1
|
Wu T, Zhang L, Tian J, Lu Y, Yang Y, Yang X, Jin P, Gu Y, Wang X, Yi L. Correlation between dyslipidaemia and gastric cancer: pathogenesis to prevention and treatment strategies. Lipids Health Dis 2025; 24:204. [PMID: 40483484 PMCID: PMC12144744 DOI: 10.1186/s12944-025-02625-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
As the sixth most common cancer in the world, the development of gastric cancer (GC) is influenced by many factors. Recent studies have revealed a strong association between dyslipidaemia and GC. In this paper, the relationships between dyslipidaemia and GC are discussed in depth. We review the development of GC through the mechanisms of the inflammatory response, signalling pathways, and apolipoprotein function, and the intersecting targets of atherosclerosis and GC are explored. The synergistic effects of Helicobacter pylori infection and dyslipidaemia on the development of GC are also analysed, and the potential value of statins in the prevention and treatment of GC is discussed. In this review, we systematically investigated the relationship between dyslipidaemia and GC to provide new ideas for GC prevention and treatment.
Collapse
Affiliation(s)
- Tao Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Lingna Zhang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China
| | - Jiao Tian
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China
| | - Yuyuan Lu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China
| | - Yan Yang
- Pharmacy Department, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Xin Yang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China
| | - Penghui Jin
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China
| | - Yuanhui Gu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Xiaoke Wang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Lin Yi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
2
|
Wang Y, Tang C, Wang K, Zhang X, Zhang L, Xiao X, Lin H, Xiong L. The role of ferroptosis in breast cancer: Tumor progression, immune microenvironment interactions and therapeutic interventions. Eur J Pharmacol 2025; 996:177561. [PMID: 40154567 DOI: 10.1016/j.ejphar.2025.177561] [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: 12/29/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Ferroptosis represents a distinctive and distinct form of regulated cellular death, which is driven by the accumulation of lipid peroxidation. It is distinguished by altered redox lipid metabolism and is linked to a spectrum of cellular activities, including cancer. In breast cancer (BC), with triple negative breast cancer (TNBC) being an iron-and lipid-rich tumor, inducing ferroptosis was thought to be a novel approach to killing breast tumor cells. However, in the recent past, a novel conceptual framework has emerged which posits that in addition to the promotion of tumor cell death, ferritin deposition has a potent immunosuppressive effect on the tumor immune microenvironment (TIME) via the influence on both innate and adaptive immune responses. TIME of BC includes various cell populations from both the innate and adaptive immune systems. In this review, the internal association between iron homeostasis and the progression of ferroptosis, along with the common inducers and protectors of ferroptosis in BC, are discussed in detail. Furthermore, a comprehensive analysis is conducted on the dual role of ferroptosis in immune cells and proto-oncogenic functions, along with an evaluation of the potential applications of immunogenic cell death-targeted immunotherapy in TIME of BC. It is anticipated that our review will inform future research endeavors that seek to integrate ferroptosis and immunotherapy in the management of BC.
Collapse
Affiliation(s)
- Yi Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chuanyun Tang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Keqin Wang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaoan Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lifang Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xinghua Xiao
- Department of Pathology, The First Affiliated Hospital, Nanchang University, 17 Yongwaizheng Road, Nanschang, 330066, China
| | - Hui Lin
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lixia Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
3
|
Delmas D, Mialhe A, Cotte AK, Connat JL, Bouyer F, Hermetet F, Aires V. Lipid metabolism in cancer: Exploring phospholipids as potential biomarkers. Biomed Pharmacother 2025; 187:118095. [PMID: 40311223 DOI: 10.1016/j.biopha.2025.118095] [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: 02/12/2025] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Aberrant lipid metabolism is increasingly recognized as a hallmark of cancer, contributing to tumor growth, metastatic dissemination, and resistance to therapy. Cancer cells reprogram key metabolic pathways-including de novo lipogenesis, lipid uptake, and phospholipid remodeling-to sustain malignant progression and adapt to microenvironmental demands. This review summarizes current insights into the role of lipid metabolic reprogramming in oncogenesis and highlights recent advances in lipidomics that have revealed cancer type- and stage-specific lipid signatures with diagnostic and prognostic relevance. We emphasize the dual potential of lipid metabolic pathways-particularly those involving phospholipids-as sources of clinically relevant biomarkers and therapeutic targets. Enzymes and transporters involved in these pathways have emerged as promising candidates for both diagnostic applications and pharmacological intervention. We also examine persistent challenges hindering the clinical translation of lipid-based approaches, including analytical variability, insufficient biological validation, and the lack of standardized integration into clinical workflows. Furthermore, the review explores strategies to overcome these barriers, highlighting the importance of incorporating lipidomics into multi-omics frameworks, supported by advanced computational tools and AI-driven analytics, to decipher the complexity of tumor-associated metabolic networks. We discuss how such integrative approaches can facilitate the identification of actionable metabolic targets, improve the specificity and robustness of lipid-based biomarkers, and enhance patient stratification in the context of precision oncology.
Collapse
Affiliation(s)
- Dominique Delmas
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France; Centre de Lutte Contre le Cancer Georges François Leclerc Center, Dijon F-21000, France; Inserm UMS58 - Biologie Santé Dijon (BioSanD), Dijon F-21000, France.
| | - Aurélie Mialhe
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Alexia K Cotte
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Jean-Louis Connat
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Florence Bouyer
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - François Hermetet
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Virginie Aires
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| |
Collapse
|
4
|
Kloeckner AA, Walker SR. Endometriosis Cell Spheroids Undergo Mesothelial Clearance in a Similar Manner to Ovarian Cancer Cell Spheroids. Cells 2025; 14:742. [PMID: 40422245 PMCID: PMC12110144 DOI: 10.3390/cells14100742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/10/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Endometriosis is a gynecological disease characterized by the presence of endometrium-like cells located outside the uterus. The most widely accepted theory for endometriosis development, retrograde menstruation, does not account for extra-pelvic lesions or ones found on other organs in the peritoneal cavity. Similar to ovarian cancer, endometriosis cells can interact with the mesothelial cells of the peritoneal cavity. In ovarian cancer metastasis, ovarian cancer cell spheroids attach and push away the mesothelial cells lining the peritoneal cavity, clearing the mesothelial layer. Since endometriosis cells are known to interact with the mesothelium, we hypothesized that endometriosis cells would be able to form spheroids capable of undergoing mesothelial clearance. To test this, we designed an in vitro mesothelial clearance assay using endometriosis spheroids and a mesothelial cell monolayer. Our results demonstrate that normal and endometriotic epithelial cell spheroids can perform mesothelial clearance similar to ovarian cancer spheroids, though normal endometrial cells do not clear as well as endometriosis cells. Additionally, we demonstrated that our mesothelial clearance assay can test potential pharmacological therapies for endometriosis prior to clinical trials. These results give insight into the development of endometriosis lesions, but further research is needed to determine the mechanisms behind mesothelial clearance in endometriosis.
Collapse
Affiliation(s)
| | - Sarah R. Walker
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA
| |
Collapse
|
5
|
Paul JK, Azmal M, Talukder OF, Ghosh A. Statin use and Pancreatic Cancer: A Meta-analysis of its Association with Incidence in the General Population and Survival in Patients. J Gastrointest Cancer 2025; 56:121. [PMID: 40379858 DOI: 10.1007/s12029-025-01238-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2025] [Indexed: 05/19/2025]
Abstract
PURPOSE Statins have been appearing as a potential anti-cancer agent in numerous studies. The study aimed to unravel the impact of statins in pancreatic cancer in terms of reducing the occurrence (morbidity) and improving survival (mortality). METHODS A comprehensive search of databases was carried out to collect the eligible studies up to July 2024. This meta-analysis evaluates two distinct questions: (1) whether statin use reduces the incidence of pancreatic ductal adenocarcinoma (PDAC) in the general population, and (2) whether statins improve survival among patients diagnosed with PDAC. In total, 39 studies were included in the meta-analysis, comprising 15 case-control studies, 20 cohort studies, three randomized controlled trials, and one non-randomized controlled trial. A generic inverse variance weighted random-effects model was applied to calculate the pooled risk ratio and 95% confidence intervals. Subgroup analyses were performed based on the availability of relevant information. RESULTS In the total meta-analysis, aggregated results demonstrated a substantial decrease in pancreatic cancer risk in all statin users (RR 0.94; 95% CIs, 0.90-0.97, and p-value = 0.0008). The pooled risk ratio estimate of lipophilic statins was 0.97 (95% CI, 0.87-1.07; P = 0.50; I2 = 0.0%). The estimated pooled risk ratios of long-term and short-term statin use were 0.80 (95% CI, 0.69-0.92; P = 0.002; I2 = 42%) and 0.86 (95% CI, 0.70-1.06; P = 0.15; I2 = 96%), respectively. For long-term and short-term follow-up, the risk ratios were 0.81 (95% CI, 0.70-0.94; P = 0.007; I2 = 55%) and 0.96 (95% CI, 0.90-1.02; P = 0.16; I2 = 26%), respectively. As for the studies collectively, heterogeneity was tested using the Cochrane chi square test (p-value = = 0.40, I2 = 4%). No publication bias was found. CONCLUSION The overall outcome of the study indicates that statins might lower the occurrence and increase the survival of PDAC patients.
Collapse
Affiliation(s)
- Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Omar Faruk Talukder
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh.
| |
Collapse
|
6
|
Yen WC, Yang MH, Weng TH, Chung CH, Tsao CH, Tsao CW, Meng E, Wu ST, Chien WC, Kao CC. Dialysis and the risk of early urological cancer: A nationwide population-based cohort study in Taiwan. Medicine (Baltimore) 2025; 104:e42521. [PMID: 40388720 PMCID: PMC12091643 DOI: 10.1097/md.0000000000042521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 05/01/2025] [Indexed: 05/21/2025] Open
Abstract
Patients with end-stage renal disease (ESRD) are predisposed to a higher risk of developing malignancies. This study aimed to explore the association between chronic dialysis with ESRD treated and the subsequent development of urothelial cell carcinoma or renal cell carcinoma (UC/RCC). Data spanning 13 years were retrieved from Taiwan's National Health Insurance Research Database. A total of 11,820 patients with ESRD undergoing maintenance dialysis between January 1, 2000, and December 31, 2013, and 35,460 controls matched for sex, age, and index year, were identified. After adjusting for confounding factors, Cox proportional hazards analysis was performed to compare the risk of UC/RCC during the 13-year follow-up period, and Kaplan-Meier analysis was used to evaluate the cumulative UC/RCC incidence between the ESRD and non-ESRD cohorts. The average time before developing UC/RCC was 4.18 years after dialysis initiation in the ESRD group compared to 5.39 years in the control group. After adjusting for sex, age, monthly income, urbanization level, geographic region, and comorbidities, the hazard ratio for UC/RCC was 1.186 (95% confidence interval, 1.071-1.448; P = .005). Stratified by age, the odds ratios (ORs) for developing UC/RCC were 2.105, 1.498, 1.371, and 0.925 among patients with ESRD aged 40 to 49, 50 to 59, 60 to 69, and ≥ 70 years, respectively. Stratification by comorbidities revealed ORs of 1.204, 1.179, 1.186,1.172, 1.211, and 1.210 for patients without diabetes mellitus, hyperlipidemia, obesity, coronary artery disease, chronic obstructive pulmonary disease, and hematuria, respectively. The mean time to UC/RCC occurrence was 4.18 years after dialysis. Furthermore, younger male patients undergoing dialysis with fewer comorbidities were at higher risk of developing UC/RCC. Early or more intensive surveillance for urological cancers post-dialysis initiation is recommended for patients undergoing dialysis with longer life expectancies or a higher likelihood of undergoing renal transplantation.
Collapse
Affiliation(s)
- Wei-Chen Yen
- Department of Surgery, Divisions of Urology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hsin Yang
- Department of Surgery, Divisions of Urology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tzu-Hsuan Weng
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Hsiang Chung
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Chang-Huei Tsao
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Wei Tsao
- Department of Surgery, Divisions of Urology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - En Meng
- Department of Surgery, Divisions of Urology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sheng-Tang Wu
- Department of Surgery, Divisions of Urology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wu-Chien Chien
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Chang Kao
- Department of Surgery, Divisions of Urology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
7
|
Dang Y, Zhang Y, Wang Z. The role of statins in the regulation of breast and colorectal cancer and future directions. Front Pharmacol 2025; 16:1578345. [PMID: 40438592 PMCID: PMC12116307 DOI: 10.3389/fphar.2025.1578345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/08/2025] [Indexed: 06/01/2025] Open
Abstract
Statins, widely recognized as a cornerstone in the prevention of cardiovascular diseases, have garnered increasing attention in oncology due to their pleiotropic effects, particularly their potential roles in regulating breast and colorectal cancer. Emerging evidence suggests that statins may exert anticancer effects through multiple mechanisms, including the mitochondrial apoptosis pathway, the LKB1-AMPK-p38MAPK-p53-survivin signaling cascade, inhibition of the mevalonate pathway, modulation of the EGFR/RhoA and IGF-1 signaling pathways, and regulation of the BMP/SMAD4 signaling pathway. However, significant heterogeneity exists in the reported anticancer effects of statins, likely due to variations in statin type (lipophilic vs hydrophilic), dosage, treatment duration, and population-specific characteristics. These factors contribute to inconsistencies in study outcomes. Additionally, while combination therapies incorporating statins with chemotherapy and immunotherapy have demonstrated synergistic effects in certain studies, their clinical utility remains to be fully established. Nevertheless, current evidence suggests that statins may have a potential role in reducing breast and colorectal cancer-related mortality. Future research should prioritize elucidating their precise molecular mechanisms, defining dose-response relationships, developing personalized treatment strategies within the framework of precision medicine, and validating their efficacy through large-scale, long-term prospective studies. These efforts will provide a more robust scientific foundation for the clinical application of statins in oncology. This review systematically explores the role of statins in breast and colorectal cancer regulation, covering clinical evidence, underlying biological mechanisms, pharmacological distinctions, synergistic therapeutic potential, and translational medicine prospects.
