hormones and Diseases, Tianjin 300211, China
| | - Qing Liu
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Key Laboratory of Precision Medicine for Sex Hormones and Diseases, Tianjin 300211, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Haitao Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Key Laboratory of Precision Medicine for Sex Hormones and Diseases, Tianjin 300211, China.
| |
Collapse
|
125
|
Deng X, Luo X, Fang Z, Chen X, Luo Q. Effect of tristetraprolin on esophageal squamous cell carcinoma cell proliferation. Tissue Cell 2025; 94:102785. [PMID: 39954564 DOI: 10.1016/j.tice.2025.102785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/26/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Tristetraprolin (TTP) can inhibit the abnormal proliferation of malignant tumors but there are no studies involving TTP and esophageal squamous cell carcinoma (ESCC). We aimed to determine the effect of TTP on ESCC cell proliferation and to elucidate the underlying mechanism. METHODS The human ESCC cell line, KYSE-510, and the human ESCC cell line, KYSE-150, stably infected with tetracycline-inducible expression (Tet-on-TTP and Tet-on-EV, respectively) were screened with puromycin. After Tet-on-TTP KYSE-150 cells were treated with different concentrations of doxycycline [Dox] (0, 0.5, and 1 ug/mL), the levels of TTP mRNA and protein expression were detected by real-time fluorescent quantitative PCR and western blotting, respectively. The effects of TTP on proliferation and migration were estimated by CCK-8 and Transwell assays, respectively. Cell apoptosis and cell cycle were measured by flow cytometry. Cellular apoptosis-related gene protein expression was determined by western blotting. RESULTS TTP overexpression significantly inhibited KYSE-510 and KYSE-150 proliferation. TTP overexpression also significantly inhibited KYSE-150 migration. In addition, TTP expression upregulation promoted the KYSE-150 apoptosis and induced cell cycle arrest in the G2 phase, downregulated Bcl-2 expression, and upregulated Bax expression. CONCLUSION TTP inhibited ESCC cell proliferation, promoted ESCC cell apoptosis, and arrested cell cycle progression in the G2 phase.
Collapse
Affiliation(s)
- Xiaoya Deng
- Department of Respiratory Medicine, Shapingba Hospital affiliated to Chongqing University (Shapingba District People's Hospital of Chongqing), Chongqing 400016, China
| | - Xiaoqin Luo
- Out-patient department, Chongqing MingXing Hospital, Chongqing 405200, China
| | - Zhanglan Fang
- General Internal Medicine Department, Chongqing University Cancer Hospital, Chongqing, China
| | - Xinyu Chen
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China
| | - Qinli Luo
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
126
|
Zheng H, Wang Y, Ren Y, Wang X, Sui L, Xu H, Zheng C. Design, synthesis and biological evaluation of sulfur-containing tetrahydroxanthones as potential anti-tumor agents. Bioorg Med Chem Lett 2025; 121:130154. [PMID: 40010444 DOI: 10.1016/j.bmcl.2025.130154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/23/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Given the rising incidence and mortality rates of cancer, the development of highly effective, low-toxicity therapeutics is critical. Xanthones, a class of natural secondary metabolites, are notable for their distinct structure and exhibit promising antitumor activity, underscoring their potential as scaffolds for drug design. Sulfur heterocycles are also valuable in the development of bioactive small molecules. Therefore, we explored the introduction of sulfur in the core structure of xanthones, leading to the synthesis of a series of sulfur-containing tetrahydroxanthones. The in vitro cytotoxicity of these compounds was evaluated using the CCK8 assay, revealing that several derivatives exhibit anti-proliferative effects against HepG2 cells. Among them, compound 4k displayed potent inhibitory activity with an IC50 value of 6.08 μM and showed favorable selectivity, exhibiting low toxicity toward normal cells. Further studies demonstrated that 4k inhibited colony formation and migration of HepG2 cells, and induced apoptosis.
Collapse
Affiliation(s)
- Huimin Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China
| | - Youyi Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China
| | - Yitao Ren
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China
| | - Xueying Wang
- Institute of Medicine and Health Care, Dezhou University, Dezhou 253023, People's Republic of China
| | - Lu Sui
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China.
| | - Changwu Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China.
| |
Collapse
|
127
|
Berk Ş. Comprehensive bibliometric analysis and perspectives on therapies targeting colon cancer stem cells over a 40-year period. Regen Ther 2025; 29:19-34. [PMID: 40124468 PMCID: PMC11930536 DOI: 10.1016/j.reth.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
The presence of cancer stem cells (CSCs) is one of the primary causes of recurring therapy resistance because they have two main capacities: self-renewal and avoiding apoptotic pathways. Despite their relevance, no full bibliometric analysis has yet been done in this topic. The goal of this work is to use bibliometric analysis to map the fundamental and emergent areas in therapeutics targeting colon cancer stem cells. To perform bibliometric analysis on colon cancer stem cells (CCSCs) literature, spanning roughly the last 40 years, in order to establish a firm base for future projections by emphasizing the findings of the most notable research. All information pertinent to CCSCs was accessed from Web of Science Core Collection database. In order to identify and analyze the research hotspots and trends related to this topic, Biblioshiny (RStudio) and VOSviewer were utilized to ascertain the countries/regions, institutions, journals, authors, references, and keywords involved. The targeted time span covered 1735 research-, and review articles. The most frequent keywords were "colorectal cancer," "cancer stem cells," and "colon cancer," while the most trending keywords in the last few years were "protein stability," "spheroid formation," "ubiquitination," "exosomes," "patient-derived organoids," and "gut microbiota." Over the past 40 years, there has been a significant advancement in researchers' understanding of colon cancer stem cells. In addition, the cluster map of co-cited literature showed that colon cancer stem cell research has emerged as a research hotspot. It was also anticipated that the main focus of the future efforts appears to involve clinical applications of cell-targeted colon cancer therapy. These results provide researchers with a comprehensive understanding of this field and provide insightful ideas for further research.
Collapse
Affiliation(s)
- Şeyda Berk
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, 58140, Turkey
| |
Collapse
|
128
|
Li LY, Liang SY, Cai MP, Ge JC, Tan HS, Wang CB, Xu B. Engineered extracellular vesicles as imaging biomarkers and therapeutic applications for urological diseases. Mater Today Bio 2025; 32:101646. [PMID: 40160248 PMCID: PMC11953971 DOI: 10.1016/j.mtbio.2025.101646] [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: 01/01/2025] [Revised: 02/23/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
With the ever-increasing burden of urological diseases, the need for developing novel imaging biomarkers and therapeutics to manage these disorders has never been greater. Extracellular vesicles (EVs) are natural membranous nanoparticles and widely applied in both diagnostics and therapeutics for many diseases. A growing body of research has demonstrated that EVs can be engineered to enhance their efficiency, specificity, and safety. We systematically examine the strategies for achieving targeted delivery of EVs as well as the techniques for engineering them in this review, with a particular emphasis on cargo loading and transportation. Additionally, this review highlights and summarizes the wide range of imaging biomarkers and therapeutic applications of engineered EVs in the context of urological diseases, emphasizing the potential applications in urological malignancy and kidney diseases.
