1
|
Li L, Zhang J, Li H, Qin L, Wu H, Li Z, Cai L, Chen D, Yang J, Chen Y, Xie Y. Targeted inhibition of JMJD2C/MALAT1 axis compensates for the deficiency of metformin in reversing ovarian cancer platinum resistance. Life Sci 2025; 373:123663. [PMID: 40280301 DOI: 10.1016/j.lfs.2025.123663] [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: 11/25/2024] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
AIMS We explored JMJD2C's role in platinum resistance in ovarian cancer and its modulation by metformin to propose strategies for overcoming treatment limitations. MATERIALS AND METHODS JMJD2C and MALAT1 expression was assessed via RT-qPCR, western blotting, and immunohistochemical assays using OC cell lines, tissue from OC patients, and xenograft treatment with or without metformin. CCK-8 assays, flow cytometry, inductively coupled plasma mass spectrometry, luciferase reporter assays, and ChIP assays were employed to evaluate the impact of JMJD2C/MALAT1 on PR and the effects of metformin on JMJD2C. The effects of metformin in combination with JMJD2C knockdown were assessed in vitro and in vivo. KEY FINDINGS JMJD2C and MALAT1 expression was higher in tissue samples from platinum-resistant phase compared to those from paired platinum-sensitive phase. JMJD2C upregulated MALAT1 in platinum-resistant ovarian cancer (PROC) cells by demethylating its promoter at sites H3K9m3 and H3K36m3. Overexpression of JMJD2C or MALAT1 promoted PR by activating NF-κB/P-gp and P38 MAPK/ERCC1 signaling pathways, with their knockdown produced the opposite effect. Metformin increased JMJD2C expression in tumor tissue, cell lines, and a xenograft model of OC; however, elevated JMJD2C expression attenuated the PR-reversal efficacy of low-concentration metformin. Low-dose metformin combined with JMJD2C-knockdown effectively reversed PR both in in vitro and in vivo, achieving better results than either treatment alone. SIGNIFICANCE JMJD2C drives PR in OC by demethylating the MALAT1 promoter. Metformin upregulated JMJD2C expression, thus necessitating a higher effective dosage of metformin. Targeted inhibition of JMJD2C synergistically enhanced the efficacy of low-dose metformin in overcoming PR, thus providing a promising approach for addressing PR.
Collapse
Affiliation(s)
- Linlin Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China; Academy of Medical Sciences, Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China
| | - Jialin Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China
| | - Huiqing Li
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China
| | - Liying Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China
| | - Han Wu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China
| | - Zijiao Li
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China
| | - Lei Cai
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China
| | - Di Chen
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, and School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Jianping Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China
| | - Yibing Chen
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China.
| | - Ya Xie
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, PR China.
| |
Collapse
|
2
|
Wang J, Bai Y, Liu XY, Li S, Wang Y, Zhang YM, Hong ZY. NFAT2 promotes sorafenib resistance in hepatocellular carcinoma cells by modulating calcium ion signalling. Arch Biochem Biophys 2025; 769:110440. [PMID: 40288600 DOI: 10.1016/j.abb.2025.110440] [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: 01/05/2025] [Revised: 04/01/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Treating hepatocellular carcinoma (HCC) remains challenging due to the drug resistance of HCC cells, which limits the clinical efficacy of sorafenib. This study elucidates the role of nuclear factor activated T cells 2 (NFAT2) in sorafenib resistance in HCC cells and reveals the underlying mechanism. Sorafenib-resistant cell lines were constructed, with NFAT2 overexpressed and knocked down via genetic engineering. Fura-2 detected intracellular calcium ion concentration; transmission electron microscopy (TEM) assessed organelle damage; wound healing and transwell assays evaluated cell migration and invasion; clone formation and CCK8 assays measured cell proliferation. Flow cytometry detected apoptosis; Western blot analyzed protein expressions. Tumorigenesis was evaluated using a sorafenib-resistant HCC orthotopic xenograft mouse model. We found that NFAT2 was upregulated in MHCC97H-SR and HepG2-SR cells. Overexpression of NFAT2 inhibited Ca2+ influx in MHCC97H-SR, reduced the expression of Ca2+ regulation-related proteins (p-PLCγ, p-IP3R, p-CaMKII), ER-related proteins (CPR94, CPR78), and oxidative stress-related proteins (NOX2, NOX4). NFAT2 overexpression inhibited apoptosis and enhanced cell migration, invasion, and proliferation. NFAT2 knockdown reduced tumorigenesis. Our study uncovered a mechanism by which NFAT2 increases HCC cell resistance to sorafenib by altering intracellular calcium ion signals, highlighting NFAT2 as a promising target for HCC drug therapy.
Collapse
Affiliation(s)
- Jian Wang
- Department of Comprehensive Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Yi Bai
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Xian-Yi Liu
- Department of Comprehensive Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Shuang Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, China
| | - Ying Wang
- Department of Comprehensive Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Ya-Min Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, Tianjin, China.
| | - Zhang-Yong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
3
|
Lei Y, Cai S, Zhang JK, Ding SQ, Zhang ZH, Zhang CD, Dai DQ, Li YS. The role and mechanism of fatty acid oxidation in cancer drug resistance. Cell Death Discov 2025; 11:277. [PMID: 40514365 PMCID: PMC12166077 DOI: 10.1038/s41420-025-02554-1] [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: 03/16/2025] [Revised: 05/06/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025] Open
Abstract
Cancer is a leading cause of death globally. While drug treatment is the most commonly used method for cancer therapy, it is often hampered by drug resistance. Consequently, the mechanisms of drug resistance in cancer therapy have become a focus of current research. The mechanisms underlying cancer drug resistance are complex and may involve genetic mutation, immune escape, and metabolic reprogramming, amongst others. Metabolic reprogramming is an important marker of tumor cells, and an increasing number of studies have shown that cancer drug resistance is correlated with metabolic reprogramming, especially when fatty acid oxidation (FAO) is involved. More importantly, many preclinical studies have shown that when anti-tumor drugs are combined with FAO inhibitors, cancer cell resistance to drugs can be reversed and the effectiveness of tumor therapy is enhanced. This review provides a comprehensive overview of the mechanisms by which FAO leads to cancer resistance as well as potential targets for inhibition of FAO.
Collapse
Affiliation(s)
- Yun Lei
- Department of Surgical Oncology and 8th General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Shuang Cai
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jia-Kui Zhang
- Department of Surgical Oncology and 8th General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Si-Qi Ding
- Department of Surgical Oncology and 8th General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhan-He Zhang
- Department of Surgical Oncology and 8th General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Chun-Dong Zhang
- Department of Surgical Oncology and 8th General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Dong-Qiu Dai
- Department of Surgical Oncology and 8th General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
- Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Yong-Shuang Li
- Department of Surgical Oncology and 8th General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
4
|
El Behery M, Abo-Elmatty DM, Alsunbul M, Mohey El-Deen YI, Mohamed DI, Rashad Sindi E, Hashem MH, Al-Olayan E, Ibrahim IAA, Bamagous GA, Mali S, Saied EM. Exploration of the anticancer efficacy of a novel 1,3-thiazole analog in an ehrlich ascites carcinoma model: in vivo and in silico insights into hepatorenal protective potentials via the modulation of apoptosis, oxidative stress and inflammation. RSC Adv 2025; 15:20143-20167. [PMID: 40519680 PMCID: PMC12163905 DOI: 10.1039/d5ra01014d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/20/2025] [Indexed: 06/18/2025] Open
Abstract
Thiazoles, as a class of compounds, offer a diverse array of analogs that are pivotal in the rational design of anticancer agents. Recently, we reported a novel 1,3-thiazole analog, 2-(1-(2-(4-(4-bromophenyl)thiazol-2-yl)hydrazinylidene)ethyl)phenol (BTHP), that exhibited potential cytotoxic activity toward breast cancer cells. In the present study, we extended our investigations to explore the anticancer potential of BTHP in Ehrlich Ascites Carcinoma (EAC)-administrated female Swiss albino mice. Our findings revealed that, compared with the control group, the expression levels of antioxidant enzymes significantly decreased in the EAC-induced model group, while the level of lipid peroxidation substantially increased. Furthermore, the administration of EAC impaired hepatorenal function, as indicated by a significant increase in serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, and urea levels and a decrease in total protein and albumin levels. EAC-induced renal and hepatic damage was accompanied by elevated expression of proinflammatory biomarkers (TGF-β, NFκB, and IL6 genes) and altered serum apoptotic signaling, including reduced p53, Bax, caspase-3, and cytochrome c levels, alongside increased Bcl-2 expression. Interestingly, administration of BTHP (5 mg per kg per day, 14 days) significantly mitigated the viable EAC cell count (38%) and enhanced lifespan (131.25%) compared to untreated EAC-bearing mice. Furthermore, compared with the EAC-induced model group, the BTHP-treated EAC-induced group exhibited significantly attenuated lipid peroxidation levels and enhanced antioxidant enzyme activity (superoxide dismutase, glutathione, and catalase). Moreover, BTHP improved hepatorenal function by restoring serum ALT, AST, urea, creatinine, albumin, and total protein levels. Remarkably, BTHP reversed the apoptotic dysregulation observed in the EAC model, significantly elevating p53, Bax, caspase-3, and cytochrome c levels while suppressing Bcl-2 expression. Anti-inflammatory effects were further evidenced by diminished NFκB, TGF-β, and IL6 expression in liver and kidney tissues. Histological examinations confirmed BTHP's efficacy in attenuating EAC-induced renal and hepatic damage, preserving structural integrity. Finally, detailed molecular modeling investigations revealed that BTHP exhibits a pronounced binding affinity toward key protein targets associated with the observed anticancer activity. Overall, this study underscores the anticancer efficacy of BTHP through the regulation of antioxidant, proinflammatory, and apoptotic biomarkers, highlighting its protective effects on hepatorenal function and its therapeutic potential.