Collapse
Affiliation(s)
| | | | - Zhihao Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Pu J, Yuan K, Tao J, Qin Y, Li Y, Fu J, Li Z, Zhou H, Tang Z, Li L, Gai X, Qin D. Glioblastoma multiforme: an updated overview of temozolomide resistance mechanisms and strategies to overcome resistance. Discov Oncol 2025; 16:731. [PMID: 40353925 PMCID: PMC12069213 DOI: 10.1007/s12672-025-02567-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with high lethality. The typical treatment regimen includes post-surgical radiotherapy and temozolomide (TMZ) chemotherapy, which helps extend survival. Nevertheless, TMZ resistance occurs in approximately 50% of patients. This resistance is primarily associated with the expression of O6-methylguanine-DNA methyltransferase (MGMT), which repairs O6-methylguanine lesions generated by TMZ and is thought to be the major mechanism of drug resistance. Additionally, the mismatch repair and base excision repair pathways play crucial roles in TMZ resistance. Emerging studies also point to drug transport mechanisms, glioma stem cells, and the heterogeneous tumor microenvironment as additional influences on TMZ resistance in gliomas. A better understanding of these mechanisms is vital for developing new treatments to improve TMZ effectiveness, such as DNA repair inhibitors, inhibitors of multidrug transporting proteins, TMZ analogs, and combination therapies targeting multiple pathways. This article discusses the main resistance mechanisms and potential strategies to counteract resistance in GBM patients, aiming to broaden the understanding of these mechanisms for future research and to explore the therapeutic effects of traditional Chinese medicines and their active components in overcoming TMZ resistance.
Collapse
Affiliation(s)
- Jianlin Pu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Kai Yuan
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Jian Tao
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Yuliang Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Yongxin Li
- Department of Rehabilitation Medicine, Mojiang Hani Autonomous Country Hospital of Traditional Chinese Medicine, Mojiang, China
| | - Jing Fu
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhong Li
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, China
| | - Haimei Zhou
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhengxiu Tang
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China
| | - Li Li
- Department of Emergency Trauma Surgery, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xuesong Gai
- Department of Rehabilitation Medicine, The First People's Hospital of Yunnan Province, Kunming, China.
| | - Dongdong Qin
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming, China.
| |
Collapse
|
9
|
Yeom S, Lee DH, Song J. Therapeutic Potential of Anti-Diabetes Drugs and Anti-Dyslipidemia Drugs to Mitigate Head and Neck Cancer Risk in Metabolic Syndrome. CNS Neurosci Ther 2025; 31:e70446. [PMID: 40387523 PMCID: PMC12087305 DOI: 10.1111/cns.70446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/24/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Head and neck cancer (HNC) encompasses a heterogeneous group of malignancies originating in the oral cavity, pharynx, nasopharynx, larynx, paranasal sinuses, and salivary glands. Accumulating evidence indicates that metabolic syndrome (MetS) characterized by a constellation of conditions including central adiposity, hyperglycemia, dyslipidemia, hypertension, and insulin resistance, may significantly influence cancer pathogenesis and progression. RESULTS MetS has been epidemiologically linked to elevated risk for multiple malignancies through various metabolic mechanisms involving chronic systemic inflammation, insulin resistance, and dysregulated lipid metabolism. Especially in HNC, recent studies demonstrated that MetS and metabolic imbalance conditions may contribute to carcinogenesis, disease progression, and clinical outcomes, but the exact mechanisms behind the association between excess fat accumulation and HNC risk remain unclear. Considering previous studies, pharmacological agents targeting metabolic pathways, including biguanides (metformin), thiazolidinediones, sodium-glucose cotransporter-2 (SGLT-2) inhibitors, and HMG-CoA reductase inhibitors (statins) are being investigated for potential repurposing in cancer prevention and adjuvant therapy. CONCLUSIONS Here, we summarize the latest evidence on the relationship between MetS and HNC, highlighting the therapeutic potential of anti-diabetes drugs and anti-dyslipidemia drugs in ameliorating various pathological problems in HNC patients with MetS.
Collapse
Affiliation(s)
- Sujung Yeom
- Department of Otolaryngology‐Head and Neck SurgeryChonnam National University Medical School & Hwasun HospitalHwasunRepublic of Korea
| | - Dong Hoon Lee
- Department of Otolaryngology‐Head and Neck SurgeryChonnam National University Medical School & Hwasun HospitalHwasunRepublic of Korea
| | - Juhyun Song
- Department of AnatomyChonnam National University Medical SchoolHwasunRepublic of Korea
| |
Collapse
|
10
|
Ren X, Jin C, Li Q, Fu C, Fang Y, Xu Z, Liang Z, Wang T. Fatty acid binding proteins-mediated mitochondrial dysfunction in the development of age-related diseases: A review. Int J Biol Macromol 2025; 309:142913. [PMID: 40203912 DOI: 10.1016/j.ijbiomac.2025.142913] [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: 12/30/2024] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Fatty acid-binding proteins (FABPs) act as lipid chaperones and play a role in the pathological processes of various lipid signaling pathways. Mitochondria are crucial for the regulation of lipid metabolism. As an aging marker, lipid-mediated mitochondrial dysfunction has been observed in the etiology of numerous diseases, including neurodegenerative diseases, metabolic syndromes, cardiovascular diseases, and tumorigenesis. Members of the FABP family have been identified to regulate mitochondrial function. Targeting FABPs specifically may provide a promising approach to improve mitochondrial function and treat age-related diseases. This review summarizes the connection between FABPs and mitochondrial function and highlights certain FABPs involved in age-related diseases that hold significant therapeutic promise.
Collapse
Affiliation(s)
- Xingxing Ren
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Chaoyuan Jin
- Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Qilin Li
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Congyi Fu
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Yu Fang
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Zihang Xu
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200125, China
| | - Zi Liang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Tianshi Wang
- Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201207, China.
| |
Collapse
|
11
|
Eckel O, Mirea MA, Gschwendtner A, Pistek M, Kinslechner K, Röhrl C, Stangl H, Hengstschläger M, Mikula M. Expression of the cholesterol transporter SR-B1 in melanoma cells facilitates inflammatory signaling leading to reduced cholesterol synthesis. Neoplasia 2025; 63:101154. [PMID: 40120430 PMCID: PMC11981749 DOI: 10.1016/j.neo.2025.101154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
Scavenger receptor class B type 1 (SR-B1) is a cholesterol transporter, abundantly expressed in human melanoma, yet its precise role for melanoma progression is not fully understood. This study investigates the involvement of SR-B1 in cholesterol homeostasis of tumor cells and its implications for potential therapy. We found that SR-B1 depletion in melanoma cells does not alter total cholesterol levels, but induces cholesterol biosynthesis. This effect was characterized by an increased expression of HMG-CoA reductase (HMGCR), a rate limiting enzyme of cholesterol biosynthesis. Notably, further analyses indicated that this regulation occurs at the post-translational level, mediated via the hypoxia-inducible factor (HIF) signaling pathway. Importantly, we identified SR-B1 as a transporter of the lipid hormone sphingosine-1-phosphate (S1P) and we found that S1P exposure leads to HIF1A up-regulation. Finally, we used a pluripotent stem cell-derived skin organoid model to show that targeting SR-B1 in combination with targeted melanoma therapy can lead to increased apoptosis and suppressed proliferation of transplanted tumor cells. Our study shows that functional SR-B1 is linked to inflammatory signaling, which reduces cholesterol synthesis, while enabling melanoma cell survival during chemotherapy treatment.
Collapse
Affiliation(s)
- Oliver Eckel
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Madalina A Mirea
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Anna Gschwendtner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Martina Pistek
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Katharina Kinslechner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Clemens Röhrl
- University of Applied Sciences Upper Austria, Faculty of Engineering, Stelzhamerstraße 23, 4600, Wels, Austria
| | - Herbert Stangl
- Institute of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Mario Mikula
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| |
Collapse
|
12
|
Zhang W, Xu Y, Fang Y, Li M, Li D, Guo H, Li H, He J, Miao L. Ubiquitination in lipid metabolism reprogramming: implications for pediatric solid tumors. Front Immunol 2025; 16:1554311. [PMID: 40370434 PMCID: PMC12075147 DOI: 10.3389/fimmu.2025.1554311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Pediatric solid tumors represent a significant subset of childhood cancers, accounting for approximately 60% of new diagnoses. Despite advancements in therapeutic strategies, survival rates remain markedly disparate between high-income and resource-limited settings, underscoring the urgent need for novel and effective treatments. Lipid metabolic reprogramming is a fundamental hallmark of cancer, driving tumor progression, therapeutic resistance, and immune evasion through enhanced fatty acid uptake, increased de novo lipid synthesis, and activated fatty acid β-oxidation (FAO). Ubiquitination, a dynamic post-translational modification mediated by the ubiquitin-proteasome system (UPS), plays a crucial role in regulating lipid metabolism by modulating the stability and activity of key metabolic enzymes and transporters involved in cholesterol and fatty acid pathways. This review comprehensively examines the complex interplay between ubiquitination and lipid metabolic reprogramming in pediatric solid tumors. It delineates the mechanisms by which ubiquitination influences cholesterol biosynthesis, uptake, efflux, and fatty acid synthesis and oxidation, thereby facilitating tumor growth and survival. Furthermore, the review identifies potential UPS-mediated therapeutic targets and explores the feasibility of integrating ubiquitination-based strategies with existing treatments. By targeting the UPS to disrupt lipid metabolism pathways, novel therapeutic avenues may emerge to enhance treatment efficacy and overcome resistance in pediatric oncology. This synthesis of current knowledge aims to provide a foundation for the development of innovative, precision medicine approaches to improve clinical outcomes for children afflicted with solid tumors.
Collapse
Affiliation(s)
- Weixin Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Yile Xu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Yingjin Fang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
| | - Meng Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Di Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Huiqin Guo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Hang Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Ghogare SS, Pathan EK. Intratumor fungi specific mechanisms to influence cell death pathways and trigger tumor cell apoptosis. Cell Death Discov 2025; 11:188. [PMID: 40258837 PMCID: PMC12012188 DOI: 10.1038/s41420-025-02483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer, uncontrolled cell growth due to the loss of cell cycle regulation, is often found to be associated with viral infections and, as recent studies show, with bacterial infections as well. Emerging reports also suggest a strong link between fungi and cancer. The crucial virulence trait of fungi, the switch from yeast (Y) to hyphal (H) form, is found to be associated with carcinogenesis. The physicochemical properties and signal transduction pathways involved in the switch to the hyphal form overlap with those of tumor cell formation. Inhibiting differentiation causes apoptosis in fungi, whereas preventing apoptosis leads to cancer in multicellular organisms. Literature on the fungi-cancer linkage, though limited, is increasing rapidly. This review examines cancer-specific fungal communities, the impact of fungal microbiome on cancer cell progression, similarities between fungal differentiation and cells turning cancerous at biochemical and molecular levels, including the overlaps in signal transduction pathways between fungi and cancer. Based on the available evidence, we suggest that molecules inhibiting the yeast-hyphal transition in fungi can be combined with those targeting tumor cell apoptosis for effective cancer treatment. The review points out fertile research areas where mycologists and cancer researchers can collaborate to unravel common molecular mechanisms. Moreover, antibodies targeting fungal-specific chitin and glucan can be used for the selective neutralization of tumor cells. These new combinations of potential therapies are expected to facilitate the development of target-specific, less harmful and commercially feasible anticancer therapies. We bring together available evidence to argue that fungal infections could either trigger cancer or have a significant role in the development and progression of cancer. Hence, cancer-associated fungal populations could be utilized as a target for a combination therapy involving the integration of anticancer and antifungal drugs as well as inhibitors of fungal morphogenesis to develop more effective anticancer therapies.