Collapse
Affiliation(s)
- Liao-Yuan Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Si-Yuan Liang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mao-Ping Cai
- Department of Urology, Cancer Center, Fudan University, Shanghai, China
| | - Jian-Chao Ge
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hai-Song Tan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cheng-Bang Wang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
129
|
Rofaeil RR, Sharata EE, Attya ME, Abo-Youssef AM, Hemeida RA, Khalaf MM. Repurposing levomilnacipran to attenuate premature ovarian insufficiency induced by cyclophosphamide in female Wistar albino rats through modulation of TLR4/p38-MAPK/NF-κB p65, caspase-3-driven apoptosis, and Klotho protein expression. Food Chem Toxicol 2025; 200:115406. [PMID: 40154831 DOI: 10.1016/j.fct.2025.115406] [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: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
AIM This study aims to explore the mitigative impact of levomilnacipran (LVM) against cyclophosphamide (CPA)-induced premature ovarian insufficiency by targeting TLR4/p38 MAPK/NF-κB p65, and klotho expression. METHODS Rats were allocated into five groups as follows: control, LVM, CPA, CPA + LVM, and CPA + TRI. Serum hormones and histopathological examination were performed. To assess oxidative stress, ovarian MDA, GSH, and SOD were evaluated. The ovarian contents of caspase-3 and inflammatory markers were assessed using the ELISA method. The expression of ovarian NF-κB p65 was examined using an immunohistochemical technique. RT-qPCR was used to measure Bax and Bcl-2 mRNA expression. Utilizing a Western blot, the TLR4, p38 MAPK, α-Klotho, and cleaved caspase-3 levels were estimated. The estrous cycle was also monitored. FINDINGS LVM attenuated CPA-induced ovarian toxicity by regulating hormones and alleviating histopathological aberrations. It also raised SOD and GSH levels and lowered MDA's ovarian content. Moreover, Bcl-2 levels were raised, Bax and caspase-3 expression levels were reduced, and IL-18, IL-1β, and TNF-α levels were all reduced. LVM-induced ovarian protection by diminishing TLR4/p38 MAPK/NF-κB p65 expression and boosting the protein levels of α-Klotho. SIGNIFICANCE LVM mitigated POI caused by CPA by downregulating TLR4/p38 MAPK/NF-κB p65, enhancing the α-Klotho level and attenuating caspase-3 derived apoptosis.
Collapse
Affiliation(s)
- Remon Roshdy Rofaeil
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt; Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia, 61519, Egypt.
| | - Ehab E Sharata
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt.
| | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia, 61519, Egypt.
| | - Amira M Abo-Youssef
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Ramadan Am Hemeida
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt.
| | - Marwa M Khalaf
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| |
Collapse
|
130
|
Romanczuk P, Zajda J, Matczuk M, Zuchowska A. Multi-Organ-on-Chip approach to study the impact of inter-organ communication on the efficacy and side effects of cancer therapy. Chem Biol Interact 2025; 413:111460. [PMID: 40057013 DOI: 10.1016/j.cbi.2025.111460] [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: 09/26/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 03/24/2025]
Abstract
Cancer is one of the pathological conditions of the human body, which, due to its tissue diversity, is not fully understood. Currently used preclinical in vitro cell or animal models do not reflect the complexity and functional features of the human body, including its pathological conditions such as cancer. This fact is related to poor predictions of the effectiveness of newly developed drugs. Therefore, in our work, we focused on creating a tool that allows the reproduction of important morphological and biochemical features of the tumor in vivo, such as three-dimensional (3D) structure, heterogeneity, the presence of extracellular matrix (ECM), and the appropriate scale (volume to surface ratio). Moreover, the presented Multi-Organ-on-Chip (MOC) tool allows us to evaluate the effects of anticancer therapy, considering hepatic metabolism (liver model) and the assessment of its side effects on a selected organ (skin model). Our research shows that incorporating multiple organ models in one in vitro tool affects the viability and metabolic activity of the cells that constitute them. Moreover, we have shown how important it is to consider hepatic metabolism when evaluating the therapeutic effectiveness of two selected chemotherapy drugs, 5-Fluorouracil (5-FU) and its prodrug Capecitabine (CAP).
Collapse
Affiliation(s)
- Paweł Romanczuk
- Medical Biotechnology, Warsaw University of Technology, Poland
| | - Joanna Zajda
- Analytical Chemistry, Warsaw University of Technology, Poland
| | | | | |
Collapse
|
131
|
Ikpeama CR, Edjekpewhu CLJ. Knowledge of Cervical Cancer Among Women of Childbearing Age: A Qualitative Study of a Local Council in South-Eastern Nigeria. SOCIAL WORK IN PUBLIC HEALTH 2025; 40:216-231. [PMID: 39952781 DOI: 10.1080/19371918.2025.2467399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Abstract
The study investigated the knowledge of cervical cancer among women of childbearing age in a local council of a southeastern Nigerian state, aiming at improving attitudes toward screening uptake and early detection. The health belief model served as a guide for the study's qualitative exploratory research design. Using a combination of purposive and availability sampling strategies, a sample of 20 women participants with varying ages (23-36 years) were selected from 10 villages. The findings revealed that participants have limited knowledge of cervical cancer, with misconceptions and cultural beliefs influencing their understanding. The findings identified factors contributing to the lack of knowledge, including limited access to information and resources, fear, social disapproval, and financial constraints resulting from poverty. The findings also revealed that lack of knowledge has detrimental effects on women's mental, psychological, and social lives, including their relationships with friends and family. In addition, education, awareness campaigns, collaboration with medical personnel, and community engagements were effective strategies to enhance knowledge and awareness of cervical cancer programs among women. The study revealed that the findings imply social work. However, the study recommended that future investigations should consider a larger, more diverse sample to validate these findings and explore strategies to improve knowledge and awareness of cervical cancer among women in Nigeria.
Collapse
|
132
|
Li J, Yang C, Zhang Y, Hong X, Jiang M, Zhu Z, Li J. Deciphering aging-associated prognosis and heterogeneity in gastric cancer through a machine learning-driven approach. iScience 2025; 28:112316. [PMID: 40256325 PMCID: PMC12008712 DOI: 10.1016/j.isci.2025.112316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy with a high mortality rate and limited treatment options. Aging significantly contributes to tumor progression, and GC was confirmed as an aging-related heterogeneous disease. This study established an aging-associated index (AAI) using a machine learning-derived gene panel to stratify GC patients. High AAI scores associated with poor prognosis and indicated potential benefits from adjuvant chemotherapy, while showing resistance to immunotherapy. Single-cell transcriptome analysis revealed that AAI was enriched in monocyte cells within the tumor microenvironment. Two distinct molecular subtypes of GC were identified through unsupervised clustering, leading to the development of a subtype-specific regulatory network highlighting SOX7 and ELK3 as potential therapeutic targets. Drug sensitivity analyses indicated that patients with high ELK3 expression may respond to FDA-approved drugs (axitinib, dacarbazine, crizotinib, and vincristine). Finally, a user-friendly Shiny application was created to facilitate access to the prognostic model and molecular subtype classifier for GC.