Collapse
Affiliation(s)
- Mohammed El Behery
- The Division of Biochemistry, Chemistry Department, Faculty of Science, Port Said University Port Said 42526 Egypt
| | - Dina M Abo-Elmatty
- Biochemistry Department, Faculty of Pharmacy, Suez Canal University Ismailia 415222 Egypt
| | - Maha Alsunbul
- Department of Pharmaceutical Sciences., College of Pharmacy, Princess Nourah Bint Abdulrahman University P.O. Box 84428 Riyadh 11671 Saudi Arabia
| | - Yassmina I Mohey El-Deen
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University Ismailia 41522 Egypt
| | - Doaa I Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University Cairo 11566 Egypt
| | - Emad Rashad Sindi
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah Jeddah 23890 Saudi Arabia
| | - Maha H Hashem
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University Cairo 11566 Egypt
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University Riyadh Saudi Arabia
| | - Ibrahim Abdel Aziz Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University Makkah Saudi Arabia
| | - Ghazi A Bamagous
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University Makkah Saudi Arabia
| | - Suraj Mali
- School of Pharmacy, D.Y. Patil University (Deemed to be University) Sector 7, Nerul 400706 Navi Mumbai India
| | - Essa M Saied
- Chemistry Department, Faculty of Science, Suez Canal University Ismailia 41522 Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin 12489 Berlin Germany
| |
Collapse
|
5
|
Lyu WY, Cao J, Deng WQ, Huang MY, Guo H, Li T, Lin LG, Lu JJ. Xerophenone H, a naturally-derived proteasome inhibitor, triggers apoptosis and paraptosis in lung cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156647. [PMID: 40112632 DOI: 10.1016/j.phymed.2025.156647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Polycyclic polyprenylated acylphloroglucinols (PPAPs) characterized by unique chemical architectures, exhibit diverse pharmacological activities. Xerophenone H (XeH) is a PPAP extracted from the plant Garcinia multiflora Champ. ex Benth. (Clusiaceae) with a novel and unique chemical structure. Although in vitro screening has revealed the anti-cancer activity of XeH, whose in vivo effectiveness and mechanistic basis required systematic investigation. METHODS Cytotoxic effects were evaluated through MTT and colony formation assays. A subcutaneous xenograft model was established to assess in vivo anti-cancer efficacy. To elucidate the underlying mechanism of the anti-cancer effect of XeH, RNA-sequencing and western blotting were performed. A proteasome activity assay was conducted to quantify the effect of XeH. Molecular docking and cellular thermal shift assays were conducted to identify the potential molecular target for XeH. RESULTS XeH demonstrated concentration-dependent cytotoxicity in A549 cells (IC₅₀ = 12.16 μM at 48 h). Intratumoral administration (10 mg/kg triweekly) achieved 38.6 % tumor growth inhibition. XeH simultaneously triggered apoptosis and paraptosis in A549 and H460 cells. Mechanistically, XeH promoted the formation of protein aggregates and induced significant endoplasmic reticulum stress in lung cancer cells by directly interacting with PSMB5 and inhibiting proteasome activity. CONCLUSIONS XeH, a novel PPAP, was identified as a novel proteasome inhibitor. It effectively downregulated proteasome activity, and induced both apoptosis and paraptosis in lung cancer cells.
Collapse
Affiliation(s)
- Wen-Yu Lyu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Jun Cao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Hongwei Guo
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & Pharmaceutical College, Guangxi Medical University, Nanning, 530021, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, 999078, China.
| | - Li-Gen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, 999078, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao SAR, 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, 999078, China.
| |
Collapse
|
6
|
Shang Y, Pang Y, Liu T, Wang W. Application of mass cytometry in the immune microenvironment of breast cancer. Med Oncol 2025; 42:215. [PMID: 40388018 DOI: 10.1007/s12032-025-02770-w] [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: 02/08/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
The rapid development of immunotherapy has shown preliminary clinical efficacy and significant anti-tumor effects in some cancer patients. Although immunotherapy has been approved for breast cancer, some breast cancer patients still do not benefit from it due to issues such as immunotherapy insensitivity and resistance. Mass cytometry, as a mature single-cell proteomic analysis method, with its high-throughput capabilities, has been widely used in the analysis of tumor immune microenvironments and immune cell subpopulations. Using mass cytometry to analyze the immune microenvironment of breast cancer and explore new immunotherapy targets can help improve the current status of breast cancer immunotherapy and develop personalized treatment plans for more patients. This review surveys the recent advancements in analyzing the single-cell components of breast cancer using mass cytometry technology and reviews the immune microenvironment of breast cancer as well as potential targets for immunotherapy. These results provide new insights for the subsequent research of the immune microenvironment of breast cancer and targeted immunotherapy.
Collapse
Affiliation(s)
- Yuefeng Shang
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Yuheng Pang
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Tong Liu
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, No.8, Xi Tou Tiao, Youanmen Wai, Fengtai District, Beijing, 100069, People's Republic of China.
| |
Collapse
|
7
|
Shah S, D'Souza GGM. Modeling Tumor Microenvironment Complexity In Vitro: Spheroids as Physiologically Relevant Tumor Models and Strategies for Their Analysis. Cells 2025; 14:732. [PMID: 40422235 DOI: 10.3390/cells14100732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite their widespread use, conventional two-dimensional monolayer cultures fail to reproduce these complexities, contributing to the poor translational predictability of many preclinical candidates. Three-dimensional multicellular tumor spheroids have emerged as more representative in vitro models that capture essential features of tumor architecture, stromal interactions, and microenvironmental resistance mechanisms. Spheroids exhibit spatially organized regions of proliferation, quiescence, and hypoxia, and can incorporate non-tumor cells to mimic tumor-stroma crosstalk. Advances in spheroid analysis now enable detailed evaluation of drug penetration, cellular migration, cytotoxic response, and molecular gradients using techniques such as optical and confocal imaging, large-particle flow cytometry, biochemical viability assays, and microfluidic integration. By combining physiological relevance with analytical accessibility, spheroid models support mechanistic studies of drug transport and efficacy under tumor-like conditions. Their adoption into routine preclinical workflows has the potential to improve translational accuracy while reducing reliance on animal models.