Collapse
Affiliation(s)
- Simran S Ghogare
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) Lavale, Pune, 412115, Maharashtra, India
| | - Ejaj K Pathan
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
14
|
Addanki S, Kim L, Stevens A. Understanding and Targeting Metabolic Vulnerabilities in Acute Myeloid Leukemia: An Updated Comprehensive Review. Cancers (Basel) 2025; 17:1355. [PMID: 40282531 PMCID: PMC12025543 DOI: 10.3390/cancers17081355] [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: 02/21/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Acute Myeloid Leukemia (AML) is characterized by aggressive proliferation and metabolic reprogramming that support its survival and resistance to therapy. This review explores the metabolic distinctions between AML cells and normal hematopoietic stem cells (HSCs), emphasizing the role of altered mitochondrial function, oxidative phosphorylation (OXPHOS), and biosynthetic pathways in leukemic progression. AML cells exhibit distinct metabolic vulnerabilities, including increased mitochondrial biogenesis, reliance on glycolysis and amino acid metabolism, and unique signaling interactions that sustain leukemic stem cells (LSCs). These dependencies provide potential therapeutic targets, as metabolic inhibitors have demonstrated efficacy in disrupting AML cell survival while sparing normal hematopoietic cells. We examine current and emerging metabolic therapies, such as inhibitors targeting glycolysis, amino acid metabolism, and lipid biosynthesis, highlighting their potential in overcoming drug resistance. However, challenges remain in translating these strategies into clinical practice due to AML's heterogeneity and adaptability. Further research into AML's metabolic plasticity and precision medicine approaches is crucial for improving treatment outcomes. Understanding and exploiting AML's metabolic vulnerabilities could pave the way for novel, more effective therapeutic strategies.
Collapse
Affiliation(s)
- Sridevi Addanki
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Alexandra Stevens
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
15
|
Aizawa F, Yagi K, Sato M, Niimura T, Goda M, Izawa-Ishizawa Y, Ishizawa K. Influence of statin intervention on peripheral neuropathy in patients treated with anticancer drugs identified from the insurer database. J Pharm Health Care Sci 2025; 11:27. [PMID: 40197323 PMCID: PMC11978124 DOI: 10.1186/s40780-025-00428-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Statins, hydroxymethylglutaryl-CoA reductase inhibitors, possess neuroprotective properties. Given the potential neuroprotective properties of statins and their prevalent use in clinical settings, we aimed to investigate their impact on chemotherapy-induced peripheral neuropathy (CIPN) in Japan by assessing both their safety and efficacy in this context. METHODS We conducted a retrospective observational study using the Japan Medical Data Centre database, which includes data from 2005 to 2021. We included patients who underwent anticancer therapy and were categorized into non-statin (10,920) and statin (1,537) groups. These groups were matched using a propensity score, resulting in 2,548 non-statin and 1,274 statin users. The primary endpoints were the incidence of CIPN post-first prescription of each anticancer drug and overall survival. RESULTS Treatment with statins did not increase the incidence of CIPN (non-statin 27.2% vs. statin 28.4%, P = 0.443). Nevertheless, the incidence of CIPN was significantly high among women (non-statin 28.0% vs. statin 33.2%, P = 0.025). Overall survival was not impacted by statin use (hazard ratio 0.98, 95%CI: 0.83-1.16, P = 0.8846). Among men treated with paclitaxel, we observed an improvement in overall survival (hazard ratio: 0.72; 95% CI: 0.56-0.92; P = 0.0110). CONCLUSIONS The use of statins in patients with cancer was not associated with CIPN incidence. However, in men receiving paclitaxel treatment, statins may be linked to improved overall survival. Further studies are necessary to clarify the factors influencing prognosis and CIPN severity.
Collapse
Affiliation(s)
- Fuka Aizawa
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Kenta Yagi
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.
- Clinical Research Centre for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan.
| | - Maki Sato
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takahiro Niimura
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
- Clinical Research Centre for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Mitsuhiro Goda
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yuki Izawa-Ishizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
- Department of General Medicine, Taoka Hospital, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
- Clinical Research Centre for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| |
Collapse
|
16
|
Wu Y, Song W, Su M, He J, Hu R, Zhao Y. The Role of Cholesterol Metabolism and Its Regulation in Tumor Development. Cancer Med 2025; 14:e70783. [PMID: 40145543 PMCID: PMC11948085 DOI: 10.1002/cam4.70783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Within the tumor microenvironment, tumor cells undergo metabolic reprogramming of cholesterol due to intrinsic cellular alterations and changes in the extracellular milieu. Furthermore, cholesterol reprogramming within this microenvironment influences the immune landscape of tumors, facilitating immune evasion and consequently promoting tumorigenesis. These biological changes involve modifications in numerous enzymes associated with cholesterol uptake and synthesis, including NPC1L1, SREBP, HMGCR, SQLE, and PCSK9. REVIEW This review systematically summarizes the role of cholesterol metabolism and its associated enzymes in cancer progression, examines the mechanisms through which dysregulation of cholesterol metabolism affects immune cells within the tumor microenvironment, and discusses recent advancements in cancer therapies that target cholesterol metabolism. CONCLUSION Targeting cholesterol metabolism-related enzymes can inhibit tumor growth, reshape immune landscapes, and rejuvenate antitumor immunity, offering potential therapeutic avenues in cancer treatment.
Collapse
Affiliation(s)
- Yongmei Wu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Wenqian Song
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Min Su
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Jing He
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Rong Hu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Youbo Zhao
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| |
Collapse
|
17
|
Patnaik R, Varghese RL, Khan S, Huda B, Bhurka F, Amiri L, Banerjee Y. Targeting PAR-2-driven inflammatory pathways in colorectal cancer: mechanistic insights from atorvastatin and rosuvastatin treatment in cell line models. Transl Cancer Res 2025; 14:1531-1566. [PMID: 40224964 PMCID: PMC11985218 DOI: 10.21037/tcr-24-1027] [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: 06/20/2024] [Accepted: 01/27/2025] [Indexed: 04/15/2025]
Abstract
Background Colorectal cancer (CRC) is a growing health concern globally and in regions such as the United Arab Emirates, where risk factors like obesity and hyperlipidaemia are prevalent. Chronic inflammation, driven by pathways involving protease-activated receptor 2 (PAR-2), plays a pivotal role in CRC progression, creating a tumour-promoting microenvironment. The overexpression of PAR-2 has been associated with increased tumour aggressiveness and drug resistance. While previous studies have focused on broad inflammatory modulation, this study explores the selective targeting of PAR-2 by atorvastatin (ATV) and rosuvastatin (RSV), highlighting their specificity by assessing minimal impact on PAR-1 expression, which serves as a control. Methods HT-29 and Caco-2 CRC cell lines were employed to investigate the anti-inflammatory effects of ATV and RSV. Inflammation was induced with lipopolysaccharide (LPS), followed by treatment with varying concentrations of ATV and RSV. Western blotting and real-time polymerase chain reaction for quantification (qPCR) were performed to quantify PAR-2 and TNF-α at both the protein and mRNA levels. Enzyme linked immunosorbent assay (ELISA) was used to measure the secretion of TNF-α. Calcium signalling, which plays a crucial role in inflammation, was analysed using Fluo-4 AM dye, with fluorescence imaging capturing the effects of statin treatment on intracellular calcium influx. Results LPS treatment significantly upregulated PAR-2 and TNF-α expression in both cell lines, validating the inflammatory model. Co-treatment with ATV or RSV reduced PAR-2 and TNF-α expression in a dose-dependent manner. The higher concentrations of ATV (50 µg/mL) and RSV (20 µg/mL) produced the most significant reduction in these inflammatory markers at both the protein and mRNA levels. Importantly, the treatment did not substantially alter PAR-1 expression, underlining the specificity of ATV and RSV in modulating PAR-2-mediated pathways. Additionally, statin treatment attenuated LPS-induced calcium influx, with fluorescence intensity decreasing markedly at higher concentrations of both statins. Conclusions This study provides novel insights into the selective targeting of PAR-2 by ATV and RSV, distinguishing their effects from PAR-1. The reduction in PAR-2 expression and TNF-α secretion, along with the suppression of calcium signalling, underscores the potential of these statins as targeted anti-inflammatory agents in CRC. The findings highlight the therapeutic value of ATV and RSV in modulating inflammation through PAR-2-specific pathways, which may contribute to reduced cancer progression. These results pave the way for further preclinical and clinical evaluations to explore statins as adjunctive therapies in the management of CRC.
Collapse
Affiliation(s)
- Rajashree Patnaik
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Riah Lee Varghese
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Sara Khan
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Bintul Huda
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Farida Bhurka
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Layla Amiri
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Yajnavalka Banerjee
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
- Centre for Medical Education, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| |
Collapse
|
18
|
Sun D, Cui X, Yang W, Wei M, Yan Z, Zhang M, Yu W. Simvastatin inhibits PD-L1 via ILF3 to induce ferroptosis in gastric cancer cells. Cell Death Dis 2025; 16:208. [PMID: 40140647 PMCID: PMC11947124 DOI: 10.1038/s41419-025-07562-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
The treatment of gastric cancer remains challenging, with immunotherapy serving as a critical component of the holistic approach to its treatment. The results of this study indicated that statins could decrease the serum levels of interleukin-enhancing binding factor 3 (ILF3) and programmed cell death ligand 1(PD-L1) in GC patients and improve their prognosis. Functional experiments demonstrated that simvastatin induced ferroptosis by inhibiting ILF3 in GC cells and enhanced the killing effect of activated CD8+ T cells on GC cells. The CUT&Tag assay revealed that, mechanistically, simvastatin inhibited ILF3 expression by reducing the acetylation level at residue site H3K14 in ILF3. Next-generation sequencing and Kyoto Encyclopedia of Genes and Genomes analysis revealed that ILF3 regulated PD-L1 expression through the DEPTOR/mTOR signaling pathway. Overall, simvastatin induced ferroptosis in GC cells by inhibiting ILF3 expression while promoting the activation of CD8+ T cells to augment antitumor immune responses, thereby facilitating synergistic immunotherapy.
Collapse
Affiliation(s)
- Danping Sun
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Xiaohan Cui
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Wenshuo Yang
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Meng Wei
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Zhibo Yan
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China
| | - Wenbin Yu
- Department of Gastrointestinal Surgery, General Surgery, Qilu Hospital of Shandong University, 107 West Wen Hua Road, Jinan, 250012, China.
| |
Collapse
|
19
|
Wang Z, Xu C, Wang Q, Wang Y. Repurposing of nervous system drugs for cancer treatment: recent advances, challenges, and future perspectives. Discov Oncol 2025; 16:396. [PMID: 40133751 PMCID: PMC11936871 DOI: 10.1007/s12672-025-02067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
The nervous system plays a critical role in developmental biology and oncology, influencing processes from ontogeny to the complex dynamics of cancer progression. Interactions between the nervous system and cancer significantly affect oncogenesis, tumor growth, invasion, metastasis, treatment resistance, inflammation that promotes tumors, and the immune response. A comprehensive understanding of the signal transduction pathways involved in cancer biology is essential for devising effective anti-cancer strategies and overcoming resistance to existing therapies. Recent advances in cancer neuroscience promise to establish a new cornerstone of cancer therapy. Repurposing drugs originally developed for modulating nerve signal transduction represent a promising approach to target oncogenic signaling pathways in cancer treatment. This review endeavors to investigate the potential of repurposing neurological drugs, which target neurotransmitters and neural pathways, for oncological applications. In this context, it aims to bridge the interdisciplinary gap between neurology, psychiatry, internal medicine, and oncology. By leveraging already approved drugs, researchers can utilize existing extensive safety and efficacy data, thereby reducing both the time and financial resources necessary for the development of new cancer therapies. This strategy not only promises to enhance patient outcomes but also to expand the array of available treatments, thereby enriching the therapeutic landscape in oncology.
Collapse
Affiliation(s)
- Zixun Wang
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Chen Xu
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Qi Wang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Yudong Wang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
20
|
Dai CL, Qiu ZY, Wang AQ, Yan S, Zhang LJ, Luan X. Targeting cholesterol metabolism: a promising therapy strategy for cancer. Acta Pharmacol Sin 2025:10.1038/s41401-025-01531-9. [PMID: 40133625 DOI: 10.1038/s41401-025-01531-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/03/2025] [Indexed: 03/27/2025]
Abstract
Cholesterol is a crucial structural component of cell membranes, playing a vital role in maintaining membrane fluidity and stability. Cholesterol metabolism involves four interconnected processes: de novo synthesis, uptake, efflux, and esterification. Disruptions in any of these pathways can lead to imbalances in cholesterol homeostasis, which are significantly associated with cancer progression. In recent years, traditional Chinese medicine (TCM) has emerged as a comprehensive therapeutic approach with multi-target and multi-pathway effects, demonstrating significant potential in regulating cholesterol metabolism. Research has shown that certain components of TCM can modulate enzymes, transport proteins, and signaling pathways involved in cholesterol metabolism, effectively interfering with survival and migration of cancer. These mechanisms highlight the unique advantages of TCM in inhibiting tumor progression. In this review we systematically describe the execution and regulation of the four key cholesterol metabolism processes, highlights the roles of critical proteins involved, and provides a comprehensive overview of natural products from TCM that modulate cholesterol metabolism. This review provides valuable insights for the development of novel drugs and cancer therapeutic strategies targeting cholesterol metabolism.