Collapse
Affiliation(s)
- Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chuanlai Yang
- Department of Science and Technology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Yunxiao Zhang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Andrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaoning Hong
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Mingye Jiang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhongxu Zhu
- Biomics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Guangdong, Shenzhen, China
| |
Collapse
|
133
|
He M, Xie W, Yuan Z, Chen J, Wang J, Fu Y, Hu Z, Meng Q, Gao W, Hu D, Zhang Y, Pan Y, Zhou Z. Comparing PD-L1 and PD-1 inhibitors plus bevacizumab combined with hepatic arterial interventional therapies in unresetable hepatocellular carcinoma: A single-center, real-world study. Int J Cancer 2025; 156:1972-1985. [PMID: 39834172 DOI: 10.1002/ijc.35341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
With the rise of anti-vascular endothelial growth factor antibody and programmed cell death-ligand 1 (PD-L1) regimens, particularly bevacizumab and atezolizumab, as first-line treatments for advanced hepatocellular carcinoma (HCC), there is a need to explore PD-L1 and programmed cell death 1 inhibitors in combination therapies for unresectable HCC (uHCC). Integrating systemic therapies with locoregional approaches is also emerging as a potent strategy. This study compares the outcomes of atezolizumab (PD-L1 inhibitor) and sintilimab (programmed cell death 1 inhibitor) with bevacizumab or its biosimilar, combined with hepatic arterial interventional therapies (HAIT) in uHCC patients. From January 2020 to September 2023, a retrospective analysis was conducted on 138 uHCC patients at Sun Yat-sen University Cancer Center. The cohort included 69 patients treated with atezolizumab with bevacizumab (Bev/Ate) and 69 with bevacizumab biosimilar with sintilimab (Bio/Sin), combined with HAIT. The propensity score matching was also employed to further explore the efficacy and safety. The median progression-free survival (mPFS) was 13.8 months for the Bev/Ate group and 10.0 months for the Bio/Sin group (p = 0.188). The Bev/Ate group showed significantly longer intrahepatic mPFS (HR 0.381; 95% confidence interval 0.176-0.824; p = .018) and higher overall response rates compared with the Bio/Sin group (60.87% vs. 31.88%, p = .001; 69.57% vs. 49.28%, p = .024) based on Response Evaluation Criteria in Solid Tumors v1.1 and modified Response Evaluation Criteria in Solid Tumors criteria. Treatment-related adverse events were similar between groups (p > .050). Combining atezolizumab or sintilimab with bevacizumab or its biosimilar alongside HAIT provided similar overall PFS in uHCC patients. However, the atezolizumab-bevacizumab combination with HAIT showed superior intrahepatic PFS and control rates, warranting further validation.
Collapse
Affiliation(s)
- Minrui He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Wa Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Imaging Diagnostic and Interventional Center, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Ze Yuan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Neurosurgery/NeuroOncology, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Jinbin Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Juncheng Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Yizhen Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Zili Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Qi Meng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Wenqing Gao
- Department of Oncology, Tengchong People's Hospital, Baoshan, PR China
| | - Dandan Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Yangxun Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Zhongguo Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| |
Collapse
|
134
|
Slotman E, Pape M, van Laarhoven HWM, Pouw RE, van der Linden YM, Verhoeven RHA, Siesling S, Fransen HP, Raijmakers NJH. Considerations to forgo systemic treatment in patients with advanced esophageal or gastric cancer: A real-world evidence study. Int J Cancer 2025; 156:1950-1960. [PMID: 39786196 PMCID: PMC11924308 DOI: 10.1002/ijc.35314] [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: 07/17/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
The majority of patients with advanced esophageal or gastric cancer do not start palliative systemic treatment. To gain insight into the considerations underlying the decision not to start systemic treatment, we analyzed characteristics of patients starting and not starting systemic treatment, reasons for not starting systemic treatment, and receipt of local palliative treatments on a nationwide scale. Patients diagnosed with advanced esophageal or gastric cancer between 2015 and 2021 were included (n = 10,948). Survival was compared using propensity score matching on patient and disease characteristics. Most patients did not start systemic treatment (esophageal cancer 59%; gastric cancer 64%). These patients were generally older, more often female, had more comorbidities and a worse performance status. The main reason for not starting systemic treatment was patient or family preference (35%). Among patients who did not start systemic treatment, 47% (esophageal) and 19% (gastric), received local palliative treatment, most commonly radiotherapy. Patients who did not start systemic treatment had worse median overall survival compared to patients who did start (esophageal cancer 2.9 months vs. 8.9 months; gastric cancer 2.2 vs. 8.2 months). These findings indicate that patient condition and disease burden are important aspects in systemic treatment decisions. However, patient or family preference was the main reason for not starting systemic treatment, highlighting that their priorities also strongly influence the decision. Systemic treatment did show to be associated with improved overall survival in matched patients, and therefore adequately weighing treatment risks and benefits based on real world data against patient preferences is of utmost importance.
Collapse
Affiliation(s)
- Ellis Slotman
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
- Department of Health Technology and Services Research, University of Twente, Technical Medical Centre, Enschede, The Netherlands
| | - Marieke Pape
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
- Medical Oncology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Hanneke W M van Laarhoven
- Medical Oncology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Roos E Pouw
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| | - Yvette M van der Linden
- Centre of Expertise in Palliative Care, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Radiotherapy, Leiden University Medical Centre, Leiden, The Netherlands
| | - Rob H A Verhoeven
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
- Medical Oncology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Sabine Siesling
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
- Department of Health Technology and Services Research, University of Twente, Technical Medical Centre, Enschede, The Netherlands
| | - Heidi P Fransen
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Natasja J H Raijmakers
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| |
Collapse
|
135
|
Ragusa C, Pereira B, Balayssac D. Assessment of pain prevalence in cancer patients undergoing anticancer treatments and in cancer survivors after completion of anticancer treatments: A French nationwide cross-sectional study. Int J Cancer 2025; 156:1873-1884. [PMID: 39625069 DOI: 10.1002/ijc.35280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/21/2025]
Abstract
Pain is a common and disabling symptom of cancer and its treatment. This study aimed to provide an update on the prevalence, characteristics, and impact of pain on quality of life (QoL) in cancer patients and survivors in France. Data were collected using self-assessment questionnaires as part of a nationwide web-based survey conducted between January and March 2023. Pain was reported by 44.7% of the study population (n = 1029), including by 49.2% (95% CI [44.8; 53.6]) of cancer patients (n = 255/518) and 40.1% (95% CI [35.8; 44.5]) of cancer survivors (n = 205/511). Chronic pain was more prevalent among survivors (99.0%) than patients (87%), but no between-group differences in the prevalence of neuropathic pain (66.8% vs. 67.5%, respectively) or other pain characteristics (pain intensity, location, etc.) were observed. Pain had a negative impact on QoL in both groups, but the impact on global health status, functioning, symptom severity, and depression was greater among cancer patients. Analgesic use was also more frequent among patients than survivors. Breast cancer, being overweight or obese, and having a poorer global health status were identified as main factors increasing the likelihood of pain. Pain therefore remains a common symptom among cancer patients and survivors in France. Further improvements to management are needed, including strategies to target chronic and neuropathic pain, and the high frequency of pain associated with breast cancer. Multimodal interventions to improve global health status, help individuals maintain a healthy weight, and reduce the impact of cancer pain on QoL could also be evaluated.