Collapse
Affiliation(s)
- Shrey Shah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
- Atom Bioworks Inc., Cary, NC 27513, USA
| | - Gerard G M D'Souza
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| |
Collapse
|
8
|
Perez-Medina M, Benito-Lopez JJ, Aguilar-Cazares D, Lopez-Gonzalez JS. A Comprehensive Review of Long Non-Coding RNAs in the Cancer-Immunity Cycle: Mechanisms and Therapeutic Implications. Int J Mol Sci 2025; 26:4821. [PMID: 40429961 PMCID: PMC12111859 DOI: 10.3390/ijms26104821] [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: 02/19/2025] [Revised: 05/10/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as pivotal regulators of the dynamic interplay between cancer progression and immune responses. This review explored their influence on key processes of the cancer-immunity cycle, such as immune cell differentiation, antigen presentation, and tumor immunogenicity. By modulating tumor escape from the immune response, therapeutic resistance, and tumor-stroma interactions, lncRNAs actively shape the tumor microenvironment. Due to their growing knowledge in the area of immune suppression, directly intervening in the induction of regulatory T cells (Tregs), M2 macrophages, and regulating immune checkpoint pathways such as PD-L1, CTLA-4, and others, lncRNAs can be considered promising therapeutic targets. Advances in single-cell technologies and immunotherapy have significantly expanded our understanding of lncRNA-driven regulatory networks, paving the way for novel precision medicine approaches. Ultimately, we discussed how targeting lncRNAs could enhance cancer immunotherapy, offering new avenues for biomarker discovery and therapeutic intervention.
Collapse
Affiliation(s)
- Mario Perez-Medina
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico; (M.P.-M.); (J.J.B.-L.); (D.A.-C.)
- Asociación Para Evitar la Ceguera en México, I. A. P., Mexico City 04030, Mexico
| | - Jesus J. Benito-Lopez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico; (M.P.-M.); (J.J.B.-L.); (D.A.-C.)
| | - Dolores Aguilar-Cazares
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico; (M.P.-M.); (J.J.B.-L.); (D.A.-C.)
| | - Jose S. Lopez-Gonzalez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico; (M.P.-M.); (J.J.B.-L.); (D.A.-C.)
| |
Collapse
|
9
|
Pereira AR, Pires PC, Hameed H, Lopes D, Lopes J, Sousa-Oliveira I, Babaie S, Mazzola P, Veiga F, Paiva-Santos AC. Injectable nanocomposite hydrogels for targeted intervention in cancer, wound healing, and bone and myocardial tissue engineering. Drug Deliv Transl Res 2025:10.1007/s13346-025-01864-2. [PMID: 40358831 DOI: 10.1007/s13346-025-01864-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2025] [Indexed: 05/15/2025]
Abstract
Despite current medicine's fast-paced advances, many acute and chronic illnesses still lack truly effective and safe therapies. Cancer treatments often lead to off-target healthy tissue damage and poor therapeutic outcomes, wound standard treatments generally demonstrate poor healing efficacy and increased susceptibility to infection, and bone tissue engineering and myocardial tissue engineering can result in immunological rejection and limited availability. To tackle these issues, injectable hydrogels have emerged, and through the incorporation of nanoparticles, nanocomposite hydrogels have appeared as versatile platforms, offering improved biocompatibility, mechanical strength, stability, and precise controlled drug release, as well as targeted delivery with increased drug retention at the site of action, reducing systemic drug distribution to non-target sites. With the ability to deliver a diverse range of therapeutic entities, including low molecular weight drugs, proteins, antibodies, and even isolated cells, injectable nanocomposite hydrogels have revolutionized current therapies, working as multifunctional platforms capable of improving efficacy and safety in cancer treatment, including in chemotherapy, immunotherapy, photothermal therapy, magnetic hyperthermia, photodynamic therapy, chemodynamic therapy, radiotherapy, molecularly targeted therapy, and after tumor surgical removal, and in general, chronic diabetic or tumor-induced wound healing, as well as in bone tissue engineering and myocardial tissue engineering. This review provides a thorough summary and critical insight of current advances on injectable nanocomposite hydrogels as an innovative approach that could bring substantial contributions to biomedical research and clinical practice, with a focus on their applications in cancer therapy, wound healing management, and tissue engineering.
Collapse
Affiliation(s)
- Ana Rita Pereira
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 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.
- Department of Medical Sciences, Faculty of Health Sciences, RISE-Health, University of Beira Interior, Av. Infante D. Henrique, 6200 - 506, Covilhã, Portugal.
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Daniela Lopes
- 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
| | - Joana Lopes
- 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
| | - Inês Sousa-Oliveira
- 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
| | - Soraya Babaie
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences, Tabriz, 51368, Iran
| | - Priscila Mazzola
- Faculty of Pharmaceutical Sciences, Universidade Estadual de Campinas, Campinas, SP, 13083 - 970, Brazil
| | - 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.
| |
Collapse
|
10
|
Xue F, Yang H, Xu P, Zhang S, Britzen-Laurent N, Bao LL, Grützmann R, Krautz C, Pilarsky C. CRISPR/Cas9 Screening Highlights PFKFB3 Gene as a Major Contributor to 5-Fluorouracil Resistance in Esophageal Cancer. Cancers (Basel) 2025; 17:1637. [PMID: 40427135 PMCID: PMC12109790 DOI: 10.3390/cancers17101637] [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: 03/17/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Esophageal cancer (EC) is the eighth most common cancer and the sixth most common cause of death worldwide. Esophageal squamous cell carcinoma (ESCC) comprises the majority of esophageal cancers globally, and 5-Fluorouraci (5-FU) is one of the commonly used chemotherapeutics for this type of cancer. Chemoresistance to drugs is a main obstacle in the successful treatment of this malignancy. METHODS In this study, we used the CRISPR/Cas9 screening method to determine the target gene related to 5-FU drug resistance in esophageal cancer. RESULTS Our research findings indicate that the loss of PFKFB3 can increase the resistance of different human esophageal squamous cell carcinoma cell lines to 5-FU through various pathways. Specifically, in KYSE-70 cells, loss of PFKFB3 can induce epithelial-mesenchymal transition (EMT) and prolong the S phase of the cell cycle, allowing cancer cells to evade the effects of 5-FU and develop resistance. In the KYSE-270 and KYSE-150 cell lines, loss of PFKFB3 can upregulate the expression of Slug and Mcl-1, indirectly regulate Chk1 and promote its autophosphorylation, which in turn inhibits apoptosis, thus counteracting the effects of 5-FU. CONCLUSIONS Our research not only enriches our understanding of the biological characteristics of different ESCC cell lines but also provides new clinical insights for future personalized treatments. Assessing the status of PFKFB3 can help predict resistance to 5-FU in ESCC patients with different genetic backgrounds, allowing for more precise treatment planning. This personalized approach has the potential to improve treatment efficacy, reduce unnecessary drug use and side effects, and ultimately improve patient survival rates and quality of life.
Collapse
Affiliation(s)
- Feng Xue
- Department of Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (F.X.); (P.X.); (S.Z.); (N.B.-L.); (R.G.); (C.K.)
| | - Hai Yang
- Department of Surgery, Juraklinik Scheßlitz, 96110 Scheßlitz, Germany;
| | - Pengyan Xu
- Department of Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (F.X.); (P.X.); (S.Z.); (N.B.-L.); (R.G.); (C.K.)
| | - Shuman Zhang
- Department of Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (F.X.); (P.X.); (S.Z.); (N.B.-L.); (R.G.); (C.K.)
| | - Nathalie Britzen-Laurent
- Department of Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (F.X.); (P.X.); (S.Z.); (N.B.-L.); (R.G.); (C.K.)
| | - Li-Li Bao
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91052 Erlangen, Germany;
| | - Robert Grützmann
- Department of Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (F.X.); (P.X.); (S.Z.); (N.B.-L.); (R.G.); (C.K.)
| | - Christian Krautz
- Department of Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (F.X.); (P.X.); (S.Z.); (N.B.-L.); (R.G.); (C.K.)
| | - Christian Pilarsky
- Department of Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (F.X.); (P.X.); (S.Z.); (N.B.-L.); (R.G.); (C.K.)