Collapse
Affiliation(s)
- Chun-Lan Dai
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zi-Yang Qiu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - An-Qi Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shen Yan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li-Jun Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
21
|
Cho HJ, Lee E, Kim SS, Cheong JY. Impact of Antihypertensive and Lipid-Lowering Agents on Hepatocellular Carcinoma Risk in Patients with Fatty Liver Disease and Diabetes. Dig Dis Sci 2025:10.1007/s10620-025-08935-x. [PMID: 40108104 DOI: 10.1007/s10620-025-08935-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND/AIM This study aimed to evaluate the effects of antihypertensives, lipid-lowering agents, and antiplatelet medications on hepatocellular carcinoma (HCC) risk in patients with fatty liver disease (FLD) and type 2 diabetes (T2D). METHOD Using data from Korea Health Insurance Review and Assessment Service, 212,443 FLD-T2D patients were analyzed through Cox regression, propensity score matching (PSM), and Kaplan-Meier analysis. The analysis considered medication use and its relation to HCC development. Cohort admission day was set as the date of the first oral hypoglycemic prescription. RESULTS The multivariate Cox regression analysis revealed that old age, male sex, chronic viral hepatitis, alcoholic liver disease, liver cirrhosis, using a combination of insulin and oral hypoglycemic agents for antidiabetic treatment, and calcium channel blocker (CCB) use were significantly correlated with higher HCC development risk, whereas dyslipidemia and statin, ezetimibe, and fibrate use was correlated with lower HCC risk, in the study cohort of 212,443 patients. Patients who used statins (hazard ratio [HR] = 0.58, 95% confidence interval [CI] = 0.42-0.80, P = 0.001) and fibrates (HR = 0.46, 95% CI = 0.22-0.93, P = 0.031) showed a significantly lower risk of HCC development even after PSM. In contrast, CCB use was linked to an elevated HCC risk (HR = 1.35, 95% CI = 1.05-1.72, P = 0.019), highlighting the differential impact of various medications on HCC incidence. CONCLUSION The use of specific medications, such as statins and fibrates, may offer protective effects against HCC in patients with FLD-T2D, whereas that of CCB may increase the risk. This underscores the importance of tailored medication strategies for the management of chronic conditions.
Collapse
Affiliation(s)
- Hyo Jung Cho
- Department of Gastroenterology, Ajou University School of Medicine, Worldcup-Ro 164, Yeongtong-Gu, Suwon, 16499, South Korea
| | - Eunyoung Lee
- Department of Neurology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Soon Sun Kim
- Department of Gastroenterology, Ajou University School of Medicine, Worldcup-Ro 164, Yeongtong-Gu, Suwon, 16499, South Korea
| | - Jae Youn Cheong
- Department of Gastroenterology, Ajou University School of Medicine, Worldcup-Ro 164, Yeongtong-Gu, Suwon, 16499, South Korea.
| |
Collapse
|
22
|
Patil PS, Saklani A, Kumar NAN, De’Souza A, Krishnatry R, Khanvilkar S, Kazi M, Engineer R, Ostwal V, Ramaswamy A, Bal M, Ranganathan P, Gupta E, Galande S. A randomized phase II/III trial of rosuvastatin with neoadjuvant chemo-radiation in patients with locally advanced rectal cancer. Front Oncol 2025; 15:1450602. [PMID: 40177244 PMCID: PMC11961435 DOI: 10.3389/fonc.2025.1450602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 02/18/2025] [Indexed: 04/05/2025] Open
Abstract
Aim Statins have been shown to improve the possibility of a pathological complete response (pCR) in patients with locally advanced rectal cancer when given in combination with neo-adjuvant chemo-radiation (NACTRT) in observational studies. The primary objective of this phase II randomized controlled trial (RCT) is to determine the impact of rosuvastatin in improving pCR rates in patients with locally advanced rectal cancer who are undergoing NACTRT. The secondary objectives are to compare adverse events, postoperative morbidity and mortality, disease-free survival (DFS), and overall survival in the two arms and to identify potential prognostic and predictive factors determining outcomes. If the study is positive, we plan to proceed to a phase III RCT with 3-year DFS as the primary endpoint. Methods This is a prospective, randomized, open-label phase II/III study. The phase II study has a sample size of 316 patients (158 in each arm) to be accrued over 3 years to have 288 evaluable patients. The standard arm will receive NACTRT while the intervention group will receive 20 mg rosuvastatin orally once daily along with NACTRT for 6 weeks followed by rosuvastatin alone for 6-10 weeks until surgery. All patients will be reviewed after repeat imaging by a multidisciplinary tumor board at 12-16 weeks after starting NACTRT and operable patients will be planned for surgery. The pathological response rate, tumor regression grade (TRG), and post-surgical complications will be recorded. Conclusion The addition of rosuvastatin to NACTRT may improve the oncological outcomes by increasing the likelihood of pCR in patients with locally advanced rectal cancer undergoing NACTRT. This would be a low-cost, low-risk intervention that could potentially lead to the refinement of strategies, such as "watch and wait", in a select subgroup of patients. Clinical trial registration Clinical Trials Registry of India, identifier CTRI/2018/11/016459.
Collapse
Affiliation(s)
- Prachi S. Patil
- Department of Digestive Diseases and Clinical Nutrition, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Avanish Saklani
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Naveena A. N. Kumar
- Department of Surgical Oncology, Manipal Comprehensive Cancer Care Center, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Ashwin De’Souza
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Rahul Krishnatry
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Snehal Khanvilkar
- Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Mufaddal Kazi
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Reena Engineer
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha national Institute, Mumbai, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha national Institute, Mumbai, India
| | - Munita Bal
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Priya Ranganathan
- Department of Anaesthesiology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Ekta Gupta
- Laboratory of Chromatin Biology and Epigenetics, Department of Biology, Indian Institute of Science Education and Research, Pune, India
- Centre of Excellence in Epigenetics, Department of Life Sciences, Shiv Nadar Institution of Eminence, Delhi, India
| | - Sanjeev Galande
- Laboratory of Chromatin Biology and Epigenetics, Department of Biology, Indian Institute of Science Education and Research, Pune, India
- Centre of Excellence in Epigenetics, Department of Life Sciences, Shiv Nadar Institution of Eminence, Delhi, India
| |
Collapse
|
23
|
ALZHRANI RIYADF, BINOBAID LAMA, ALORAINI ABDULAZIZA, ALSAHLI MESHALS, BAKHEIT AHMEDH, ASIRI HANADIH, ATTIA SABRYM, ALHOSHANI ALI, HARISA GAMALELDINI. ERLNs augment simultaneous delivery of GFSV into PC-3 cells: Influence of drug combination on SDH, GPX-4, 5α-RD, and cytotoxicity. Oncol Res 2025; 33:919-935. [PMID: 40191728 PMCID: PMC11964872 DOI: 10.32604/or.2024.054537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/04/2024] [Indexed: 04/09/2025] Open
Abstract
Objective Prostate cancer (PCA) is the second most widespread cancer among men globally, with a rising mortality rate. Enzyme-responsive lipid nanoparticles (ERLNs) are promising vectors for the selective delivery of anticancer agents to tumor cells. The goal of this study is to fabricate ERLNs for dual delivery of gefitinib (GF) and simvastatin (SV) to PCA cells. Methods ERLNs loaded with GF and SV (ERLNGFSV) were assembled using bottom-up and top-down techniques. Subsequently, these ERLN cargoes were coated with triacylglycerol, and phospholipids and capped with chitosan (CS). The ERLNGFSV, and CS engineered ERLNGFSV (CERLNGFSV) formulations were characterized for particle size (PS), zeta potential (ZP), and polydispersity index (PDI). The biocompatibility, and cytotoxicity of the plain and GF plus SV-loaded ERLN cargoes were assessed using erythrocytes and PC-3 cell line. Additionally, molecular docking simulations (MDS) were conducted to examine the influence of GF and SV on succinate dehydrogenase (SDH), glutathione peroxidase-4 (GPX-4), and 5α-reductase (5α-RD). Results These results showed that plain, ERLNGFSV, and CERLNGFSV cargoes have a nanoscale size and homogeneous appearance. Moreover, ERLNGFSV and CERLNGFSV were biocompatible, with no detrimental effects on erythrocytes. Treatment with GF, SV, GF plus SV, ERLNGFSV, and CERLNGFSV significantly reduced the viability of PC-3 cells compared to control cells. Particularly, the blend of GF and SV, as well as ERLNGFSV and CERLNGFSV augmented PC-3 cell death. Also, treating PC-3 cells with free drugs, their combination, ERLNGFSV, and CERLNGFSV formulations elevated the percentage of apoptotic cells. MDS studies demonstrated that GF and SV interact with the active sites of SDH, GPX-4, and 5α-reductase. Conclusions This study concludes that SVGF combination and ERLNs loading induce particular delivery, and synergism on PC-3 death through action on multiple pathways involved in cell proliferation, and apoptosis, besides the interaction with SDH, GPX-4, and 5α-RD. Therefore, GFSV-loaded ERLN cargoes are a promising strategy for PCA treatment. In vivo studies are necessary to confirm these findings for clinical applications.
Collapse
Affiliation(s)
- RIYAD F. ALZHRANI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - LAMA BINOBAID
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - ABDULAZIZ A. ALORAINI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - MESHAL S. ALSAHLI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - AHMED H. BAKHEIT
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - HANADI H. ASIRI
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - SABRY M. ATTIA
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - ALI ALHOSHANI
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - GAMALELDIN I. HARISA
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
24
|
Wang L, He X, Shen Y, Chen J, Chen Y, Zhou Z, Xu X. A Novel Ferroptosis-Related Gene Prognosis Signature and Identifying Atorvastatin as a Potential Therapeutic Agent for Hepatocellular Carcinoma. Curr Issues Mol Biol 2025; 47:201. [PMID: 40136455 PMCID: PMC11940908 DOI: 10.3390/cimb47030201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Among the most common malignant tumors, hepatocellular carcinoma (HCC) is a primary liver cancer type that has a high mortality rate. HCC often presents insidiously, is prone to recurrence, and has limited treatment efficacy. Ferroptosis regulates tumorigenesis, progression, and metastasis, which is a novel form of iron-dependent cell death. Numerous studies suggest that HCC is sensitive to ferroptosis, indicating that targeted therapies aimed at inducing ferroptosis may represent a promising new approach to cancer treatment. This study aims to find genes associated with HCC and ferroptosis, as well as to screen for potential agents that may cause ferroptosis in HCC. Transcriptome and clinical sample data were obtained from the TCGA database to identify differentially expressed genes related to ferroptosis. Using various regression and survival analysis techniques, we developed a prognostic model based on four core genes and evaluated its predictive potential. Subsequently, we screened for potential therapeutic agents in the Connective Map (CMap) database, designated as compound Atorvastatin, based on differential genes from two risk groups and related to ferroptosis. Through experiments conducted in vivo and in vitro, we demonstrated that Atorvastatin can induce ferroptosis in HCC cells while inhibiting their growth and migration. In conclusion, this research targets ferroptosis therapy and provides new insights for improving the prediction and prevention of HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ximing Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; (L.W.); (X.H.); (Y.S.); (J.C.); (Y.C.); (Z.Z.)
| |
Collapse
|
25
|
Mosna MJ, Garde FJ, Stinson MG, Pastore CD, Carcagno AL. The chorioallantoic membrane (CAM) model: From its origins in developmental biology to its role in cancer research. Dev Biol 2025; 519:79-95. [PMID: 39694172 DOI: 10.1016/j.ydbio.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/24/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Over the past century, the chick embryo model, historically employed for research in developmental biology, has become a valuable tool for cancer research. The characteristics of the chick chorioallantoic membrane (CAM) make it a convenient model for the study of cancer, leading to the establishment of the CAM assay as an alternative to traditional in vivo cancer models. In this review we will explore the characteristics of the CAM that make it suitable for cancer research, as well as its consolidation as a versatile platform in this field. We will put particular emphasis on describing the key features that make this model an important asset for studying the hallmarks of cancer and for testing a wide variety of therapeutic strategies for its treatment, and which make it a suitable host for patient-derived xenografts (PDX). Additionally, we will examine the wide spectrum of methodological approaches available to study these subjects, highlighting some innovative cases. Finally, we will discuss the advantages and disadvantages of the chick CAM as a model for cancer research and how we can improve this model to its full potential.
Collapse
Affiliation(s)
- María Jimena Mosna
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Federico J Garde
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Marcelo G Stinson
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Candela D Pastore
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina
| | - Abel L Carcagno
- Laboratorio de Diferenciación Celular y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina; Departamento de Ecología, Genética y Evolución, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Autónoma de, Buenos Aires, C1428EGA, Argentina.
| |
Collapse
|
26
|
Daya T, Breytenbach A, Gu L, Kaur M. Cholesterol metabolism in pancreatic cancer and associated therapeutic strategies. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159578. [PMID: 39542394 DOI: 10.1016/j.bbalip.2024.159578] [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: 07/24/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Pancreatic cancer remains one of the most lethal cancers due to late diagnosis and high chemoresistance. Despite recent progression in the development of chemotherapies, immunotherapies, and potential nanoparticles-based approaches, the success rate of therapeutic response is limited which is further compounded by cancer drug resistance. Understanding of emerging biological and molecular pathways causative of pancreatic cancer's aggressive and chemoresistance is vital to improve the effectiveness of existing therapeutics and to develop new therapies. One such under-investigated and relatively less explored area of research is documenting the effect that lipids, specifically cholesterol, and its metabolism, impose on pancreatic cancer. Dysregulated cholesterol metabolism has a profound role in supporting cellular proliferation, survival, and promoting chemoresistance and this has been well established in various other cancers. Thus, we aimed to provide an in-depth review focusing on the significance of cholesterol metabolism in pancreatic cancer and relevant genes at play, molecular processes contributing to cellular cholesterol homeostasis, and current research efforts to develop new cholesterol-targeting therapeutics. We highlight the caveats, weigh in different experimental therapeutic strategies, and provide possible suggestions for future research highlighting cholesterol's importance as a therapeutic target against pancreatic cancer resistance and cancer progression.