Collapse
Affiliation(s)
- Charles Ragusa
- Université Clermont Auvergne, INSERM U1107, NEURO-DOL, CHU Clermont-Ferrand, Direction de la Recherche Clinique et de l'Innovation, Clermont-Ferrand, France
| | - Bruno Pereira
- CHU Clermont-Ferrand, Direction de la Recherche Clinique et de l'Innovation, Clermont-Ferrand, France
| | - David Balayssac
- Université Clermont Auvergne, INSERM U1107, NEURO-DOL, CHU Clermont-Ferrand, Direction de la Recherche Clinique et de l'Innovation, Clermont-Ferrand, France
| |
Collapse
|
136
|
Peng S, Zhu Y, Zhu J, Chen Z, Tao Y. Plasma-based untargeted metabolomics reveals potential biomarkers for screening and distinguishing of ovarian tumors. Clin Chim Acta 2025; 572:120246. [PMID: 40107594 DOI: 10.1016/j.cca.2025.120246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 03/05/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Ovarian cancer (OC), a leading cause of gynecological cancer mortality, is frequently detected at advanced stages due to asymptomatic early progression. This study investigates plasma-based untargeted metabolomics for identifying biomarkers to screen and differentiate ovarian tumors (OT). Plasma samples from OC, benign ovarian tumors (BOT), and healthy controls (HC) were analyzed. Samples were randomized into train and test sets, with differential metabolites screened via two-tailed Student's t-test and partial least squares discriminant analysis. ROC models evaluated discriminatory capacity. Key metabolites demonstrated high predictive value: TMAO and hippuric acid distinguished OT from HC (AUC > 0.95), while linoleic acid, alpha-linolenic acid, and arachidonic acid (AUC > 0.9) further supported OT screening. Kynurenine differentiated OC from BOT (AUC = 0.808). Reduced levels of specific lysophosphatidylcholines (LPC (17:0/0:0), LPC (15:0/0:0)) also distinguished OT from HC (AUC = 0.771-0.89). These findings suggest plasma metabolomics holds promise for noninvasive biomarker discovery in OT screening and distinguishing between malignant and benign cases, though further validation of metabolite quantification is warranted prior to clinical application.
Collapse
Affiliation(s)
- Shen Peng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yiming Zhu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Jing Zhu
- Department of Clinical Laboratory, Zhenjiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Zhongjian Chen
- Experimental Research Center, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China.
| | - Yi Tao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
137
|
Rodriguez MS, Mahmoud YD, Vanzulli S, Giulianelli S, Spengler E, Martínez Vazquez P, Burruchaga J, Bushweller J, Lamb CA, Lüthy IA, Lanari C, Pérez Piñero C. FGFR2-RUNX2 activation: An unexplored therapeutic pathway in luminal breast cancer related to tumor progression. Int J Cancer 2025; 156:2024-2038. [PMID: 39731522 DOI: 10.1002/ijc.35302] [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: 03/18/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 12/30/2024]
Abstract
Overcoming luminal breast cancer (BrCa) progression remains a critical challenge for improved overall patient survival. RUNX2 has emerged as a protein related to aggressiveness in triple-negative BrCa, however its role in luminal tumors remains elusive. We have previously shown that active FGFR2 (FGFR2-CA) contributes to increased tumor growth and that RUNX2 expression was high in hormone-independent mouse mammary carcinomas. To elucidate the interaction between FGFR2 and RUNX2 in human BrCa, we investigated their roles in tumor progression and treatment responsiveness. Increased FGFR2 activity resulted in higher RUNX2 expression, cell proliferation, and metastasis. In contrast, silencing FGFR2 reduced these parameters. Overexpression of RUNX2 in FGFR2-silenced cells rescued the inhibitory effects, promoting a more aggressive phenotype, even if compared with the wt RUNX2-transfected cells, which also had increased aggressiveness compared with naïve-transfected cells. RUNX2-overexpressing tumors were insensitive to endocrine- or FGFR inhibitor treatments. Notably, the CBFβ-RUNX complex inhibitor, AI-14-91, demonstrated great effectiveness in vitro. In a small cohort of luminal BrCa patients, nuclear RUNX2 expression was associated with tumor recurrence. Transcriptomic analysis strongly supported these data showing that patients with luminal carcinomas with high RUNX2 activity score have a worse progression-free interval than those with low RUNX2 activity. Our findings suggest a complex interplay between FGFR2 and RUNX2 in regulating tumor aggressiveness. This study underscores the significance of RUNX2 in luminal BrCa progression and posits RUNX2 as a promising therapeutic target and as a potential prognostic biomarker in luminal BrCa patients.
Collapse
Affiliation(s)
- María S Rodriguez
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Yamil D Mahmoud
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires, Argentina
| | - Silvia Vanzulli
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Sebastián Giulianelli
- Instituto de Biología de Organismos Marinos (IBIOMAR-CCT), CENPAT-CONICET, Puerto Madryn, Argentina
| | - Eunice Spengler
- Hospital Zonal Gral. de Agudos "Magdalena V. de Martínez", General Pacheco, Argentina
| | | | - Javier Burruchaga
- Hospital Zonal Gral. de Agudos "Magdalena V. de Martínez", General Pacheco, Argentina
| | - John Bushweller
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Caroline A Lamb
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Isabel A Lüthy
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Claudia Lanari
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Cecilia Pérez Piñero
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| |
Collapse
|
138
|
Wang F, Yin L, Hu Y. Progress of extracellular vesicles-based system for tumor therapy. J Control Release 2025; 381:113570. [PMID: 39993635 DOI: 10.1016/j.jconrel.2025.02.066] [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: 08/29/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
The increasing number of new cancer cases and cancer-related deaths worldwide highlights the urgent need to develop novel anti-tumor treatment methods to alleviate the current challenging situation. Nearly all organisms are capable of secreting extracellular vesicles (EVs), and these nano-scale EVs carrying biological molecules play an important role in intercellular communication, further affecting various physiological and pathological processes. Notably, EVs from different sources have differences in their characteristics and functions. Consequently, diverse EVs have been utilized as drug or vaccine delivery carriers for improving anti-tumor treatment due to their good safety, ease of modification and unique properties, and achieved satisfactory results. Meanwhile, the clinical trials of EV-based platform for tumor therapy are also continuously being conducted. Therefore, in this review, we summarize the recent research progress of EV-based tumor treatment methods, including the introduction of main sources and unique functions of EVs, the application of EVs in tumor treatment as well as their prospects and challenges. Additionally, considering the unique advantages of artificial EVs over natural EVs, we also highlighted their characteristics and applications in tumor treatments. We believe that this review will help researchers develop novel EV-based anti-tumor platforms through a bottom-up design and accelerate the development in this field.