| |
Collapse
|
11
|
Sarlak S, Pagès G, Luciano F. Enhancing radiotherapy techniques for Triple-Negative breast cancer treatment. Cancer Treat Rev 2025; 136:102939. [PMID: 40286498 DOI: 10.1016/j.ctrv.2025.102939] [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: 01/31/2025] [Revised: 03/22/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Breast cancer is the most prevalent cancer among women worldwide, with various subtypes that require distinct treatment approaches. Among these, Triple-Negative Breast Bancer (TNBC) is recognized as the most aggressive form, often associated with poor prognosis due to its lack of targeted therapeutic options. This review specifically focuses on Radiotherapy (RT) as a treatment modality for TNBC, evaluating recent advancements and ongoing challenges, particularly the issue of radioresistance. RT remains an essential part in the management of breast cancer, including TNBC. Over the years, multiple improvements have been made to enhance RT effectiveness and minimize resistance. The introduction of advanced techniques such as Stereotactic Body Radiation Therapy (SBRT) and Stereotactic Radiosurgery (SRS) has significantly improved precision and reduced toxicity. More recently, proton radiation therapy, a novel RT modality, has been introduced, offering enhanced dose distribution and reducing damage to surrounding healthy tissues. Despite these technological advancements, a subset of TNBC patients continues to exhibit resistance to RT, leading to recurrence and poor treatment outcomes. To overcome radioresistance, there is an increasing interest in combining RT with targeted therapeutic agents that sensitize cancer cells to radiation. Radiosensitizing drugs have been explored to enhance the efficacy of RT by making cancer cells more susceptible to radiation-induced damage. Potential candidates include DNA damage repair inhibitors, immune checkpoint inhibitors, and small-molecule targeted therapies that interfere with key survival pathways in TNBC cells. In conclusion, while RT remains a crucial modality for TNBC treatment, radioresistance remains a significant challenge. Future research should focus on optimizing RT techniques while integrating radiosensitizing agents to improve treatment efficacy. By combining RT with targeted drug therapy, a more effective and personalized treatment approach can be developed, ultimately improving patient outcomes and reducing recurrence rates in TNBC.
Collapse
Affiliation(s)
- Saharnaz Sarlak
- Cote d'Azur University (UCA), Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, France.
| | - Gilles Pagès
- Cote d'Azur University (UCA), Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, France.
| | - Frédéric Luciano
- Cote d'Azur University (UCA), Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, France.
| |
Collapse
|
12
|
Heymann D, Muñoz-Garcia J, Babuty A, Audéon A, Ollivier E, Papy-Garcia D, Chantepie S, Zykwinska A, Sinquin C, Colliec-Jouault S. A new promising anticancer agent: A glycosaminoglycan-mimetic derived from the marine bacterial infernan exopolysaccharide. Int J Biol Macromol 2025; 308:142074. [PMID: 40118403 DOI: 10.1016/j.ijbiomac.2025.142074] [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: 09/30/2024] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
Marine microorganisms are a promising source of innovative compounds for medical applications. The present study aimed to investigate anticancer potential of oversulfated low molecular weight derivatives, named OSIDs, prepared from infernan, a marine bacterial exopolysaccharide. In order to identify a lead, OSIDs with different sulfate contents and molecular weights were firstly evaluated in vitro in a large series of human and murine tumor cell lines. Among all derivatives tested, OSID4 was the most effective, showing a significant dose-dependent inhibitory effect on the viability of cancer cells. OSID4 was then able to significantly slow down progression of lung and melanoma tumor growth in immunocompetent tumor-bearing mouse models. In immunodeficient mice bearing a human lung carcinoma, a notable inhibitory effect of OSID4, comparable to doxorubicin, was observed. In combination with doxorubicin, OSID4 did not exhibit any drug interaction. The activity of OSID4 was confirmed by its modulatory effect on the transcriptomic profile of human lung cancer cells. Finally, toxicity and pharmacokinetic parameters disclosed that OSID4 presented no toxicity and no bleeding risk. In conclusion, by combining its notable anticancer and moderate anticoagulant activities, OSID4 may be promising for treatment of cancers associated with a high risk of thromboembolic events.
Collapse
Affiliation(s)
- Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, F44322 Nantes, France; Institut de cancérologie de l'Ouest, Tumor Heterogeneity and Precision Medicine Laboratory, F-44805 Saint Herblain, France; University of Sheffield, School of Medicine and Population Health, S102RX Sheffield, UK.
| | - Javier Muñoz-Garcia
- Nantes Université, CNRS, UMR6286, US2B, F44322 Nantes, France; Institut de cancérologie de l'Ouest, Tumor Heterogeneity and Precision Medicine Laboratory, F-44805 Saint Herblain, France
| | - Antoine Babuty
- Nantes Université, CNRS, UMR6286, US2B, F44322 Nantes, France; Institut de cancérologie de l'Ouest, Tumor Heterogeneity and Precision Medicine Laboratory, F-44805 Saint Herblain, France; CHU de Nantes, Department of Hemostasis, F-44201 Nantes, France
| | - Antoine Audéon
- Institut de cancérologie de l'Ouest, Tumor Heterogeneity and Precision Medicine Laboratory, F-44805 Saint Herblain, France; SATT Ouest Valorisation, F-44201 Nantes, France
| | - Emilie Ollivier
- Institut de cancérologie de l'Ouest, Tumor Heterogeneity and Precision Medicine Laboratory, F-44805 Saint Herblain, France
| | - Dulce Papy-Garcia
- Université Paris Est Créteil (UPEC), Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), F-94010 Créteil, France
| | - Sandrine Chantepie
- Université Paris Est Créteil (UPEC), Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), F-94010 Créteil, France
| | - Agata Zykwinska
- Ifremer, MASAE Microbiologie Aliment Santé Environnement, F-44000 Nantes, France
| | - Corinne Sinquin
- Ifremer, MASAE Microbiologie Aliment Santé Environnement, F-44000 Nantes, France
| | | |
Collapse
|
13
|
Naeem AA, Abdulsamad SA. Molecular Mechanisms of Castration-Resistant Prostate Cancer Progression. Cureus 2025; 17:e83813. [PMID: 40491606 PMCID: PMC12146216 DOI: 10.7759/cureus.83813] [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] [Accepted: 05/08/2025] [Indexed: 06/11/2025] Open
Abstract
Prostate cancer is a global health issue and one of the most common reasons for cancer-related mortality. This research aimed to investigate the molecular mechanisms underlying the progression to castration-resistant prostate cancer (CRPC). Differential gene expression was analyzed by contrasting the PNT2 prostate epithelial cell line and the PC3M CRPC cell line. RNA sequencing was performed on three biological replicates of each cell type, and 1,000 differentially expressed genes were identified with a fold change ≥1 and a p<0.05. A heatmap was generated to visualize the gene expression profiles, and the top 10 significantly altered genes were identified. Functional enrichment analysis, including Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis, was conducted to map the biological processes, cellular components, and molecular functions associated with the differentially expressed genes. Furthermore, weighted gene co-expression network analysis was utilized to identify co-expression modules and significant genes, thereby highlighting the top 10 most significant differentially expressed genes in significant pathways. The findings indicate substantial molecular alterations associated with the development of castration-resistant prostate cancer, with major pathways including metabolic deregulation pathways and cell cycle regulation. The identified differentially expressed genes (DEGs) and pathways provide significant insights into disease progression and potential therapeutic targets. These findings contribute to the understanding of prostate cancer at the molecular level and can be used to identify new diagnostic and therapeutic strategies. However, further validation is required to determine the clinical significance of these targets in the treatment of CRPC.