Collapse
Affiliation(s)
- Tasvi Daya
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Andrea Breytenbach
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Liang Gu
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa.
| |
Collapse
|
27
|
Chiu CC, Hao WR, Lin KJ, Chen CC, Yang TY, Fang YA, Yang TL, Lai YH, Chen MY, Hsu MH, Lin CH, Hsiu H, Chen HY, Cheng TH, Chen NH, Liu JC. Big data analysis of influenza vaccination and liver cancer risk in hypertensive patients: insights from a nationwide population-based cohort study. BMC Gastroenterol 2025; 25:109. [PMID: 39994561 PMCID: PMC11849173 DOI: 10.1186/s12876-025-03665-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 02/01/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND previous studies have indicated that influenza vaccination may be associated with reduced risks of certain types of cancer. However, the protective effect of influenza vaccination against primary liver cancer in individuals with hypertension remains unclear. METHODS In this cohort study, 37,022 patients over 55 years of age who received a diagnosis of hypertension at any time between January 1, 2001, and December 31, 2012, were enrolled from the National Health Insurance Research Database. The patients were divided into a vaccinated and an unvaccinated group. Categorical and continuous variables were analyzed using the chi-square test and t test, respectively, and the correlation between influenza vaccination and liver cancer in patients with hypertension was analyzed using time-varying COX model. Propensity score method was performed to reduce selection bias. RESULTS Compared with the unvaccinated group, the vaccinated group had a significantly lower incidence of liver cancer (hazard ratio = 0.56, 95% confidence interval = 0.46-0.64; p < .001). In addition, a protective effect was observed regardless of sex, age, or comorbidities. Besides, the association was dose-dependent which could be noted when patients were stratified based on the total number of vaccinations. The adjusted HRs for patients receiving 1, 2 to 3, and ≥ 4 vaccinations during the follow-up period were 0.60 (0.51-0.78), 0.48 (0.38-0.65), and 0.39(0.30-0.51), respectively. CONCLUSIONS In summary, influenza vaccination is linked to a decreased risk of liver cancer in individuals with hypertension. However, unmeasurable confounders may have been present in the analysis.
Collapse
Affiliation(s)
- Chun-Chih Chiu
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Wen-Rui Hao
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Kuan-Jie Lin
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chun-Chao Chen
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Tsung-Yeh Yang
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ann Fang
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Lin Yang
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan
| | - Yu-Hsin Lai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei, Taiwan
| | - Ming-Yao Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei, Taiwan
| | - Min-Huei Hsu
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Cheng-Hsin Lin
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Hsin Hsiu
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, No.43, Section 4, Keelung Road, Taipei, 10607, Taiwan
| | - Huan-Yuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11578, Taiwan
| | - Tzu-Hurng Cheng
- Department of Biochemistry, School of Medicine, College of Medicine, China Medical University, Taichung City, 404333, Taiwan
| | - Nai-Hsuan Chen
- Department of Physical medicine and rehabilitation, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Ju-Chi Liu
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan.
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
28
|
Guo L, He Y, Xin H. GDF15 promotes the resistance of epithelial ovarian cancer cells to gemcitabine via DHCR24-mediated cholesterol metabolism to elevate ABCB1 and ABCC1 levels in lipid rafts. Am J Cancer Res 2025; 15:452-469. [PMID: 40084368 PMCID: PMC11897626 DOI: 10.62347/kyiy8286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Growth differentiation factor 15 (GDF15) is upregulated in most cases of epithelial ovarian cancer (EOC); however, its functions in EOC are not fully understood. In this study, we knocked down GDF15 in EOC cells before performing high-throughput sequencing to identify genes regulated by GDF15. GDF15 was overexpressed to determine its effect on the viability, migration, and response of EOC cells to gemcitabine, carboplatin, and paclitaxel. Reprogramming of glucose and cholesterol metabolism in EOC cells was evaluated based on oxygen consumption, lactic acid production, complex I activity, and free and esterified cholesterol levels. The activities of ATP-binding cassette (ABC)B1 and ABCC1 were assessed based on the expulsion efficiency of rhodamine 12. GDF15 overexpression promoted cell viability, migration, and resistance to gemcitabine. In addition, GDF15 induced glycolysis and increased cholesterol levels in EOC cells. Cholesterol metabolism regulated by GDF15 contributed to the resistance of EOC cells to gemcitabine by elevating ABCB1 and ABCC1 levels in lipid rafts. DHCR24 plays an important role in cholesterol synthesis. DHCR24 was identified as a downstream effector of GDF15, because knockdown of DHCR24, but not treatment with statins, suppressed the cancer-promoting effect of GDF15. Overall, GDF15 promoted the resistance of EOC cells to gemcitabine via DHCR24-mediated cholesterol metabolism to elevate ABCB1 and ABCC1 levels in lipid rafts. Therefore, GDF15 and DHCR24 are potential therapeutic targets for suppressing the growth of EOC cells and improving their sensitivity to gemcitabine.
Collapse
Affiliation(s)
- Linlin Guo
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Jinzhou Medical UniversityNo. 2, Section 5, Renmin Street, Guta District, Jinzhou 121001, Liaoning, PR China
| | - Yan He
- Department of Laboratory, Animal Science, China Medical UniversityNo. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, Liaoning, PR China
| | - Huang Xin
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Jinzhou Medical UniversityNo. 2, Section 5, Renmin Street, Guta District, Jinzhou 121001, Liaoning, PR China
| |
Collapse
|
29
|
Martins M, Veiga F, Paiva-Santos AC, Pires PC. Drug Repurposing and Nanotechnology for Topical Skin Cancer Treatment: Redirecting toward Targeted and Synergistic Antitumor Effects. ACS Pharmacol Transl Sci 2025; 8:308-338. [PMID: 39974652 PMCID: PMC11833728 DOI: 10.1021/acsptsci.4c00679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Skin cancer represents a major health concern due to its rising incidence and limited treatment options. Current treatments (surgery, chemotherapy, radiotherapy, immunotherapy, and targeted therapy) often entail high costs, patient inconvenience, significant adverse effects, and limited therapeutic efficacy. The search for novel treatment options is also marked by the high capital investment and extensive development involved in the drug discovery process. In response to these challenges, repurposing existing drugs for topical application and optimizing their delivery through nanotechnology could be the answer. This innovative strategy aims to combine the advantages of the known pharmacological background of commonly used drugs to expedite therapeutic development, with nanosystem-based formulations, which among other advantages allow for improved skin permeation and retention and overall higher therapeutic efficacy and safety. The present review provides a critical analysis of repurposed drugs such as doxycycline, itraconazole, niclosamide, simvastatin, leflunomide, metformin, and celecoxib, formulated into different nanosystems, namely, nanoemulsions and nanoemulgels, nanodispersions, solid lipid nanoparticles, nanostructured lipid carriers, polymeric nanoparticles, hybrid lipid-polymer nanoparticles, hybrid electrospun nanofibrous scaffolds, liposomes and liposomal gels, ethosomes and ethosomal gels, and aspasomes, for improved outcomes in the battle against skin cancer. Enhanced antitumor effects on melanoma and nonmelanoma research models are highlighted, with some nanoparticles even showing intrinsic anticancer properties, leading to synergistic effects. The explored research findings highly evidence the potential of these approaches to complement the currently available therapeutic strategies in the hope that these treatments might one day reach the pharmaceutical market.
Collapse
Affiliation(s)
- Maria Martins
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Patrícia C. Pires
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- RISE-Health,
Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
30
|
Gambaro R, Chain CY, Scioli-Montoto S, Moreno A, Huck-Iriart C, Ruiz ME, Cisneros JS, Lamas DG, Tau J, Gehring S, Islan GA, Rodenak-Kladniew B. Phytoactive-Loaded Lipid Nanocarriers for Simvastatin Delivery: A Drug Repositioning Strategy Against Lung Cancer. Pharmaceutics 2025; 17:255. [PMID: 40006622 PMCID: PMC11858925 DOI: 10.3390/pharmaceutics17020255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Drug repurposing explores new applications for approved medications, such as simvastatin (SV), a lipid-lowering drug that has shown anticancer potential but is limited by solubility and side effects. This study aims to enhance SV delivery and efficacy against lung cancer cells using bioactive lipid nanoparticles formulated with plant-derived monoterpenes as both nanostructuring agents and anticancer molecules. Methods: Lipid nanoparticles were produced by ultrasonication and characterized for morphology, size, zeta potential, and polydispersity index (PDI). Monoterpenes (linalool-LN-, limonene, 1,8-cineole) or Crodamol® were used as liquid lipids. Encapsulation efficiency (EE), release profiles, stability, biocompatibility, protein adsorption, cytotoxicity, and anticancer effects were evaluated. Results: The nanoparticles exhibited high stability, size: 94.2 ± 0.9-144.0 ± 2.6 nm, PDI < 0.3, and zeta potential: -4.5 ± 0.7 to -16.3 ± 0.8 mV. Encapsulation of SV in all formulations enhanced cytotoxicity against A549 lung cancer cells, with NLC/LN/SV showing the highest activity and being chosen for further investigation. Sustained SV release over 72 h and EE > 95% was observed for NLC/LN/SV. SAXS/WAXS analysis revealed that LN altered the crystallographic structure of nanoparticles. NLC/LN/SV demonstrated excellent biocompatibility and developed a thin serum protein corona in vitro. Cellular studies showed efficient uptake by A549 cells, G0/G1 arrest, mitochondrial hyperpolarization, reactive oxygen species production, and enhanced cell death compared to free SV. NLC/LN/SV more effectively inhibited cancer cell migration than free SV. Conclusions: NLC/LN/SV represents a promising nanocarrier for SV repurposing, combining enhanced anticancer activity, biocompatibility, and sustained stability for potential lung cancer therapy.
Collapse
Affiliation(s)
- Rocío Gambaro
- Children’s Hospital, University Medical Center of the Johannes, Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (R.G.); (S.G.)
| | - Cecilia Y. Chain
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), La Plata 1900, Buenos Aires, Argentina; (C.Y.C.); (J.S.C.)
| | - Sebastian Scioli-Montoto
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Buenos Aires, Argentina; (S.S.-M.); (M.E.R.)
| | - Ailin Moreno
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), CCT-La Plata, Facultad de Ciencias Médicas UNLP, La Plata 1900, Buenos Aires, Argentina; (A.M.); (J.T.)
| | - Cristián Huck-Iriart
- Instituto de Tecnologías Emergentes y Ciencias Aplicadas (ITECA), Universidad Nacional de San Martín (UNSAM)--Investigaciones Científicas y Tecnológicas (CONICET), Escuela de Ciencia y Tecnología (ECyT), Laboratorio de Cristalografía Aplicada (LCA), Campus Miguelete, San Martín 1650, Buenos Aires, Argentina; (C.H.-I.); (D.G.L.)
- ALBA Synchrotron Light Source, Carrer de la Llum 2–26, Cerdanyola del Vallès, 08290 Barcelona, Spain
| | - María Esperanza Ruiz
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Buenos Aires, Argentina; (S.S.-M.); (M.E.R.)
| | - José S. Cisneros
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), La Plata 1900, Buenos Aires, Argentina; (C.Y.C.); (J.S.C.)
| | - Diego G. Lamas
- Instituto de Tecnologías Emergentes y Ciencias Aplicadas (ITECA), Universidad Nacional de San Martín (UNSAM)--Investigaciones Científicas y Tecnológicas (CONICET), Escuela de Ciencia y Tecnología (ECyT), Laboratorio de Cristalografía Aplicada (LCA), Campus Miguelete, San Martín 1650, Buenos Aires, Argentina; (C.H.-I.); (D.G.L.)
| | - Julia Tau
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), CCT-La Plata, Facultad de Ciencias Médicas UNLP, La Plata 1900, Buenos Aires, Argentina; (A.M.); (J.T.)
| | - Stephan Gehring
- Children’s Hospital, University Medical Center of the Johannes, Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (R.G.); (S.G.)
| | - Germán A. Islan
- Children’s Hospital, University Medical Center of the Johannes, Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; (R.G.); (S.G.)
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), Laboratorio de Nanobiomateriales, Departamento de Química, Facultad de Ciencias Exactas, Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), CCT-La Plata, La Plata 1900, Buenos Aires, Argentina
| | - Boris Rodenak-Kladniew
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Investigaciones Científicas y Tecnológicas (CONICET)-Universidad Nacional de La Plata (UNLP), CCT-La Plata, Facultad de Ciencias Médicas UNLP, La Plata 1900, Buenos Aires, Argentina; (A.M.); (J.T.)
| |
Collapse
|
31
|
Zhu Z, Wu X, Zhang J, Zhu M, Tian M, Zhao P. Advances in understanding ferroptosis mechanisms and their impact on immune cell regulation and tumour immunotherapy. Discov Oncol 2025; 16:153. [PMID: 39930297 PMCID: PMC11811334 DOI: 10.1007/s12672-025-01911-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Ferroptosis is a novel mode of iron-dependent non-apoptotic cell death that occurs mainly due to excessive accumulation of lipid peroxides. Numerous studies in recent years have shown that ferroptosis plays a vital role in the organism and has important interactions with immune cells. Ferroptosis has been shown to have great potential in tumour therapy through studying its mechanism of action. In addition, ferroptosis plays a major role in many types of tumour cells that can potently suppress the tumourigenesis and metastasis, provide a basis for the treatment of many malignant tumour diseases and become a novel therapeutic modality of antitumour immunity in the clinic. Current tumour immunotherapy for ferroptosis in combination with other conventional oncological modalities is not well elaborated. In this paper, we mainly discuss the connection of ferroptosis with immune cells and their mediated tumour immunotherapy in order to provide a better theoretical basis and new thinking about ferroptosis mediated antitumour immunity.