Collapse
Affiliation(s)
- Fei Wang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; Nanjing University (Suzhou) High-tech Institute, Renai Road 150, Suzhou Industrial Park, Suzhou 215123, China
| | - Le Yin
- Affiliated Tongzhou Hospital of Xinglin College, Nantong University, 999 Jianshe Road, Jinsha Town, Tongzhou District, Nantong, Jiangsu 226300, China.
| | - Yong Hu
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; Nanjing University (Suzhou) High-tech Institute, Renai Road 150, Suzhou Industrial Park, Suzhou 215123, China.
| |
Collapse
|
139
|
Liang C, Ma Y, Ding M, Gao F, Yu K, Wang S, Qu Y, Hua H, Li D. Asiatic acid and its derivatives: Pharmacological insights and applications. Eur J Med Chem 2025; 289:117429. [PMID: 40015163 DOI: 10.1016/j.ejmech.2025.117429] [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: 01/17/2025] [Revised: 02/07/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Centella asiatica (L.) Urban has been utilized in wound healing remedies for nearly 3000 years. Asiatic acid (AA), a pentacyclic triterpenoid characterized by ursane-type skeleton, serves as principal bioactive constituent of Centella asiatica, exhibits remarkable therapeutic potential across a spectrum of health conditions. Pharmacological investigations have revealed that AA exerts direct regulatory effects on a multitude of enzymes, receptors, inflammatory mediators, and transcription factors. This article systematically examines the therapeutic applications of AA and its derivatives in the management of neurodegenerative diseases, cancer, cardiovascular disorders, and infections. Additionally, recent advancements in the structural modification of AA are summarized, offering new insights for the development of low-toxicity, effective AA-based therapeutics and diagnostic agents. However, several challenges remain, including the paucity of clinical trials, uncertainties in dosage and treatment regimens, limited data on long-term safety and side effects, and poor bioavailability. Addressing these limitations is crucial for advancing AA-based therapies and ensuring their clinical applicability.
Collapse
Affiliation(s)
- Chaowei Liang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Yongzhi Ma
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Minni Ding
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Fang Gao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Kewang Yu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Siyu Wang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Ying Qu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China.
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
140
|
Shukla S, Bagchi D, Divya, Khushi, Manohara Reddy YV, Park JP. Multifunctional metal-organic frameworks in breast cancer therapy: Advanced nanovehicles for effective treatment. Eur J Med Chem 2025; 289:117424. [PMID: 39999692 DOI: 10.1016/j.ejmech.2025.117424] [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: 01/12/2025] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025]
Abstract
Breast cancer is the second-most common cause of cancer-related death among women worldwide, with a gradual annual increase of 0.5 % in its occurrence rate in recent years. This complex ailment exhibits considerable diversity, with a mortality rate of 2.5 %. One promising area of research for its treatment is the development of MOFs, which are intricate three-dimensional (3D) structures constructed from metal ions or clusters joined with organic ligands through coordinate bonds. MOFs have emerged as versatile platform overcoming the limitations of conventional chemotherapeutics including poor drug solubility, non-specific targeting, and multidrug resistance. These applications are attributed to their adjustable porosity, chemical makeup, dimensions, straightforward surface customization capabilities, biocompatibility, nontoxicity etc. These properties position MOFs as excellent candidates for diverse regimes of cancer therapeutics including innovative approaches such as phototherapy, chemotherapy, immunotherapy, gene therapy, sonodynamic therapy, and various combination therapies. The article emphasizes the functionalization and applications of MOFs, with a primary focus on their therapeutic capabilities, synergistic approaches, and theranostic strategies that integrate diagnostic and therapeutic functions. Strategies to improve MOF biocompatibility and stability, such as surface modifications and biocompatible coatings are also discussed. Insights on various challenges and future prospects are provided to address current limitations and inspire further research, paving the way for clinical translation of MOF-based breast cancer therapies.
Collapse
Affiliation(s)
- Shefali Shukla
- Department of Chemistry, Sri Venkateswara College, University of Delhi, New Delhi, India.
| | - Dipankar Bagchi
- Department of Chemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Divya
- Department of Chemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Khushi
- Department of Chemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Y Veera Manohara Reddy
- Department of Chemistry, Sri Venkateswara College, University of Delhi, New Delhi, India.
| | - Jong Pil Park
- Department of Food Science and Technology, GreenTech-based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|
141
|
Zhao G, Li C, Liu W, Wu J, Yang X. Understanding the Molecular Mechanisms of SORBS2 in TNBC Lung Metastasis. Biochem Biophys Res Commun 2025; 762:151762. [PMID: 40199126 DOI: 10.1016/j.bbrc.2025.151762] [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: 01/21/2025] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/10/2025]
Abstract
Metastasis is the leading cause of recurrence and mortality in triple-negative breast cancer (TNBC), an aggressive subtype that predominantly spreads to the lungs, brain, bones, and liver, with lung metastasis being particularly prevalent. Despite the clinical significance of TNBC metastasis, the molecular mechanisms that drive lung-specific metastasis remain poorly understood. RNA-binding proteins (RBPs) are crucial regulators of post-transcriptional gene expression and are frequently dysregulated in cancers. This study identifies SORBS2 as a critical RBP implicated in TNBC lung metastasis. Using RNA sequencing (RNA-seq) and LACE-seq, we demonstrate that SORBS2 regulates a specific set of genes through direct binding to coding sequences (CDS), introns, and 3' untranslated regions (UTRs), and its binding targets are linked to various pathways, including a possible association with Wnt/β-catenin signaling, among others. Functional assays confirm that SORBS2 knockdown inhibits proliferation, migration, and invasion in TNBC cells. These findings highlight SORBS2 as a key regulator of TNBC lung metastasis, with a context-dependent role that promotes metastatic behavior in highly metastatic TNBC cells, providing potential avenues for novel therapeutic strategies.
Collapse
Affiliation(s)
- Gongke Zhao
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China
| | - Chunzheng Li
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China
| | - Wan Liu
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China
| | - Jianing Wu
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China
| | - Xianguang Yang
- State Key Laboratory Base of Cell Differentiation and Regulation, Henan Normal University, Xin xiang, China.
| |
Collapse
|
142
|
Ashi A, Awaji AA, Bond J, Johnson CA, Shaaban AM, Bell SM. Threonine and tyrosine kinase (TTK) mRNA and protein expression in breast cancer; prognostic significance in the neoadjuvant setting. Histopathology 2025; 86:916-932. [PMID: 39775836 PMCID: PMC11964583 DOI: 10.1111/his.15399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
AIMS Threonine and tyrosine kinase (TTK) is up-regulated in triple-negative breast cancer (TNBC), yet its expression in patients undergoing neoadjuvant chemotherapy (NACT) remains unexplored. This investigation aims to assess TTK protein expression in treatment-naïve pre-treatment cores and paired pre- and post-NACT breast cancer (BC) cohorts, as well as its correlation with microcephaly 1 (MCPH1) protein expression. METHODS AND RESULTS Transcriptomic data were sourced from the Gene Expression Omnibus microarray database for mRNA expression analysis. TTK protein expression was evaluated using immunohistochemistry staining, employing receiver operating characteristic curve analysis to determine an optimal TTK expression cut-off point. The association between TTK expression, clinicopathological parameters and survival outcomes was examined. Additionally, MCPH1 protein expression was assessed in a pilot study. Analysis revealed a significantly elevated TTK mRNA expression in BC tissue compared to normal breast tissue, with high TTK mRNA levels predicting reduced overall survival. Notably, TTK protein expression increased significantly post-NACT in a paired cohort. Conversely, decreased TTK protein expression pre-NACT was correlated with improved overall survival. CONCLUSIONS High TTK and low MCPH1 protein expression was significantly correlated, highlighting TTK's potential as a biomarker for BC and a therapeutic target for MCPH1-deficient cancer cells.