Collapse
Affiliation(s)
- Abdulghani A Naeem
- Basic Sciences, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Basic Sciences, King Abdullah International Medical Research Center, Jeddah, SAU
| | - Saud A Abdulsamad
- Basic Sciences, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Basic Sciences, King Abdullah International Medical Research Center, Jeddah, SAU
| |
Collapse
|
14
|
Amereh M, Seyfoori A, Shojaei S, Lane S, Zhao T, Shokrollahi Barough M, Lum JJ, Walter P, Akbari M. Tumoroid Model Reveals Synergistic Impairment of Metabolism by Iron Chelators and Temozolomide in Chemo-Resistant Patient-derived Glioblastoma Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412505. [PMID: 40285641 PMCID: PMC12120723 DOI: 10.1002/advs.202412505] [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] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/18/2025] [Indexed: 04/29/2025]
Abstract
Chemoresistance poses a significant clinical challenge in managing glioblastoma (GBM), limiting the long-term success of traditional treatments. Here, a 3D tumoroid model is used to investigate the metabolic sensitivity of temozolomide (TMZ)-resistant GBM cells to iron chelation by deferoxamine (DFO) and deferiprone (DFP). This work shows that TMZ-resistant GBM cells acquire stem-like characteristics, higher intracellular iron levels, higher expression of aconitase, and elevated reliance on oxidative phosphorylation and proteins associated with iron metabolism. Using a microphysiological model of GBM-on-a-chip consisting of extracellular matrix (ECM)-incorporated tumoroids, this work demonstrates that the combination of iron chelators with TMZ induces a synergistic effect on an in vitro tumoroid model of newly diagnosed and recurrent chemo-resistant patient-derived GBM and reduced their size and invasion. Investigating downstream metabolic variations reveal reduced intracellular iron, increased reactive oxygen species (ROS), upregulated hypoxia-inducible factor-1α, reduced viability, increased autophagy, upregulated ribonucleotide reductase (RRM2), arrested proliferation, and induced cell death in normoxic TMZ-resistant cells. Hypoxic cells, while showing similar results, display reduced responses to iron deficiency, less blebbing, and an induced autophagic flux, suggesting an adaptive mechanism associated with hypoxia. These findings show that co-treatment with iron chelators and TMZ induces a synergistic effect, making this combination a promising GBM therapy.
Collapse
Affiliation(s)
- Meitham Amereh
- Laboratory for Innovations in Micro Engineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBCV8P 5C2Canada
| | - Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBCV8P 5C2Canada
| | - Shahla Shojaei
- Department of Human Anatomy and Cell ScienceMax Rady College of MedicineRady Faculty of Health SciencesUniversity of ManitobaWinnipegMBR3T 2N2Canada
| | - Sarah Lane
- Department of BiologyUniversity of VictoriaBCCanada
| | - Tian Zhao
- Trev and Joyce Deeley Research CentreBC CancerVictoriaBCV8R 6V5Canada
| | - Mahdieh Shokrollahi Barough
- Laboratory for Innovations in Micro Engineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBCV8P 5C2Canada
| | - Julian J. Lum
- Trev and Joyce Deeley Research CentreBC CancerVictoriaBCV8R 6V5Canada
- Department of Biochemistry and MicrobiologyUniversity of VictoriaVictoriaBCV8W 2Y2Canada
| | | | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBCV8P 5C2Canada
- Terasaki Institute for Biomedical InnovationsLos AngelesCA91367USA
| |
Collapse
|
15
|
Lee Y. Principles and therapeutics of cancer. Mol Cells 2025; 48:100201. [PMID: 40024317 PMCID: PMC12034218 DOI: 10.1016/j.mocell.2025.100201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Affiliation(s)
- Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea.
| |
Collapse
|
16
|
Chen R, Zhang R, Ke F, Guo X, Zeng F, Liu Q. Mechanisms of breast cancer metastasis: the role of extracellular matrix. Mol Cell Biochem 2025; 480:2771-2796. [PMID: 39652293 DOI: 10.1007/s11010-024-05175-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/23/2024] [Indexed: 05/03/2025]
Abstract
The components of the extracellular matrix (ECM) are dynamic, and they mediate mechanical signals that modulate cellular behaviors. Disruption of the ECM can induce the migration and invasion of cancer cells via specific signaling pathways and cytokines. Metastasis is a leading cause of high mortality in malignancies, and early intervention can improve survival rates. However, breast cancer is frequently diagnosed subsequent to metastasis, resulting in poor prognosis and distant metastasis poses substantial hurdles in therapy. In breast cancer, there is notable tissue remodeling of ECM proteins, with several identified as essential components for metastasis. Moreover, specific ECM molecules, receptors, enzymes, and various signaling pathways play crucial roles in breast cancer metastasis, drug treatment, and resistance. The in-depth consideration of these elements could provide potential therapeutic targets to enhance the survival rates and quality of life for breast cancer patients. This review explores the mechanisms by which alterations in the ECM contribute to breast cancer metastasis and discusses current clinical applications targeting ECM in breast cancer treatment, offering valuable perspectives for future ECM-based therapies.
Collapse
Affiliation(s)
- Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Famin Ke
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
17
|
Muttiah B, Hanafiah A. Gut Microbiota and Cardiovascular Diseases: Unraveling the Role of Dysbiosis and Microbial Metabolites. Int J Mol Sci 2025; 26:4264. [PMID: 40362500 PMCID: PMC12072866 DOI: 10.3390/ijms26094264] [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: 04/09/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular diseases (CVDs), including heart failure (HF), hypertension, myocardial infarction (MI), and atherosclerosis, are increasingly linked to gut microbiota dysbiosis and its metabolic byproducts. HF, affecting over 64 million individuals globally, is associated with systemic inflammation and gut barrier dysfunction, exacerbating disease progression. Similarly, hypertension and MI correlate with reduced microbial diversity and an abundance of pro-inflammatory bacteria, contributing to vascular inflammation and increased cardiovascular risk. Atherosclerosis is also influenced by gut dysbiosis, with key microbial metabolites such as trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs) playing crucial roles in disease pathogenesis. Emerging evidence highlights the therapeutic potential of natural compounds, including flavonoids, omega-3 fatty acids, resveratrol, curcumin, and marine-derived bioactives, which modulate the gut microbiota and confer cardioprotective effects. These insights underscore the gut microbiota as a critical regulator of cardiovascular health, suggesting that targeting dysbiosis may offer novel preventive and therapeutic strategies. Further research is needed to elucidate underlying mechanisms and optimize microbiome-based interventions for improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- GUT Research Group, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
18
|
Li Y, Wang W, Xu J, Zhao B, Xiong L, Ge D, Wu Y, Dou X, Fu Y, Wang L, Zhao C, Chen M. Tumor signal amplification and immune decoy strategy using bacterial membrane-coated nanoparticles for immunotherapy. Biomater Sci 2025; 13:2368-2380. [PMID: 40104961 DOI: 10.1039/d4bm01535e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
In cancer therapy, tumor cells can diminish their signals through mechanisms such as immune escape, thereby evading recognition and elimination by the immune system. Providing tumor signals to enhance the recognition of tumor sites is considered a crucial approach in cancer treatment. Inspired by the decoy-induced directed feeding of fish, we propose a biomimetic nanoparticle system for tumor signal amplification. This biomimetic system comprises magnetically responsive nanoparticles and immune-inducing bacterial membranes. These designs work together to create a baiting effect at the tumor site, attracting and activating immune cells to attack. It has been demonstrated that the generated nanoparticles have the potential to be targeted and delivered to the tumor site under the influence of an external magnetic field, as demonstrated in preliminary in vitro and in vivo studies. Moreover, the nanoparticles utilize the bacterial membrane and cell membrane-translocated calreticulin to induce an immune response, simulating a decoy mechanism to recruit immune cells. The nanoparticles were proved to be effective in recruiting macrophages and neutrophils and reducing tumor size in animal experiments. These features make the nanoparticles an ideal candidate for treating tumors.
Collapse
Affiliation(s)
- Yifan Li
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Weiwei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Jiale Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Bei Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Longying Xiong
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Dan Ge
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Yanping Wu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Xiaotan Dou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Yuping Fu
- Department of Pharmacy, Zhangjiajie People's Hospital, Hunan, 427099, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Cheng Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Min Chen
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
19
|
Liu Y, Yang Y, Qi X. Combination Therapy for Overcoming Multidrug Resistance in Breast Cancer Through Hedgehog Signaling Pathway Regulation. Pharmaceutics 2025; 17:572. [PMID: 40430864 PMCID: PMC12114799 DOI: 10.3390/pharmaceutics17050572] [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: 03/09/2025] [Revised: 04/09/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: The ineffective delivery of drugs into tumors and the existence of multidrug resistance (MDR) are the primary causes of chemotherapy failure. Downregulation of the Sonic Hedgehog (Shh) pathway has been shown to reduce P-glycoprotein (P-gp) expression on cell membranes and to resist MDR. Methods: In this study, we combine cyclopamine (CYP, a potent Shh antagonist) with paclitaxel (PTX, an antitumor drug that can produce MDR) in a nano-drug delivery system (CYP NP and PTX NP) for the treatment of drug-resistant breast cancer. Nanoparticles were characterized for size, zeta potential, and encapsulation efficiency. P-gp expression, nanoparticle accumulation, cytotoxicity, and apoptosis were evaluated in MCF-7 and MCF-7/Adr cells. Penetration ability was assessed using 3D multicellular tumor spheroids. Antitumor efficacy and nanoparticle biodistribution were validated in MCF-7/Adr-bearing nude mice models. Results: Our engineered CYP nanoparticles (~200 nm) demonstrated prolonged intratumoral retention, enabling sustained Shh pathway inhibition and P-gp functional suppression. This size-optimized formulation created a favorable tumor microenvironment for the smaller PTX nanoparticles (~30 nm), facilitating deeper tumor penetration and enhanced cellular uptake. Meanwhile, by down-regulating P-gp expression, CYP NPs could convert drug-resistant cells to PTX-sensitive cells in both cytotoxicity and apoptosis induction through the Shh pathway. The combination of CYP NP and PTX NP augmented the antitumor effects in MCF-7/Adr-bearing nude mice models. Conclusions: The CYP NP and PTX NP combination offers a new therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; (Y.L.); (Y.Y.)
| | - Yiliang Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; (Y.L.); (Y.Y.)