Collapse
Affiliation(s)
- Zengjun Zhu
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, China
| | - Xuanxuan Wu
- School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, China
| | - Jian Zhang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255036, China
| | - Minghui Zhu
- Department of Clinical Laboratory, Huantai County People's Hospital, Zibo, 256400, China
| | - Maojin Tian
- Department of Critical Care Medicine, Zibo Central Hospital, Zibo, 255036, China.
| | - Peiqing Zhao
- Center of Translational Medicine, Zibo Central Hospital, Zibo, 255036, China.
| |
Collapse
|
32
|
Nedeljković M, Vuletić A, Mirjačić Martinović K. Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:1396. [PMID: 40003864 PMCID: PMC11855393 DOI: 10.3390/ijms26041396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant type of breast cancer with limited treatment options and poor prognosis. One of the most significant impediments in TNBC treatment is the high heterogeneity of this disease, as highlighted by the detection of several molecular subtypes of TNBC. Each subtype is driven by distinct mutations and pathway aberrations, giving rise to specific molecular characteristics closely connected to clinical behavior, outcomes, and drug sensitivity. This review summarizes the knowledge regarding TNBC molecular subtypes and how it can be harnessed to devise tailored treatment strategies instead of blindly using targeted drugs. We provide an overview of novel targeted agents and key insights about new treatment modalities with an emphasis on the androgen receptor signaling pathway, cancer stem cell-associated pathways, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, growth factor signaling, and immunotherapy.
Collapse
Affiliation(s)
- Milica Nedeljković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (A.V.); (K.M.M.)
| | | | | |
Collapse
|
33
|
Abdelrady YA, Thabet HS, Sayed AM. The future of metronomic chemotherapy: experimental and computational approaches of drug repurposing. Pharmacol Rep 2025; 77:1-20. [PMID: 39432183 DOI: 10.1007/s43440-024-00662-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Metronomic chemotherapy (MC), long-term continuous administration of anticancer drugs, is gaining attention as an alternative to the traditional maximum tolerated dose (MTD) chemotherapy. By combining MC with other treatments, the therapeutic efficacy is enhanced while minimizing toxicity. MC employs multiple mechanisms, making it a versatile approach against various cancers. However, drug resistance limits the long-term effectiveness of MC, necessitating ongoing development of anticancer drugs. Traditional drug discovery is lengthy and costly due to processes like target protein identification, virtual screening, lead optimization, and safety and efficacy evaluations. Drug repurposing (DR), which screens FDA-approved drugs for new uses, is emerging as a cost-effective alternative. Both experimental and computational methods, such as protein binding assays, in vitro cytotoxicity tests, structure-based screening, and several types of association analyses (Similarity-Based, Network-Based, and Target Gene), along with retrospective clinical analyses, are employed for virtual screening. This review covers the mechanisms of MC, its application in various cancers, DR strategies, examples of repurposed drugs, and the associated challenges and future directions.
Collapse
Affiliation(s)
- Yousef A Abdelrady
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104, Freiburg, Germany
| | - Hayam S Thabet
- Microbiology Department, Faculty of Veterinary Medicine, Assiut University, Asyut, 71526, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Asyut, 71516, Egypt
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Kingdom of Saudi Arabia
| |
Collapse
|
34
|
Noh JK, Lee MK, Lee Y, Bae M, Min S, Kong M, Lee JW, Kim SI, Lee YC, Ko S, Woo SR, Eun Y. Targeting ferroptosis for improved radiotherapy outcomes in HPV-negative head and neck squamous cell carcinoma. Mol Oncol 2025; 19:540-557. [PMID: 39297393 PMCID: PMC11792990 DOI: 10.1002/1878-0261.13720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/16/2024] [Accepted: 08/01/2024] [Indexed: 02/05/2025] Open
Abstract
To enhance the efficacy of radiotherapy (RT) in human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC), we explored targeting ferroptosis, a regulated cell death process. We developed a gene signature associated with ferroptosis using Cox proportional hazard modeling in HPV-negative HNSCC patients who underwent RT. This ferroptosis-related gene signature (FRGS) was a significant predictor of overall survival and recurrence-free survival in HPV-negative HNSCC patients who received RT. Subtype B of the FRGS, characterized by decreased expression of ferroptosis inducers [nuclear receptor coactivator 4 (NCOA4) and natural resistance-associated macrophage protein 2 homolog/divalent metal transporter 1 (NRAMP2/DMT1)] and increased expression of suppressors [phospholipid hydroperoxide glutathione peroxidase (GPX4) and ferritin heavy chain (FTH1)], was associated with poorer prognosis, potentially indicating the inhibition of ferroptosis. Furthermore, our in vitro and in vivo studies demonstrated that treatment with statins, such as atorvastatin and simvastatin, induced ferroptosis and sensitized radioresistant HNSCC cells to irradiation, improving radiosensitivity and potentially enhancing the response to RT. Additionally, in xenograft models, the combination of statins and RT led to a significant reduction in tumor initiation. These findings provide valuable insights for enhancing treatment and improving prognosis in HPV-negative HNSCC by targeting ferroptosis and utilizing statins to sensitize tumors to RT-induced cell death.
Collapse
Affiliation(s)
- Joo Kyung Noh
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Min Kyeong Lee
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Yeonseo Lee
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Minji Bae
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Soonki Min
- Department of Radiation OncologyKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Moonkyoo Kong
- Department of Radiation OncologyKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Jung Woo Lee
- Department of Oral and Maxillofacial Surgery, School of DentistryKyung Hee UniversitySeoulKorea
| | - Su Il Kim
- Department of Otolaryngology‐Head and Neck SurgeryKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Young Chan Lee
- Department of Otolaryngology‐Head and Neck SurgeryKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Seong‐Gyu Ko
- Department of Preventive Medicine, College of Korean MedicineKyung Hee UniversitySeoulKorea
| | - Seon Rang Woo
- Department of Otolaryngology‐Head and Neck SurgeryKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Young‐Gyu Eun
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
- Department of Otolaryngology‐Head and Neck SurgeryKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| |
Collapse
|
35
|
Codenotti S, Asperti M, Poli M, Lorenzi L, Pietrantoni A, Cassandri M, Marampon F, Fanzani A. Synthetic inhibition of SREBP2 and the mevalonate pathway blocks rhabdomyosarcoma tumor growth in vitro and in vivo and promotes chemosensitization. Mol Metab 2025; 92:102085. [PMID: 39706565 PMCID: PMC11750561 DOI: 10.1016/j.molmet.2024.102085] [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: 09/26/2024] [Revised: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024] Open
Abstract
OBJECTIVE The aim of the present study was to investigate the effects of targeting the mevalonate pathway (MVP) in rhabdomyosarcoma (RMS), a soft tissue tumor with a prevalence in young people. METHODS In silico analyses of RNA datasets were performed to correlate MVP with RMS patient survival. The sensitivity of RMS cell lines to MVP inhibitors was assessed in vitro by analysis of cell growth (crystal violet and clonogenic assays), cell migration (wound healing assay), cell survival (neutral red assay), and oxidative stress (ROS assay). The effects of MVP inhibitors were tested in vivo by analyzing RMS xenografts grown in NOD/SCID mice. Quantification of protein targets was performed using immunoblotting or immunohistochemistry analyses. RESULTS In silico analysis showed upregulation of sterol regulatory element-binding protein 2 (SREBP2) and MVP genes, including 3-Hydroxy-3-Methylglutaryl-CoA Reductase (HMGCR), farnesyl-diphosphate synthase (FDPS), squalene epoxidase (SQLE), which correlated with worse overall patient survival. Targeting of MVP in human RD and RH30 lines by inhibitors of SREBP2 (fatostatin), HMGCR (lovastatin and simvastatin), and FDPS (zoledronic acid) resulted in impaired cell growth, migration, and viability, and increased oxidative cell death in combination with actinomycin D. Conversely, cholesterol (CHO) supplementation enhanced cell growth and migration. Fatostatin and lovastatin produced rapid attenuation of Erk1/2 and Akt1 signaling in RMS lines, and oral administration of lovastatin reduced tumor mass growth of xenografted RD cells in NOD/SCID mice. Finally, we found that forced Akt1 activation in RD cells was sufficient to drive SREBP2, HMGCR and SQLE protein expression, promoting increased susceptibility to MVP inhibitors. CONCLUSIONS These data suggest that the Akt1, SREBP2 and MVP axis is critical for RMS tumor growth, migration, and oxidative stress protection primarily through maintaining adequate CHO levels that enable proper intracellular signaling. Therefore, stimulating CHO depletion via SREBP2 and MVP inhibition may represent a viable option to improve the combination therapy protocol, especially in pAkt1-positive RMS.
Collapse
Affiliation(s)
- Silvia Codenotti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Michela Asperti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luisa Lorenzi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Alberto Pietrantoni
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; ASST Spedali Civili di Brescia, 25123, Brescia, Italy
| | - Matteo Cassandri
- Department of Radiological Sciences, Oncology and Anatomic Pathology, "Sapienza" University of Rome, 00161, Rome, Italy
| | | | - Alessandro Fanzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
36
|
Jain A, Das R, Giri M, Mane P, Shard A. Carbohydrate kinase inhibition: a promising strategy in cancer treatment. Drug Discov Today 2025; 30:104308. [PMID: 39912130 DOI: 10.1016/j.drudis.2025.104308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025]
Abstract
Carbohydrate kinases (CKs) are pivotal in various biological processes, including energy consumption, cell signaling, and biosynthesis. They are a group of enzymes that facilitate the phosphorylation of carbohydrates, playing a crucial role in cellular metabolism. These enzymes facilitate the transfer of a phosphate group from a high-energy donor like ATP to a specified location on a carbohydrate substrate. Dysregulated kinase activity drives tumor growth and progression. Inhibitors targeting these enzymes have been developed and used in cancer therapy. The CK family encompasses three major types: hexokinases, ribokinases, and phosphatidylinositol kinases, with inhibitors of paramount importance in cancer treatment. This review explores the role of CKs in cancer and its inhibitors, providing insights into improving existing inhibitors and designing new ones.
Collapse
Affiliation(s)
- Archit Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Muskan Giri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Pranita Mane
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
37
|
Wu X, Li Y, Wang W, Xu J, Zhao B, Sun W, Ge D, Xiong L, Dou X, Zou X, Wang L, Chen M. DRAM1 enhances the proliferation and metastasis of gastric cancer through the PI3K/AKT/mTOR signaling pathway and energy metabolism. Sci Rep 2025; 15:3542. [PMID: 39875526 PMCID: PMC11775094 DOI: 10.1038/s41598-025-87389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
Gastric cancer (GC) is a prevalent malignant tumor of the digestive system that is often diagnosed at advanced stages owing to inconspicuous early symptoms and a lack of specific examination methods. Effective treatment of advanced stages remains challenging, emphasizing the need for new therapeutic targets. Metabolic reprogramming, a hallmark of tumors, plays a pivotal role in tumor progression, immune evasion, and immune surveillance. DNA damage-regulated autophagy modulator 1 (DRAM1) encodes a hexameric transmembrane protein that is predominantly located in lysosomes and induces autophagy; however, its mechanism of action in gastric cancer remains unclear. Our study found that elevated DRAM1 expression in patients with GC correlated with survival and prognosis. DRAM1 knockdown suppressed energy metabolism in GC cells through the PI3K/AKT/mTOR signaling pathway, thereby mitigating GC progression. Atorvastatin, a focus of recent tumor research, significantly enhanced apoptosis levels in DRAM1 knockdown GC cells compared to the control group. Therefore, through metabolic reprogramming, DRAM1 may serve as a potential therapeutic target for GC prevention.
Collapse
Affiliation(s)
- Xinrong Wu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University of Chinese Medicine, Nanjing, 210000, Jiangsu, China
| | - Yifan Li
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University of Chinese Medicine, Nanjing, 210000, Jiangsu, China
| | - Weiwei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University of Chinese Medicine, Nanjing, 210000, Jiangsu, China
| | - Jiale Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University of Chinese Medicine, Nanjing, 210000, Jiangsu, China
| | - Bei Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, 210000, China
| | - Wenqi Sun
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, 210000, China
| | - Dan Ge
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, 210000, China
| | - Longying Xiong
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, 210000, China
| | - Xiaotan Dou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, 210000, China
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, 210000, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, 210000, China.
| | - Min Chen
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University of Chinese Medicine, Nanjing, 210000, Jiangsu, China.