Collapse
Affiliation(s)
- Abrar Ashi
- Division of Molecular Medicine, Leeds Institute of Medical Research, St James's University HospitalUniversity of LeedsLeedsUK
| | - Aeshah A Awaji
- Division of Molecular Medicine, Leeds Institute of Medical Research, St James's University HospitalUniversity of LeedsLeedsUK
| | - Jacquelyn Bond
- Division of Molecular Medicine, Leeds Institute of Medical Research, St James's University HospitalUniversity of LeedsLeedsUK
| | - Colin A Johnson
- Division of Molecular Medicine, Leeds Institute of Medical Research, St James's University HospitalUniversity of LeedsLeedsUK
| | - Abeer M Shaaban
- Histopathology, St James's Institute for OncologySt James's University HospitalLeedsUK
- Histopathology and Cancer SciencesQueen Elizabeth Hospital Birmingham and University of BirminghamBirminghamUK
| | - Sandra M Bell
- Division of Molecular Medicine, Leeds Institute of Medical Research, St James's University HospitalUniversity of LeedsLeedsUK
| |
Collapse
|
143
|
Proesmans K, Luik AI, Lahousse L. Sex-specific associations between sleep apnoea and lung cancer risk in patients with COPD: a nationwide prospective cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2025; 52:101269. [PMID: 40224371 PMCID: PMC11987683 DOI: 10.1016/j.lanepe.2025.101269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 04/15/2025]
Abstract
Background COPD is an established risk factor for lung cancer. Sleep apnoea is prevalent in COPD and the inflammation caused by intermittent hypoxaemia may increase this lung cancer risk. Females have more systemic inflammation for a similar apnoea-hypopnoea index than males. Therefore, this study aims to investigate sex-specific associations between sleep apnoea and lung cancer in COPD. Methods The sex-specific absolute and relative risk of sleep apnoea on newly diagnosed lung cancer was estimated in a nationwide observational study of Belgian patients with COPD (≥55 years), between 2017 and 2022, using an Aalan-Johanson estimator and a cause-specific Cox regression model adjusted for age, socioeconomic status, smoking status, alcoholism, frailty, comorbidities, and comedication. Findings The study consisted of 62,903 COPD patients (42·80% female), of whom 2898 (4·60%) developed lung cancer. We found a significant sex interaction of sleep apnoea on lung cancer hazard ( χ -squared: 13·239, P-interaction < 0·01). In females, sleep apnoea was associated with a higher lung cancer risk (cumulative incidence: 1545 vs 1350 per 100,000 PY; aHR: 1·31 (95% CI: 1·05-1·63)). For males, sleep apnoea patients had a lower lung cancer risk (cumulative incidence: 1632 and 2305 per 100,000 PY; aHR: 0·82 (95% CI: 0·70-0·95)). The impact of sleep apnoea on lung cancer development was especially strong in female COPD patients with hypoxia-related comorbidities e.g., with a history of emphysema (aHR: 2·65 (95% CI: 1·11-6·34)). Interpretation Sleep apnoea was associated with a higher risk of lung cancer in female COPD patients while, in males, there was a lower risk. Especially in female COPD patients with hypoxia, sleep apnoea is strongly associated with an increased lung cancer risk. Funding Emmanuel van der Schueren cancer research fellowship "Kom Op Tegen Kanker".
Collapse
Affiliation(s)
- Kristiaan Proesmans
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent, 9000, Belgium
| | - Annemarie I. Luik
- Department of Epidemiology, Erasmus Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015, the Netherlands
- Trimbos Institute, The Netherlands Institute of Mental Health and Addiction, Utrecht, the Netherlands
| | - Lies Lahousse
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, Ghent, 9000, Belgium
- Department of Epidemiology, Erasmus Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015, the Netherlands
| |
Collapse
|
144
|
Wang Y, Huang M, Zhou X, Li H, Ma X, Sun C. Potential of natural flavonoids to target breast cancer angiogenesis (review). Br J Pharmacol 2025; 182:2235-2258. [PMID: 37940117 DOI: 10.1111/bph.16275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023] Open
Abstract
Angiogenesis is the process by which new blood vessels form and is required for tumour growth and metastasis. It helps in supplying oxygen and nutrients to tumour cells and plays a crucial role in the local progression and distant metastasis of, and development of treatment resistance in, breast cancer. Tumour angiogenesis is currently regarded as a critical therapeutic target; however, anti-angiogenic therapy for breast cancer fails to produce satisfactory results, owing to issues such as inconsistent efficacy and significant adverse reactions. As a result, new anti-angiogenic drugs are urgently needed. Flavonoids, a class of natural compounds found in many foods, are inexpensive, widely available, and exhibit a broad range of biological activities, low toxicity, and favourable safety profiles. Several studies find that various flavonoids inhibit angiogenesis in breast cancer, indicating great therapeutic potential. In this review, we summarize the role of angiogenesis in breast cancer and the potential of natural flavonoids as anti-angiogenic agents for breast cancer treatment. We discuss the value and significance of nanotechnology for improving flavonoid absorption and utilization and anti-angiogenic effects, as well as the challenges of using natural flavonoids as drugs. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Yuetong Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengge Huang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xintong Zhou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Xiaoran Ma
- Department of Oncology, Linyi People's Hospital, Linyi, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
145
|
Holthuijsen DDB, Rijnhart JJM, Bours MJL, van Roekel EH, Ueland PM, Breukink SO, Janssen-Heijnen MLG, Konsten JL, Keulen ETP, McCann A, Brezina S, Gigic B, Ulrich CM, Weijenberg MP, Eussen SJPM. Longitudinal associations of dietary intake with fatigue in colorectal cancer survivors up to 1 year post-treatment, and the potential mediating role of the kynurenine pathway. Brain Behav Immun 2025; 126:144-159. [PMID: 39922470 DOI: 10.1016/j.bbi.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/22/2025] [Accepted: 02/01/2025] [Indexed: 02/10/2025] Open
Abstract
INTRODUCTION A healthy diet may help to reduce cancer-related fatigue, but evidence is limited and mechanisms remain unclear. Both diet and fatigue following colorectal cancer (CRC) have been linked to metabolites (kynurenines) of the kynurenine pathway (KP). We investigated longitudinal associations between dietary intake and fatigue, and the potential mediating role of the KP, in CRC survivors up to 1 year post-treatment. METHODS Measurements at 6 weeks, 6 months, and 1 year post-treatment were performed in 209 stage I-III CRC survivors. Diet was assessed by 7-day food records. Plasma kynurenines were analyzed using LC-MS/MS. Fatigue, including subjective fatigue, was assessed using validated questionnaires. To analyse longitudinal associations between diet and fatigue and to explore potential mediation by the KP, we used confounder-adjusted multilevel parallel-multiple mediator models with all kynurenines included simultaneously, and simple mediator models with established KP ratios to estimate total (c: diet-fatigue), direct (c': diet-fatigue, while controlling for mediators), metabolite-specific indirect (ab: diet-metabolite-fatigue), and total indirect (ab: diet-metabolites-fatigue) effects. RESULTS Higher intake of total carbohydrates and mono- and disaccharides was longitudinally associated with more subjective fatigue, while higher intake of plant protein, total fat, and unsaturated fats was associated with less subjective fatigue (c). Most associations remained statistically significant after controlling for KP metabolites, except for mono- and disaccharides (c'). All kynurenines simultaneously did not mediate longitudinal associations between diet and subjective fatigue (ab). The kynurenic acid-to-quinolinic acid (KA/QA) ratio significantly mediated associations of intakes of carbohydrate, mono- and disaccharides, alcohol, magnesium, and zinc with subjective fatigue, whereas the HKr significantly mediated the association between polysaccharide intake and subjective fatigue (ab). CONCLUSION Our findings suggest that carbohydrate intake is associated with greater fatigue, while protein and fat intake are associated with lower fatigue in CRC survivors up to 1 year post-treatment. While all KP metabolites simultaneously did not significantly mediate associations between diet and fatigue in our population, the KA/QA ratio and HKr were significant mediators in several diet-fatigue associations. These results should be repeated in larger observational studies.