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, China
| | - Xianrong Qi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; (Y.L.); (Y.Y.)
| |
Collapse
|
20
|
Ei ZZ, Racha S, Chunhacha P, Yokoya M, Moriue S, Zou H, Chanvorachote P. Substituents introduction of methyl and methoxy functional groups on resveratrol stabilizes mTOR binding for autophagic cell death induction. Sci Rep 2025; 15:14675. [PMID: 40287470 PMCID: PMC12033263 DOI: 10.1038/s41598-025-98616-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
The regulation of the mammalian target of rapamycin (mTOR) protein by cancer cells can lead to uncontrol of cancer cell growth and cancer therapy resistance. The drug discovery of the anticancer agent 5-(3-hydroxy-4-methoxyphenethyl)-2-methoxy-3-methylphenol (SM-3), a derivative of resveratrol by substituting a methyl group at the hydroxy group of ring A and adding a methoxy group at the para position of ring B, shows promising potential for targeting autophagy to induce cell death and suppress cancer stem cells (CSCs) through the inhibition of the mTOR protein. In human lung cancer cells, SM-3 showed greater efficacy, with lower IC50 values of 72.74 ± 0.13, 67.66 ± 0.10, and 43.24 ± 0.11 µM in A549, H292, and H460 cells, respectively, compared to the parent compound, Resveratrol (Res). Moreover, the selectivity index (SI) values for BEAS2B cells compared to tumor cells treated with SM-3 were 10.99, 11.81, and 18.49 for A549, H292, and H460 cell lines, respectively. Therefore, SM-3 treatment led to reduced proliferation rates and colony formation in lung cancer cells. In our study, spheroids treated with SM-3 showed a higher proportion of dead spheroids compared to those treated with Res. Additionally, SM-3 treatment resulted in decreased expression of stem cell markers (CD133, CD44, and ALDH1A1) and transcription factors (OCT4, NANOG, and SOX2) in spheroids and organoids from human lung cancer cells by inhibiting the mTOR/pAkt pathway. SM-3 was also found to induce autophagic cell death, as indicated by Monodansylcadaverine staining, acidic vesicle formation, and the conversion of LC3BI to LC3BII. Using MM/GBSA calculations, SM-3 exhibited a stronger binding affinity (-25.09 kcal/mol) compared to Res (-18.85 kcal/mol). SM-3 also displayed greater stability during the entire simulation, maintaining lower RMSD values of 2-3 Å even after 80 ns. In summary, the introduction of methyl and methoxy functional groups on Res to create SM-3 effectively suppressed cancer spheroids and organoids formation in lung cancer cells by targeting the upstream mTOR/pAkt pathway.
Collapse
Affiliation(s)
- Zin Zin Ei
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn university, Bangkok, 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Satapat Racha
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn university, Bangkok, 10330, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Interdisciplinary Program in Pharmacology, Graduate School, Chulalongkorn university, Bangkok, 10330, Thailand
| | - Preedakorn Chunhacha
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Masashi Yokoya
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Sohsuke Moriue
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Hongbin Zou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn university, Bangkok, 10330, Thailand.
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand.
- Sustainable Environment Research Institute, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
21
|
Ebrahim NAA, Soliman SMA, Othman MO, Tahoun NS. Molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated Schwann cells in cancer progression and metastasis. Med Oncol 2025; 42:171. [DOI: ebrahim, n.a.a., soliman, s.m.a., othman, m.o.et al.molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated schwann cells in cancer progression and metastasis.med oncol 42, 171 (2025).https:/doi.org/10.1007/s12032-025-02729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
|
22
|
Ebrahim NAA, Soliman SMA, Othman MO, Tahoun NS. Molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated Schwann cells in cancer progression and metastasis. Med Oncol 2025; 42:171. [PMID: 40259163 DOI: 10.1007/s12032-025-02729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Perineural invasion (PNI) is a pathological process wherein cancer cells invade and spread along peripheral nerves, contributing to tumor aggressiveness and poor clinical outcomes, including increased recurrence, metastasis, and reduced survival. Tumor-associated Schwann cells (SCs) play a pivotal role in facilitating PNI by promoting epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) remodeling, and immune modulation. These cells actively support tumor progression through neurotrophin, cytokine, chemokine, and neurotransmitter signaling, enhancing cancer cell migration along neural pathways. Recent advances in imaging techniques, single-cell transcriptomics, and molecular profiling have provided deeper insights into the tumor microenvironment's role in PNI. Emerging therapeutic strategies targeting neurotrophin-mediated signaling and SC-tumor interactions have shown promise in preclinical models. However, significant research gaps remain, particularly in understanding the heterogeneity of SCs and their molecular subtypes in PNI across different malignancies. This review highlights the clinical significance, molecular mechanisms, and potential therapeutic targets associated with PNI. A comprehensive understanding of tumor-SC interactions is essential for developing targeted interventions to mitigate PNI-driven malignancies. Future research should focus on integrating multi-omics approaches and novel therapeutics to improve early detection and treatment, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Noura A A Ebrahim
- Oncologic Pathology Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt.
| | | | - Moamen O Othman
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Neveen S Tahoun
- Oncologic Pathology Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt
| |
Collapse
|
23
|
Ebrahim NAA, Soliman SMA, Othman MO, Tahoun NS. Molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated Schwann cells in cancer progression and metastasis. Med Oncol 2025; 42:171. [DOI: https:/doi.org/10.1007/s12032-025-02729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
|
24
|
Duzgun D, Oltean S. Aberrant Splicing as a Mechanism for Resistance to Cancer Therapies. Cancers (Basel) 2025; 17:1381. [PMID: 40282556 PMCID: PMC12025770 DOI: 10.3390/cancers17081381] [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: 02/17/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Cancer is biologically diverse, highly heterogeneous, and associated with molecular alterations, significantly contributing to mortality worldwide. Currently, cancer patients are subjected to single or combination treatments comprising chemotherapy, surgery, immunotherapy, radiation therapy, and targeted therapy. Chemotherapy remains the first line of treatment in cancer but faces a major obstacle in the form of chemoresistance. This obstacle has resulted in relapses and poor patient survival due to decreased treatment efficacy. Aberrant pre-mRNA alternative splicing can significantly modulate gene expression and function involved in the resistance mechanisms, potentially shaping the intricate landscape of tumour chemoresistance. Thus, novel strategies targeting abnormal pre-mRNA alternative splicing and understanding the molecular mechanisms of chemotherapy resistance could aid in overcoming the chemotherapeutic challenges. This review first highlights drug targets, drug pumps, detoxification mechanisms, DNA damage response, and evasion of apoptosis and cell death as key molecular mechanisms involved in chemotherapy resistance. Furthermore, the review discusses the progress of research on the dysregulation of alternative splicing and molecular targets involved in chemotherapy resistance in major cancer types.