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Jiangsu, 210000, China.
| |
Collapse
|
38
|
HARISA GAMALELDINI, ALZHRANI RIYADF, ALLUHAIDAN ABDULRAHMANA, ALAMRI SULTANM, BAKHEIT AHMEDH, ASIRI HANADIH, ATTIA SABRYM. Chitosan capped-NLCs enhanced codelivery of gefitinib and simvastatin into MDR HCC: impact of compositions on cell death, JNK3, and Telomerase. Oncol Res 2025; 33:477-492. [PMID: 39866231 PMCID: PMC11754001 DOI: 10.32604/or.2024.053337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/08/2024] [Indexed: 01/28/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a health problem due to multi-drug resistance (MDR). Codelivery of multiple oncotherapy in one cargo as chimeric cancer therapy (CCT) is suggested as a solution for MDR. This study aims to engineer chitosan-coated nanostructure lipid carriers (NLCs) loaded with gefitinib (GF) and simvastatin (SV) as CCT for HCC. Methods Both GF and SV-loaded nanostructure lipids carriers (GFSVNLC) and chitosan-capped GF and SV-loaded nanostructure lipids carriers (CGFSVNLC) formulations were assembled by top-down techniques. Moreover, particle size (PS), zeta potential (ZP), and polydispersity index (PDI) were measured by Zetasizer. The biosafety of GFSVNLC preparations was investigated by using erythrocytes as a biological model. The cytotoxic, and apoptotic effects of the prepared GFSVNLCs were investigated using HepG2 cell lines as a substitute model for HCC. The effect of GF, SV, and NLC composition on JNK3, HDAC6, and telomerase was studied using molecular docking simulation (MDS). Results The present results revealed that the obtained GFSVNLC and CGFSVNLC have nanosized and consistent, CS coating shifts anionic ZP of GFSVNLC into CGFSVNLC with cationic ZP. Moreover, both formulations are biocompatible as indicated by their gentle effect on erythrocyte hemolysis. The treatment of HepG2 cells with GFSVNLC, and CGFSVNLC induced marked cell death compared to other groups with a decrease of IC50. Equally, the percentage of the apoptotic HepG2 cells was increased upon treatment of the cells with GFSV, GFSVNLC, and CGFSVNLC compared to the control group. Additionally, GF, SV, stearic acid (SA), and oleic acid (OA) modulate the activity of JNK3, HDAC6, and telomerase. Conclusions This study suggests CGFSVNLC achieves codelivery, selective targeting, and enhancing the synergistic effect of GF and SV for inducing HepG2 cell death. Mechanistically, CGFSVNLC inhibits key cascades implicated in MDR and HepG2 cell survival. CGFSVNLC is promising for overcoming drug resistance mechanisms and improving therapeutic outcomes against HepG2 cells.
Collapse
Affiliation(s)
- GAMALELDIN I. HARISA
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - RIYAD F. ALZHRANI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | | | - SULTAN M. ALAMRI
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - AHMED H. BAKHEIT
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - HANADI H. ASIRI
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - SABRY M. ATTIA
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
39
|
Biscetti F, Polito G, Rando MM, Nicolazzi MA, Eraso LH, DiMuzio PJ, Massetti M, Gasbarrini A, Flex A. Residual Traditional Risk in Non-Traditional Atherosclerotic Diseases. Int J Mol Sci 2025; 26:535. [PMID: 39859250 PMCID: PMC11765428 DOI: 10.3390/ijms26020535] [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: 01/05/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Individuals with chronic inflammatory and immune disorders are at an increased risk of atherosclerotic events and premature cardiovascular (CV) disease. Despite extensive literature exploring the relationship between "non-traditional" atherosclerotic conditions and CV risk, many aspects remain unresolved, including the underlying mechanisms promoting the "non-traditional CV risk", the development of an innovative and comprehensive CV risk assessment tool, and recommendations for tailored interventions. This review aims to evaluate the available evidence on key "non-traditional" CV risk-enhancer conditions, with a focus on assessing and managing CV risk factors. We conducted a comprehensive review of 412 original articles, narrative and systematic reviews, and meta-analyses addressing the CV risk associated with "non-traditional" atherosclerotic conditions. The analysis examined the underlying mechanisms of these relationships and identified strategies for assessing and mitigating elevated risk. A major challenge highlighted is the difficulty in quantifying the contribution of individual risk factors and disease-specific elements to CV risk. While evidence supports the cardiovascular benefits of statins beyond lipid lowering, such as pleiotropic and endothelial effects, current guidelines lack specific recommendations for the use of statins or other therapies targeting non-traditional CV risk factors. Additionally, the absence of validated cardiovascular risk scores that incorporate non-traditional risk factors hinders accurate CV risk evaluation and management. The growing prevalence of "non-traditional CV risk-enhancer conditions" underscores the need for improved awareness of CV risk assessment and management. A thorough understanding of all contributing factors, including disease-specific elements, is crucial for accurate prediction of cardiovascular disease (CVD) risk. This represents an essential foundation for informed decision-making in primary and secondary prevention. We advocate for future research to focus on developing innovative, disease-specific CV risk assessment tools that incorporate non-traditional risk factors, recognizing this as a promising avenue for translational and clinical outcome research.
Collapse
Affiliation(s)
- Federico Biscetti
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Giorgia Polito
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Maria Margherita Rando
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Maria Anna Nicolazzi
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Luis H. Eraso
- Division of Vascular and Endovascular Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Paul J. DiMuzio
- Division of Vascular and Endovascular Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Massimo Massetti
- Dipartimento di Scienze Cardiovascolari e Pneumologiche, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Andrea Flex
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
40
|
Turpo-Peqqueña AG, Luna-Prado S, Valencia-Arce RJ, Del-Carpio-Carrazco FL, Gómez B. A Theoretical Study on the Efficacy and Mechanism of Combined YAP-1 and PARP-1 Inhibitors in the Treatment of Glioblastoma Multiforme Using Peruvian Maca Lepidium meyenii. Curr Issues Mol Biol 2025; 47:40. [PMID: 39852155 PMCID: PMC11763394 DOI: 10.3390/cimb47010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/25/2024] [Accepted: 01/05/2025] [Indexed: 01/26/2025] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive and treatment-resistant forms of brain cancer. Current therapeutic strategies, including surgery, chemotherapy, and radiotherapy, often fail due to the tumor's ability to develop resistance. The proteins YAP-1 (Yes-associated protein 1) and PARP-1 (Poly-(ADP-ribose)-polymerase-1) have been implicated in this resistance, playing crucial roles in cell proliferation and DNA repair mechanisms, respectively. This study explored the inhibitory potential of natural compounds from Lepidium meyenii (Peruvian Maca) on the YAP-1 and PARP-1 protein systems to develop novel therapeutic strategies for GBM. By molecular dynamics simulations, we identified N-(3-Methoxybenzyl)-(9Z,12Z,15Z)- octadecatrienamide (DK5) as the most promising natural inhibitor for PARP-1 and stearic acid (GK4) for YAP-1. Although synthetic inhibitors, such as Olaparib (ODK) for PARP-1 and Verteporfin (VER) for YAP-1, only VER was superior to the naturally occurring molecule and proved a promising alternative. In conclusion, natural compounds from Lepidium meyenii (Peruvian Maca) offer a potentially innovative approach to improve GBM treatment, complementing existing therapies with their inhibitory action on PARP-1 and YAP-1.
Collapse
Affiliation(s)
- Albert Gabriel Turpo-Peqqueña
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Medicina Humana, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
- Facultad de Biología, Universidad Nacional de San Agustín, Av. Alcides Carrión s/n, Arequipa 04001, Peru
| | - Sebastian Luna-Prado
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
| | - Renato Javier Valencia-Arce
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
| | - Fabio Leonardo Del-Carpio-Carrazco
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
| | - Badhin Gómez
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
| |
Collapse
|
41
|
Deng H, Zhou J, Liu Z, Huang L, Gu Y, Chen P, Xiao H. Concomitant medication effects on patients with lung cancer taking immune checkpoint inhibitors a review. Med Oncol 2025; 42:40. [PMID: 39762456 DOI: 10.1007/s12032-024-02591-3] [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: 10/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
In the past decade, a variety of immune checkpoint inhibitors (ICIs) are currently approved for lung cancer in the world. As a new therapeutic approach, ICIs have shown significant clinical benefits in the first-line or second-line treatment for advanced lung cancer, improving the survival and quality of life of patients. Patients need to take multiple drugs in the meantime due to their own disease or side effects during treatment. In view of drug interactions, concomitant medications have a positive or negative impact on the prognosis of lung cancer patients. In this review, we reviewed the effects of multiple drugs on the prognosis of patients with lung cancer taking ICIs. Several studies indicate that antibiotics, proton pump inhibitors (PPIs), corticosteroids, and opioid analgesics can decrease the efficacy of ICIs. Aspirin and bone-targeting drugs can enhance the efficacy of ICIs and improve the survival rate. The effects of metformin (MET), renin-angiotensin-aldosterone system inhibitors (RASI), nonsteroidal anti-inflammatory drug (NSAIDS) (except aspirin), and statins on ICIs are controversial. Future research should further explore the effects of these concomitant medications on ICIs and develop personalized prescriptions based on the specific needs of patients.
Collapse
Affiliation(s)
- Han Deng
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Junxiang Zhou
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Zhixi Liu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Lu Huang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Yanru Gu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Peng Chen
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Hongtao Xiao
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China.
| |
Collapse
|
42
|
Wang B, Huang S, Li S, Deng Y, Li Z, Wang Y, Shi X, Zhang W, Shi L, Wang X, Tang X. Hepatotoxicity of statins: a real-world study based on the US Food and Drug Administration Adverse Event Reporting System database. Front Pharmacol 2025; 15:1502791. [PMID: 39840096 PMCID: PMC11747658 DOI: 10.3389/fphar.2024.1502791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/13/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Statins, as an important class of lipid-lowering drugs, play a key role in the prevention and treatment of cardiovascular diseases. However, with their widespread use in clinical practice, some adverse events have gradually emerged. In particular, the hepatotoxicity associated with statins use has become one of the clinical concerns that require sufficient attention. METHODS In this study, we conducted a comprehensive and detailed analysis of the hepatotoxicity of statins based on the data of the US Food and Drug Administration Adverse Event Reporting System database from the first quarter (Q1) of 2004 to the Q1 of 2024 and used Reporting Odds Ratios and Empirical Bayes Geometric Mean to mine the signal of adverse events. RESULTS In this study, hepatic disorder related seven statins all exhibited positive signals. Through signal mining, we identified a total of 14,511 cases of adverse events associated with hepatic disorder caused by these statin drugs, with atorvastatin, simvastatin, and rosuvastatin occurring at a higher rate. A total of 148 positive signals related to adverse events of hepatic disorder were captured. Autoimmune hepatitis and drug-induced liver injury both presented positive signals across multiple statin drugs. Notably, atorvastatin had the most significant signal strength in cholestatic pruritus and bilirubin conjugation abnormal. Fluvastatin also showed notable signal strength in autoimmune hepatitis, while simvastatin had a relatively weaker signal strength for hepatic enzyme increased. CONCLUSION This study discovered specific adverse event signal values, revealing potential hepatotoxic risks associated with the use of statin drugs. The results provide an important reference for the safe clinical use of drugs, help to improve the understanding of the safety of statins, and also provide a scientific basis for clinicians to make more accurate and safe decisions when making treatment plans.
Collapse
Affiliation(s)
- Bojing Wang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shu Huang
- Department of Gastroenterology, Lianshui County People’ Hospital, Huaian, China
- Department of Gastroenterology, Lianshui People’ Hospital of Kangda College Affiliated to Nanjing Medical University, Huaian, China
| | - Shiqi Li
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yaqi Deng
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ziyan Li
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yizhou Wang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaomin Shi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Zhang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lei Shi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaohong Wang
- Department of Gastroenterology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Xiaowei Tang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
43
|
Jiang M, Hong C, Zou W, Ye Z, Lu L, Liu Y, Zhang T, Ding Y. Recent advances in the anti-tumor activities of saponins through cholesterol regulation. Front Pharmacol 2025; 15:1469392. [PMID: 39845802 PMCID: PMC11752913 DOI: 10.3389/fphar.2024.1469392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025] Open
Abstract
Abnormal cholesterol metabolism has become a popular therapeutic target in cancer therapy. In recent years there has been a surge in interest in the anti-tumor activities of saponins, particularly their ability to disrupt cholesterol homeostasis in tumor cells. Cholesterol regulation by saponins is a complex process that involves multiple mechanisms. However, there are now a notable dearth of comprehensive reviews addressing their anti-tumor effects through cholesterol modulation. This review will explore the intricate mechanisms by which saponins regulate cholesterol, including modulation of synthesis, metabolism, and uptake, as well as complex formation with cholesterol. It will also outline how saponins exert their anti-cancer activities through cholesterol regulation, enhancing cytotoxicity, inhibiting tumor cell metastasis, reversing drug resistance, inducing immunotoxin macromolecule escape, and ferroptosis. This comprehensive analysis offers insights into the potential for the use of saponins anti-tumor therapies and their combinations with other drugs, advancing the understanding of their effects on cancer cells.