Collapse
Affiliation(s)
- Daniëlle D B Holthuijsen
- Department of Epidemiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands.
| | | | - Martijn J L Bours
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Eline H van Roekel
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | | | - Stéphanie O Breukink
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Centre+, Maastricht, the Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Maryska L G Janssen-Heijnen
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Clinical Epidemiology, VieCuri Medical Centre, Venlo, the Netherlands
| | - Joop L Konsten
- Department of Surgery, VieCuri Medical Centre, Venlo, the Netherlands
| | - Eric T P Keulen
- Department of Internal Medicine and Gastroenterology, Zuyderland Medical Centre Sittard-Geleen, Geleen, the Netherlands
| | | | - Stefanie Brezina
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Biljana Gigic
- Department of General Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Cornelia M Ulrich
- Huntsman Cancer Institute and Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Matty P Weijenberg
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
146
|
Saif A, Islam MT, Raihan MO, Yousefi N, Rahman MA, Faridi H, Hasan AR, Hossain MM, Saleem RM, Albadrani GM, Al-Ghadi MQ, Ahasan Setu MA, Kamel M, Abdel-Daim MM, Aktaruzzaman M. Pan-cancer analysis of CDC7 in human tumors: Integrative multi-omics insights and discovery of novel marine-based inhibitors through machine learning and computational approaches. Comput Biol Med 2025; 190:110044. [PMID: 40120182 DOI: 10.1016/j.compbiomed.2025.110044] [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: 08/28/2024] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Cancer remains a significant global health challenge, with the Cell Division Cycle 7 (CDC7) protein emerging as a potential therapeutic target due to its critical role in tumor proliferation, survival, and resistance. However, a comprehensive analysis of CDC7 across multiple cancers is lacking, and existing therapeutic options have come with limited clinical success. The aim of this is to integrate a comprehensive pan-cancer analysis of CDC7 with the identification of novel marine-derived inhibitors, bridging the understanding of CDC7's role as a prognostic biomarker and therapeutic target across diverse cancer types. In this study, we conducted a pan-cancer analysis of CDC7 across 33 tumor types using publicly available datasets to evaluate its expression, genetic alterations, immune interactions, survival, and prognostic significance. Additionally, a marine-derived compound library of 31,492 molecules was screened to identify potential CDC7 inhibitors using chemoinformatics and machine learning. The top candidates underwent rigorous evaluations, including molecular docking, pharmacokinetics, toxicity, Density Functional Theory (DFT) calculations, and Molecular Dynamics (MD) simulations. The findings revealed that CDC7 is overexpressed in several cancers and is associated with poor survival outcomes and unfavorable prognosis. Enrichment analysis linked CDC7 to critical DNA replication pathways, while its role in modulating tumor-immune interactions highlighted its potential as a target for immunotherapy. Among all tested compounds, Tetrahydroaltersolanol D (CMNPD21999) exhibited the strongest binding affinity and stability, along with better drug-likeness and zero toxicity. These attributes highlight its potential as a promising drug candidate for CDC7 inhibition and future cancer treatment development. Furthermore, additional in vitro and in vivo studies are required to confirm the effectiveness of this drug candidate against the CDC7 protein.
Collapse
Affiliation(s)
- Ahmed Saif
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh; Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh.
| | - Md Tarikul Islam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh; Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh.
| | - Md Obayed Raihan
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmaceutical Sciences, College of Health Sciences and Pharmacy, Chicago State University, Chicago, IL, USA.
| | - Niloofar Yousefi
- Department of Industrial Engineering and Management Systems, University of Central Florida, USA, Orlando, FL, USA
| | - Md Ajijur Rahman
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Hafeez Faridi
- Department of Pharmaceutical Sciences, College of Health Sciences and Pharmacy, Chicago State University, Chicago, IL, USA
| | - Al Riyad Hasan
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mirza Mahfuj Hossain
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Computer Science and Engineering, Faculty of Engineering and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Rasha Mohammed Saleem
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, 65431, Saudi Arabia
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 84428, Riyadh, 11671, Saudi Arabia
| | - Muath Q Al-Ghadi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Md Ali Ahasan Setu
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Microbiology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Md Aktaruzzaman
- Laboratory of Advanced Computational Biology, Biological Research on the Brain (BRB), Jashore, 7408, Bangladesh; Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.
| |
Collapse
|
147
|
Paudice M, Greppi M, Valle L, Piol N, Barra F, Mammoliti S, Ferrero S, Marcenaro E, Vellone VG. The role of the Androgen Receptor (AR) in endometrial cancer aggressiveness: Correlation with other prognostic markers and therapeutic implications. A retrospective observational study. Pathol Res Pract 2025; 269:155922. [PMID: 40186889 DOI: 10.1016/j.prp.2025.155922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
Endometrial carcinoma (EC) is the most common gynecological malignancy, with increasing incidence linked to rising risk factors. This retrospective observational study investigates the role of the Androgen Receptor (AR) in EC aggressiveness, its correlation with other prognostic markers, and its potential therapeutic implications. A total of 143 cases of EC treated with hysterectomy were analyzed for AR expression and its association with clinicopathological and molecular markers, including estrogen receptor (ER), progesterone receptor (PR), Ki-67, p53, β-catenin, E-cadherin, Bcl-2, Cyclin D1, and mismatch repair (MMR) status. AR expression was significantly higher in low-grade endometrioid carcinoma (LGEC) compared to high-grade endometrioid carcinoma (HGEC) and other high-risk histologies (p = 0.015), suggesting a role in less aggressive tumor phenotypes. AR strongly correlated with ER and PR (p < 0.0001), indicating shared regulatory pathways. A borderline association with tumor-infiltrating lymphocytes (TILs) suggests a potential role in immune response. However, AR expression did not significantly correlate with markers of proliferation (Ki-67) or tumor suppression (p53), nor with β-catenin, E-cadherin, Bcl-2, Cyclin D1, or MMR status. These findings support AR as a prognostic marker in hormone-responsive EC subtypes and suggest that AR-targeted therapies could be beneficial, particularly in ER/PR-negative tumors. The study highlights the potential integration of AR status into molecular profiling, aiding in personalized treatment strategies for improved patient outcomes in EC management.