Collapse
Affiliation(s)
| | - Sebastian Oltean
- Department of Clinical and Biomedical Sciences, Faculty of Health Sciences, University of Exeter, Exeter EX1 2LU, UK
| |
Collapse
|
25
|
Singh M, Arora HL, Naik R, Joshi S, Sonawane K, Sharma NK, Sinha BK. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. Int J Mol Sci 2025; 26:3852. [PMID: 40332483 PMCID: PMC12028135 DOI: 10.3390/ijms26083852] [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: 02/11/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer drug resistance occurs when cancer cells evade cell death following treatment with chemotherapy, radiation therapy, and targeted therapies. This resistance is often linked to the reprogramming of programmed cell death (PCD) pathways, allowing cancer cells to survive drug-induced stress. However, certain anticancer therapies, when combined with specific agents or inhibitors, can induce ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation. Currently, extensive preclinical and clinical research is underway to investigate the molecular, cellular, and tissue-specific mechanisms underlying ferroptosis, with the goal of identifying strategies to overcome drug resistance in cancers unresponsive to conventional PCD pathways. By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes in patients with cancer. This short review aims to enhance the understanding of ferroptosis inducers in cancer therapy and stimulate further research into ferroptosis-based approaches for more effective clinical cancer treatment.
Collapse
Affiliation(s)
- Mansaa Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Hasmiq L. Arora
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Rutuja Naik
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Shravani Joshi
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Birandra K. Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
26
|
Soltani M, Ahmadzadeh N, Nasiraei Haghighi H, Khatamian N, Homayouni Tabrizi M. Targeted cancer therapy potential of quercetin-conjugated with folic acid-modified nanocrystalline cellulose nanoparticles: a study on AGS and A2780 cell lines. BMC Biotechnol 2025; 25:29. [PMID: 40241055 PMCID: PMC12001405 DOI: 10.1186/s12896-025-00962-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: 12/01/2024] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
This study investigates the effects of quercetin-conjugated nanocrystalline cellulose/cetyltrimethylammonium bromide/folic acid nanoparticles (NCC/CTAB/FA NPs) on AGS and A2780 cancer cell lines, focusing on their cytotoxicity and antioxidant capacity. Dynamic light scattering (DLS) analysis revealed an average particle size of 388.70 nm, suitable for cellular uptake and release kinetics. The NCC/CTAB/FA NPs exhibited a rod and spherical morphology and uniform distribution, as confirmed by field emission scanning electron microscopy (FESEM). Fourier-transform infrared (FTIR) spectroscopy confirmed the successful synthesis and functional group integration, supporting the NPs' ability for drug delivery. The encapsulation efficiency value was 81.17%, demonstrating the effective incorporation of Quercetin. Cytotoxicity assays indicated significant reductions in cell viability for AGS and A2780 cells with IC50 values of 3.2 µg/mL and 16.04 µg/mL, respectively, while HDF cells exhibited higher viability. Flow cytometry analysis revealed a dose-dependent induction of apoptosis in AGS cells, supported by changes in gene expression related to apoptosis and inflammation. Furthermore, antioxidant capacity assays demonstrated practical free radical scavenging abilities, with IC50 values of 151.65 µg/mL for ABTS and 349.54 µg/mL for DPPH. NCC/CTAB/FA/Quercetin NPs exhibit promising characteristics for targeted cancer therapy and antioxidant applications.
Collapse
Affiliation(s)
- Mozhgan Soltani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Negar Ahmadzadeh
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Niloufar Khatamian
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | |
Collapse
|
27
|
Zhakupova A, Zeinolla A, Kokabi K, Sergazy S, Aljofan M. Drug Resistance: The Role of Sphingolipid Metabolism. Int J Mol Sci 2025; 26:3716. [PMID: 40332322 PMCID: PMC12027666 DOI: 10.3390/ijms26083716] [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: 03/04/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
A significant challenge in cancer treatment is the rising problem of drug resistance that reduces the effectiveness of therapeutic strategies. Current knowledge shows that multiple mechanisms play a role in cancer drug resistance. Another mechanism that has gained attention is the alteration in sphingolipid trafficking and the dysregulation of its metabolism, which was reported to cause cancer-associated drug resistance. Sphingolipids are lipids containing sphingosine and have multiple roles, ranging from lipid raft formation, apoptosis, and cell signaling to immune cell trafficking. Recent studies show that in developing cancer cells, altered or dysregulated sphingolipids are associated with drug efflux and promote the survival of cancer cells by bypassing apoptosis. Upregulated levels of the glucosylceramide synthase (GCS), an enzyme that functions in sphingolipid metabolism, lead to the upregulated ABCB1 gene that induces drug efflux from the cancer cells. These bypass mechanisms make drugs that induce apoptosis in tumor cells ineffective. By highlighting the current findings, this review aims to provide a mechanism of drug resistance caused by the dysregulation of glucosylceramide synthase, sphingosine kinase, and acid ceramidase enzymes as possible therapeutic targets to enhance the effectiveness of the currently used chemotherapeutic agents.
Collapse
Affiliation(s)
- Assem Zhakupova
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan; (A.Z.); (A.Z.); (K.K.)
| | - Adelina Zeinolla
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan; (A.Z.); (A.Z.); (K.K.)
| | - Kamilya Kokabi
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan; (A.Z.); (A.Z.); (K.K.)
| | - Shynggys Sergazy
- Drug Discovery and Development Laboratory, National Laboratory Astana, Astana 010000, Kazakhstan
- LLP “VICTUS PHARM”, Astana 010000, Kazakhstan
| | - Mohamad Aljofan
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan; (A.Z.); (A.Z.); (K.K.)
- Drug Discovery and Development Laboratory, National Laboratory Astana, Astana 010000, Kazakhstan
| |
Collapse
|
28
|
Rajabi F, Hajipour-Verdom B, Abdolmaleki P. Static magnetic field promotes the doxorubicin toxicity effects on osteosarcoma cells. Sci Rep 2025; 15:11902. [PMID: 40195518 PMCID: PMC11977194 DOI: 10.1038/s41598-025-96802-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 03/31/2025] [Indexed: 04/09/2025] Open
Abstract
Osteosarcoma, a highly aggressive bone cancer, primarily affects adolescents and is frequently treated with conventional chemotherapy, such as doxorubicin (DOX). However, the efficacy of DOX is often limited by severe side effects and drug resistance. This study investigates the synergistic effects of static magnetic fields (SMF) and DOX on G292 osteosarcoma cells and HFF normal fibroblasts. Cell viability was assessed using the MTT assay, intracellular reactive oxygen species (ROS) levels were quantified via DCFDA staining and flow cytometry, and iron and calcium homeostasis were analyzed using ICP-OES. Apoptosis and necrosis were determined through Annexin V-FITC/PI staining. Results demonstrated that the combination of SMF and DOX significantly reduced G292 cell viability compared to DOX alone, with IC50 values decreased from 3.2 µM (at 3 mT, p < 0.01) to 0.8 µM (at 24 mT, p < 0.001) at 24 h. Apoptosis rates increased from 8.12% with DOX alone to 16% with SMF + DOX. While DOX alone elevated ROS levels by 59.15% in G292 cells, SMF further amplified apoptosis by enhancing ROS generation and disrupting iron and calcium homeostasis. These findings suggest that SMF enhances DOX-induced cytotoxicity in osteosarcoma cells by promoting ROS production, altering metal ion homeostasis, and increasing apoptosis. SMF represents a promising adjuvant therapy for osteosarcoma treatment, though further in vivo studies are necessary to optimize treatment parameters and evaluate clinical applicability.
Collapse
Affiliation(s)
- Fatemeh Rajabi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154, Tehran, Iran
| | - Behnam Hajipour-Verdom
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154, Tehran, Iran
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), 1517964311, Tehran, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, 14115-154, Tehran, Iran.
| |
Collapse
|
29
|
Miyazaki J, Wagatsuma R, Okamoto K. Photothermal imaging of cellular responses to glucose deprivation. RSC Chem Biol 2025; 6:571-582. [PMID: 39927218 PMCID: PMC11801213 DOI: 10.1039/d4cb00269e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/30/2025] [Indexed: 02/11/2025] Open
Abstract
In solid tumours, cancer cells modify their metabolic processes to endure environments with nutrient and oxygen scarcity due to inadequate blood flow. A thorough understanding of this adaptive mechanism, which requires reliable microscopic techniques, is crucial for developing effective cancer treatments. In the present study, we used multi-wavelength photothermal (PT) microscopy to visualise the cellular response to glucose deprivation in living cells derived from cervical cancer. We found increased mitochondrial PT signal intensity under glucose deprivation conditions, which is indicative of a correlation between mitochondrial crista density and PT signal intensity. Furthermore, PT microscopy revealed that the activity of the autophagy-lysosome system can be evaluated by detecting substances accumulated in lysosomes. Using this method, we confirmed that ferritin and denatured proteins from the endoplasmic reticulum were present within the lysosomes. The detectability of these substances using PT microscopy at visible wavelengths indicated the presence of iron ions. This method does not require labeling of molecules and provides reliable information and detailed insights into the cellular responses associated with the adaptation of cancer cell metabolism to nutrient stress conditions.