Collapse
Affiliation(s)
- Min Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Hong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenkui Zou
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheng Ye
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Lu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yun Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- National Innovation Platform for Medical Industry-Education Integration, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
44
|
Gallucci G, Larocca M, Navazio A, Turazza FM, Inno A, Canale ML, Oliva S, Besutti G, Tedeschi A, Aschieri D, Russo A, Gori S, Silvestris N, Pinto C, Tarantini L. Atherosclerosis and the Bidirectional Relationship Between Cancer and Cardiovascular Disease: From Bench to Bedside, Part 2 Management. Int J Mol Sci 2025; 26:334. [PMID: 39796190 PMCID: PMC11719480 DOI: 10.3390/ijms26010334] [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: 11/16/2024] [Revised: 12/25/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
The first part of this review highlighted the evolving landscape of atherosclerosis, noting emerging cardiometabolic risk factors, the growing impact of exposomes, and social determinants of health. The prominent role of atherosclerosis in the bidirectional relationship between cardiovascular disease and cancer was also discussed. In this second part, we examine the complex interplay between multimorbid cardio-oncologic patients, cardiometabolic risk factors, and the harmful environments that lend a "syndemic" nature to these chronic diseases. We summarize management strategies targeting disordered cardiometabolic factors to mitigate cardiovascular disease and explore molecular mechanisms enabling more tailored therapies. Importantly, we emphasize the early interception of atherosclerosis through multifactorial interventions that detect subclinical signs (via biomarkers and imaging) to treat modifiable risk factors and prevent clinical events. A concerted preventive effort-referred to by some as a "preventome"-is essential to reduce the burden of atherosclerosis-driven chronic diseases, shifting from mere chronic disease management to the proactive promotion of "chronic health".
Collapse
Affiliation(s)
| | - Mario Larocca
- Provincial Medical Oncology, Department of Oncology and Advanced Technologies, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy; (M.L.); (C.P.)
| | - Alessandro Navazio
- Cardiologia Ospedaliera, Department of Specialized Medicine, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| | | | - Alessandro Inno
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy; (A.I.)
| | - Maria Laura Canale
- Division of Cardiology, Azienda USL Toscana Nord-Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy;
| | - Stefano Oliva
- UOSD Cardiologia di Interesse Oncologico IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Giulia Besutti
- Radiology Unit, Department of Imaging and Laboratory Medicine, AUSL—IRCCS di Reggio Emilia, 42100 Reggio Emilia, Italy;
- Department of Surgical and Medical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Andrea Tedeschi
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29100 Piacenza, Italy; (A.T.); (D.A.)
| | - Daniela Aschieri
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29100 Piacenza, Italy; (A.T.); (D.A.)
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy;
| | - Stefania Gori
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy; (A.I.)
| | - Nicola Silvestris
- Medical Oncology Department, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Carmine Pinto
- Provincial Medical Oncology, Department of Oncology and Advanced Technologies, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy; (M.L.); (C.P.)
| | - Luigi Tarantini
- Cardiologia Ospedaliera, Department of Specialized Medicine, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| |
Collapse
|
45
|
Maurya SK, Chaudhri S, Kumar S, Gupta S. Repurposing of Metabolic Drugs Metformin and Simvastatin as an Emerging Class of Cancer Therapeutics. Pharm Res 2025; 42:49-67. [PMID: 39775614 DOI: 10.1007/s11095-024-03811-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Metabolic alterations are commonly associated with various cancers and are recognized as contributing factors to cancer progression, invasion, and metastasis. Drug repurposing, a strategy in drug discovery, utilizes existing knowledge to recommend established drugs for new indications based on clinical data or biological evidence. This approach is considered a less risky alternative to traditional drug development. Metformin, a biguanide, is a product of Galega officinalis (French lilac) primarily prescribed for managing type 2 diabetes, is recognized for its ability to reduce hepatic glucose production and enhance insulin sensitivity, particularly in peripheral tissues such as muscle. It also improves glucose uptake and utilization while decreasing intestinal glucose absorption. Statins, first isolated from the fungus Penicillium citrinum is another class of medication mainly used to lower cholesterol levels in individuals at risk for cardiovascular diseases, work by inhibiting the enzyme 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, which is essential for cholesterol biosynthesis in the liver. Metformin is frequently used in conjunction with statins to investigate their potential synergistic effects. Combination of metformin and simvastatin has gathered much attention in cancer research because of its potential advantages for cancer prevention and treatment. In this review, we analyze the effects of metformin and simvastatin, both individually and in combination, on key cancer hallmarks, and how this combination affects the expression of biomolecules and associated signaling pathways. We also summarize preclinical research, including clinical trials, on the efficacy, safety, and potential applications of repurposing metformin and simvastatin for cancer therapy.
Collapse
Affiliation(s)
- Santosh Kumar Maurya
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Smriti Chaudhri
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, 151401, Punjab, India.
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA.
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
46
|
Zhang H, Rutkowska A, González-Martín A, Mirza MR, Monk BJ, Vergote I, Pothuri B, Graybill WAS, Goessel C, Barbash O, Bergamini G, Feng B. Potential Synergistic Effect between Niraparib and Statins in Ovarian Cancer Clinical Trials. CANCER RESEARCH COMMUNICATIONS 2025; 5:178-186. [PMID: 39636225 PMCID: PMC11775730 DOI: 10.1158/2767-9764.crc-24-0191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/17/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
SIGNIFICANCE The presented retrospective analysis suggests, to the best of our knowledge for the first time, a potential significant interaction between statins and niraparib in clinical settings. Nevertheless, further investigations are required to gain a better understanding of the potential clinical benefit.
Collapse
Affiliation(s)
- Hailei Zhang
- Precision Medicine, R&D, GSK, Waltham, Massachusetts
| | | | - Antonio González-Martín
- Medical Oncology Department, Clínica Universidad de Navarra, Madrid, Spain
- Center for Applied Medical Research (CIMA), Pamplona, Spain
- Grupo Español de Investigación en Cáncer de Ovario (GEICO), Madrid, Spain
| | - Mansoor R. Mirza
- Nordic Society of Gynaecological Oncology (NSGO) and Department of Oncology Rigshospitalet–Copenhagen University Hospital, Copenhagen, Denmark
| | - Bradley J. Monk
- Florida Cancer Specialists and Research Institute, West Palm Beach, Florida
| | - Ignace Vergote
- Gynecologic Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Bhavana Pothuri
- Department of Obstetrics and Gynecology, NYU School of Medicine, New York City, New York
| | | | | | - Olena Barbash
- Precision Medicine, GSK, Collegeville, Pennsylvania, Philadelphia
| | | | - Bin Feng
- Precision Medicine, R&D, GSK, Waltham, Massachusetts
| |
Collapse
|
47
|
Basso J, Matos AM, Ghavami S, Fortuna A, Vitorino R, Vitorino C. Are we better together? Addressing a combined treatment of pitavastatin and temozolomide for brain cancer. Eur J Pharmacol 2024; 985:177087. [PMID: 39491742 DOI: 10.1016/j.ejphar.2024.177087] [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: 09/13/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Pitavastatin is commonly prescribed to treat hypercholesterolemia through the regulation of cholesterol biosynthesis. Interestingly, it has also demonstrated a great potential for treating brain tumors, although the detailed cytotoxic mechanism, particularly in glioblastoma, remains incompletely understood. This work explores the activity of pitavastatin in 2D and 3D glioblastoma models, in an attempt to provide a more representative and robust overview of its anticancer potential in glioblastoma. The results show that not only is pitavastatin 10-1000 times-fold more effective in reducing tumoral metabolic activity than temozolomide, but also demonstrate a synergistic activity with this alkylating drug. In addition, low micromolar concentrations of this statin strongly impair the growth and the invasion ability of multicellular tumor spheroids. The obtained qRT-PCR and proteomics data highlight the modulation of cell death via apoptosis (BAX/BCL2, CASP9) and autophagy (BECN1, BNIP3, BNIP3L and LC3B), as well as an epithelial to mesenchymal transition blockage (HTRA1, SERPINE1, WNT5A, ALDH3B1 and EPHA2) and remodeling of the extracellular matrix (VCAN, SERPINE1 and TGFBI). Overall, these results lay the foundation for further investigations on the potential combinatory clinical treatment with temozolomide.
Collapse
Affiliation(s)
- João Basso
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Ana Miguel Matos
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Chemical Engineering and Renewable Resources for Sustainability, CERES, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB, R3E 0J9, Canada; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB, R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Ana Fortuna
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal; UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences-IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
48
|
Li ZZ, Zhou K, Wu Q, Liu B, Bu LL. Lymph node metastasis in cancer: Clearing the clouds to see the dawn. Crit Rev Oncol Hematol 2024; 204:104536. [PMID: 39426554 DOI: 10.1016/j.critrevonc.2024.104536] [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: 05/22/2024] [Revised: 09/26/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024] Open
Abstract
Lymph node metastasis (LNM) is often regarded as an indicator of poor prognosis in various cancers. Despite over three centuries of exploration since its discovery, the molecular mechanisms underlying LNM remain inconclusive. This review summarizes the molecular mechanisms of LNM, using the "PUMP+" principle for clarification. Pathological examination remains the gold standard for LNM diagnosis, yet there is a need to explore early diagnostic strategies that can effectively improve patient outcomes. With the advent of immunotherapy, discussions on the fate of lymph nodes (LN) have emerged, emphasizing the importance of preserving LN integrity prior to immunotherapy. This, in turn, poses higher demands for diagnostic accuracy and precision treatment of LNM. This review comprehensively discusses the molecular mechanisms, diagnostic methods, and treatment strategies for cancer lymph node metastasis, along with current bottlenecks and future directions in this field.
Collapse
Affiliation(s)
- Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
49
|
Kato C, Iizuka-Ohashi M, Honda M, Konishi E, Yokota I, Boku S, Mizuta N, Morita M, Sakaguchi K, Taguchi T, Watanabe M, Naoi Y. Additional statin treatment enhances the efficacy of HER2 blockade and improves prognosis in Rac1-high/HER2-positive breast cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167458. [PMID: 39128642 DOI: 10.1016/j.bbadis.2024.167458] [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: 01/27/2024] [Revised: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024]
Abstract
The prognosis of HER2-positive breast cancer (BC) has improved with the development of anti-HER2 therapies; however, the problem remains that there are still cases where anti-HER2 therapies do not respond well. We found that the expression of SREBF2, a master transcriptional factor in the mevalonate pathway, was correlated with ERBB2 (HER2) expression and a poor prognosis in HER2-positive BC. The target gene expressions of SREBF2 were associated with higher expression of ERBB2 in HER2-positive BC cells. Statins, anti-hypercholesterolemia drugs that inhibit the mevalonate pathway, enhanced the efficacy of HER2-targeting agents with inducing apoptosis in a geranylgeranylation-dependent manner. Mechanistically, statins specifically inhibited membrane localization of Rac1, a target protein of geranylgeranylation, and suppressed the activation of HER2 downstreams AKT and ERK pathways. Consistently, retrospective analysis showed a longer recurrence-free survival in Rac1-high/HER2-positive BC patients treated with HER2-targeting agents with statins than without statins. Our findings thus suggest that Rac1 expression could be used as a biomarker to stratify HER2-positive BC patients that could benefit from dual blockade, i.e., targeting HER2 with inhibition of geranylgeranylation of Rac1 using statins, thereby opening avenues for precision medicine in a new subset of Rac1-high/HER2-positive BC.
Collapse
Affiliation(s)
- Chikage Kato
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mahiro Iizuka-Ohashi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mizuki Honda
- Department of Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Isao Yokota
- Department of Biostatistics, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University Hospital, Osaka, Japan
| | | | - Midori Morita
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Sakaguchi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Taguchi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Motoki Watanabe
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Yasuto Naoi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
50
|
Kobayashi K, Murakami K, Baba K. Effects of Lipophilic Statins on Cell Viability and Tissue Factor Expression in Canine Haemangiosarcoma Cells. Vet Comp Oncol 2024; 22:581-591. [PMID: 39319370 DOI: 10.1111/vco.13012] [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: 05/27/2024] [Revised: 08/18/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024]
Abstract
Canine haemangiosarcoma (HSA) is a highly aggressive cancer often associated with coagulation abnormalities. Statins, inhibitors of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) clinically prescribed for hypercholesterolemia, are also believed to possess antitumour and anticoagulant properties by inhibiting downstream Akt activation. Akt phosphorylation is involved in the mechanism of the antitumour and tissue factor (TF)-lowering effects of statins. In the present study, we aimed to investigate whether statins could inhibit cell viability while concurrently inducing anticoagulant properties by regulating the expression of TFs in canine HSA cells. Using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), we initially exclusively detected HMGCR mRNA expression in canine HSA tissues and cell lines but not in normal cephalic vein and spleen tissues. Moreover, treatment with lipophilic statins, including atorvastatin, fluvastatin, and simvastatin, inhibited cell viability in a concentration-dependent manner and decreased TF expression both at the mRNA and protein levels, as evidenced by cell viability assays, RT-qPCR, and immunoblotting, respectively. Further investigation using cell viability assays and flow cytometry revealed that simvastatin decreased Akt phosphorylation, and MK-2206, a specific Akt inhibitor, mirrored the effect of simvastatin on cell viability and cell cycle arrest. However, MK-2206 exhibited different effects on TF expression depending on the cell type, indicating that Akt phosphorylation may not consistently regulate TF expression. Overall, this study provides insights into the potential therapeutic use of statins in targeting tumour growth and coagulation abnormalities in canine HSA. Further research is warranted to fully elucidate the underlying mechanisms and clinical applications of statins in canine HSA treatment.
Collapse
Affiliation(s)
- Kosuke Kobayashi
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Kohei Murakami
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Kenji Baba
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|