Collapse
Affiliation(s)
- Michele Paudice
- Department of Integrated Diagnostic and Surgical Sciences (DISC), University of Genoa, Genoa 16132, Italy; Pathology University Unit, IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa 16132, Italy
| | - Luca Valle
- Pathology University Unit, IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Nataniele Piol
- Pathology University Unit, IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Fabio Barra
- Department of Health Sciences (DISSAL), University of Genoa, 16132 Genoa, Italy Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Serafina Mammoliti
- Obstetrics & Gynecology University Unit, IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Simone Ferrero
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa 16132, Italy; IRCCS Ospedale Policlinico San Martino, Genova 16132, Italy
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa 16132, Italy; IRCCS Ospedale Policlinico San Martino, Genova 16132, Italy.
| | - Valerio Gaetano Vellone
- Department of Integrated Diagnostic and Surgical Sciences (DISC), University of Genoa, Genoa 16132, Italy; Pathology Unit, IRCCS Istituto Giannina Gaslini, Genoa 16147, Italy.
| |
Collapse
|
148
|
Jian Y, Yue P, Qiao H. 28-Day Repeated Dose Toxicity and Toxicokinetics Study on Dihydroartemisinin (DHA) in SD Rats. J Appl Toxicol 2025; 45:755-766. [PMID: 39710504 DOI: 10.1002/jat.4738] [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] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024]
Abstract
Dihydroartemisinin (DHA) is an effective antimalarial drug with potential antitumor efficacy, yet toxicological information is limited. The present study was designed to evaluate the potential toxicity of oral DHA. DHA was administered orally by gavage to SD rats at doses of 0, 25, 50, and 75/60 mg/kg b.w./day for 28 days, followed by a 4-week recovery period. Concomitant toxicokinetics was also evaluated. Due to potential toxicity affecting survival, only the female top dose was adjusted from 75 to 60 mg/kg on study day 14 (D14). Female rats in the low-dose group and male rats in the low- and medium-dose groups did not show any signs of toxicity. In contrast, male rats in the high-dose group and female rats in the medium- and high-dose groups showed significant toxic effects, including weight loss, hair loss, and gastrointestinal reactions (soft stools, perianal dirt, and fecal abnormalities). At the end of administration, female rats in the 75/60 (dose-adjusted) mg/kg dose group had significantly higher reticulocytes (Ret% and RETIC) and alanine aminotransferase (ALT), increased liver weights, and significantly lower hemoglobin (HGB). In addition, histopathology showed mild vacuolation of hepatocytes. These findings suggest that female rats have a greater toxic response than males, and toxicokinetics further demonstrate this sex difference. However, the toxic effects of DHA were reversed at the end of the 4-week recovery period. Therefore, the liver was identified as the primary target organ. The no-observed-adverse-effect-level (NOAEL) was 25 and 50 mg/kg b.w./day in female and male rats, respectively.
Collapse
Affiliation(s)
- Yang Jian
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | - Peng Yue
- Jiangsu Center for Safety Evaluation of Drugs, Jiangsu Provincial Institute of Materia Medica, Nanjing, China
| | - Hongqun Qiao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| |
Collapse
|
149
|
Wong P, Tran TB, Pollini T, Hernandez S, Zampese M, Todeschini L, Aguilar LL, Maker VK, Maker AV. Impact of coronary artery stenting on perioperative mortality and complications in patients undergoing pancreaticoduodenectomy. J Gastrointest Surg 2025; 29:102020. [PMID: 40118202 DOI: 10.1016/j.gassur.2025.102020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Postoperative morbidity and mortality rates from pancreaticoduodenectomy (PD) have significantly decreased, allowing for greater consideration of patients with severe comorbidities. This study aimed to evaluate the effect of previous coronary artery intervention on morbidity and mortality among patients who underwent PD. METHODS Patients who underwent PD were identified from the American College of Surgeon National Surgical Quality Improvement Program database. Patients with previous coronary artery intervention received either balloon dilatation or stent placement. The main outcome measures included in-hospital mortality and postoperative myocardial infarction (MI). RESULTS Of 10,848 patients who underwent PD, 698 (6.4%) received previous coronary artery intervention. Compared with patients without coronary artery intervention, those with previous coronary artery intervention were older (65 vs 70 years, respectively; P <.001), were less likely to be female (50.2% vs 26.4%, respectively; P <.001), and had higher median body mass index (26 vs 27 kg/m2, respectively; P =.003). Compared with patients not in the angioplasty/stent cohort, those in the angioplasty/stent cohort were more likely to have diabetes mellitus (22.0% vs 39.3%, respectively), functional impairment (2.4% vs 4.9%, respectively), chronic obstructive pulmonary disease (4.1% vs 8.2%, respectively), hypertension (51.2% vs 86.2%, respectively), and bleeding disorders (2.2% vs 8.0%, respectively) (all P <.001). Compared with patients not in the angioplasty/stent cohort, those in the stent/angioplasty group were more likely to have postoperative complications (41.0% vs 51.4%, respectively; P <.001). Previous stent/angioplasty procedure (odds ratio [OR], 2.61 [95% CI, 1.42-4.57]; P =.001) was associated with developing postoperative MI but was not an independent predictor of in-hospital mortality (OR, 1.19 [95% CI, 0.81-1.70]; P =.369). CONCLUSION Previous stent placement/angioplasty was not associated with increased in-hospital mortality in patients who underwent PD, despite being correlated with an increased risk of MI and severe complications. Previous coronary artery angioplasty and/or stenting is not an absolute contraindication for PD, but patients should be medically optimized preoperatively to mitigate the risk of major adverse cardiac events.
Collapse
Affiliation(s)
- Paul Wong
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Thuy B Tran
- Division of Surgical Oncology, Department of Surgery, University of Illinois Chicago, Chicago, IL, United States
| | - Tommaso Pollini
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Sophia Hernandez
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Marco Zampese
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Letizia Todeschini
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Luis Laurean Aguilar
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Vijay K Maker
- Division of Surgical Oncology, Department of Surgery, University of Illinois Chicago, Chicago, IL, United States
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
150
|
Zhang X, Han X. Synchronous occurrence of primary gastric and liver cancer: A case report. Oncol Lett 2025; 29:222. [PMID: 40103599 PMCID: PMC11916645 DOI: 10.3892/ol.2025.14968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/02/2024] [Indexed: 03/20/2025] Open
Abstract
Primary gastric and liver cancers rank among the most prevalent malignant tumors of the digestive tract. Despite their serious implications for health, the global age-standardized incidence remains relatively low, at ~11.1 per 100,000 for primary gastric cancer and ~8.657 per 100,000 for primary liver cancer. Although the occurrence of multiple primary malignancies is not uncommon in clinical practice, reports of synchronous primary gastric and liver cancer are exceedingly rare. The present study describes a case involving a 60-year-old man diagnosed with synchronous primary gastric and liver cancer. The patient underwent endoscopic submucosal dissection for lesions located at the gastric angle, followed by laparoscopic resection of a small liver tumor. Pathological examinations revealed moderately differentiated intramucosal adenocarcinoma at the gastric angle and well-differentiated hepatocellular carcinoma in the liver. Following a 3-year follow-up, the patient remained in good health, with no evidence of disease recurrence. In conclusion, clinicians should exercise caution in patients presenting with distinct lesions to ensure that subtle malignancies are not overlooked, particularly in those with confirmed cancer. For patients with multiple cancers, it is crucial to ascertain whether the malignancies are primary, as this determination influences treatment strategies.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Gastroenterology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441021, P.R. China
| | - Xiaoying Han
- Department of Gastroenterology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441021, P.R. China
| |
Collapse
|