Collapse
Affiliation(s)
- Jun Miyazaki
- Faculty of Systems Engineering, Wakayama University Wakayama 640-8510 Japan
| | - Ryotaro Wagatsuma
- Faculty of Systems Engineering, Wakayama University Wakayama 640-8510 Japan
| | - Koji Okamoto
- Graduate School of Frontier Biosciences, Osaka University Osaka 565-0871 Japan
| |
Collapse
|
30
|
Mostafa MAH, Khojah HMJ. Nanoparticle-based delivery systems for phytochemicals in cancer therapy: molecular mechanisms, clinical evidence, and emerging trends. Drug Dev Ind Pharm 2025:1-17. [PMID: 40116905 DOI: 10.1080/03639045.2025.2483425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVE This review examines recent advancements in nanoparticle-based delivery systems for phytochemicals, focusing on their role in overcoming multidrug resistance, improving therapeutic efficacy, and facilitating clinical translation. SIGNIFICANCE This review highlights recent advances in nanoparticle-enabled phytochemical delivery to enhance bioavailability, improve therapeutic outcomes, and enable targeted applications. By comparing various nanoparticle systems, formulation methods, and efficacy data, it identifies gaps in current research and guides the development of more effective, next-generation phytochemical-loaded nanocarriers. METHODS A systematic review of literature published between 2000 and 2024 was conducted using PubMed, Scopus, and Web of Science. Articles focusing on nanoparticle-based phytochemical delivery in cancer therapy were included. KEY FINDINGS Compounds such as curcumin, resveratrol, quercetin, and epigallocatechin gallate demonstrate enhanced anti-cancer efficacy when encapsulated in nanoparticles, leading to improved bioavailability, increased tumor cell targeting, and reduced toxicity. Clinical trials indicate tumor regression and fewer adverse effects. Emerging approaches-such as nanogels, hybrid nanoparticles, and combination therapies with immune checkpoint inhibitors-further refine treatment efficacy. CONCLUSIONS Nanoparticle-based delivery systems significantly improve the therapeutic potential of phytochemicals, making them promising candidates for safer, more effective cancer treatments. However, challenges related to regulatory guidelines, scalability, and long-term safety must be addressed to fully realize their clinical potential.
Collapse
Affiliation(s)
- Mahmoud A H Mostafa
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University (Assiut Branch), Assiut, Egypt
| | - Hani M J Khojah
- Department of Pharmacy Practice, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
31
|
Shang T, Jia Z, Li J, Cao H, Xu H, Cong L, Ma D, Wang X, Liu J. Unraveling the triad of hypoxia, cancer cell stemness, and drug resistance. J Hematol Oncol 2025; 18:32. [PMID: 40102937 PMCID: PMC11921735 DOI: 10.1186/s13045-025-01684-4] [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: 12/15/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
In the domain of addressing cancer resistance, challenges such as limited effectiveness and treatment resistance remain persistent. Hypoxia is a key feature of solid tumors and is strongly associated with poor prognosis in cancer patients. Another significant portion of the development of acquired drug resistance is attributed to tumor stemness. Cancer stem cells (CSCs), a small tumor cell subset with self-renewal and proliferative abilities, are crucial for tumor initiation, metastasis, and intra-tumoral heterogeneity. Studies have shown a significant association between hypoxia and CSCs in the context of tumor resistance. Recent studies reveal a strong link between hypoxia and tumor stemness, which together promote tumor survival and progression during treatment. This review elucidates the interplay between hypoxia and CSCs, as well as their correlation with resistance to therapeutic drugs. Targeting pivotal genes associated with hypoxia and stemness holds promise for the development of novel therapeutics to combat tumor resistance.
Collapse
Affiliation(s)
- Tongxuan Shang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Ziqi Jia
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiayi Li
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, 100730, China
| | - Heng Cao
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hengyi Xu
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Cong
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Dongxu Ma
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiang Wang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jiaqi Liu
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
32
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
33
|
Tiwari SK, Chandrasekharan A, Lupitha SS, Mathew KA, Jancy SV, Halikar AM, Sanjeev VS, Sivakumar KC, Prasad T, Anurup KG, Rather AA, Tiffee P J J, Jayaprasad AG, Sivasailam A, Santhoshkumar TR. Hypoxia induced mitophagy generates reversible metabolic and redox heterogeneity with transient cell death switch driving tumorigenesis. Free Radic Biol Med 2025; 230:190-208. [PMID: 39947492 DOI: 10.1016/j.freeradbiomed.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Tumor hypoxia determines tumor growth, metastasis, drug resistance, and tumor heterogeneity through multiple mechanisms, largely dependent on the extent of hypoxia, further modulated by re-oxygenation events. In order to track the cell fates under hypoxia and re-oxygenation, we have developed a sensor cell for real-time tracking of apoptotic, necrotic, and surviving mitophagy cells under hypoxia and re-oxygenation. The study using this sensor revealed a cell death switch from apoptosis to necrosis by hypoxia-exposed cells under re-oxygenation, where mitophagy plays a key role in acquiring temporally evolving functional phenotypes, including metabolic heterogeneity and mitochondrial redox heterogeneity. RNA transcriptomics also revealed a temporally evolving genomic landscape supporting the complex transcriptional plasticity of cells as a non-genetic adaptive event. Interestingly, cells regained from these distinct stages retained their metastatic potential despite slow growth in animal models. Overall, the study demonstrated that cells acquire distinct functions by tumor hypoxia and re-oxygenation, secondarily acquiring transient functional traits and metabolic heterogeneity governed by cell inherent mitochondrial dynamics. Such cell autonomous temporal alterations in cell states governed by organelle integrity with distinct cell proliferation and apoptosis-necrosis switch may be advantageous for the growing tumor to evolve under complex microenvironmental stress, further contributing to tumorigenesis.
Collapse
Affiliation(s)
- Shivanshu Kumar Tiwari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Aneesh Chandrasekharan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Santhik Subhasingh Lupitha
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Krupa Ann Mathew
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Shine Varghese Jancy
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Aman Munirpasha Halikar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Vishnu S Sanjeev
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - K C Sivakumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Tilak Prasad
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - K G Anurup
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Aijaz Ahmad Rather
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Jain Tiffee P J
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Aparna Geetha Jayaprasad
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Aswathy Sivasailam
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India; Research Centre, University of Kerala, Thiruvananthapuram, Kerala, 695534, India
| | - T R Santhoshkumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India.
| |
Collapse
|
34
|
Doddapaneni R, Tucker JD, Lu PJ, Lu QL. Synergistic Effect of Ribitol and Shikonin Promotes Apoptosis in Breast Cancer Cells. Int J Mol Sci 2025; 26:2661. [PMID: 40141303 PMCID: PMC11942206 DOI: 10.3390/ijms26062661] [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: 02/18/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
The mortality rate of breast cancer remains high, despite remarkable advances in chemotherapy. Therefore, it is imperative to identify new treatment options. In the present study, we investigated whether the metabolite ribitol enhances the cytotoxic effect of shikonin against breast cancer in vitro. Here, we screened a panel of small molecules targeting energy metabolism against breast cancer. The results of the study revealed that ribitol enhances shikonin's growth-inhibitory effects, with significant synergy. A significant (p < 0.01) increase in the percentage (56%) of apoptotic cells was detected in the combined treatment group, compared to shikonin single-treatment group (38%), respectively. The combined ribitol and shikonin treatment led to significant arrest of cell proliferation (40%) (p < 0.01) compared to untreated cells, as well as the induction of apoptosis. This was associated with upregulation of p53 (p < 0.05) and downregulation of c-Myc (p < 0.01), Bcl-xL (p < 0.001), and Mcl-1 (p < 0.05). Metabolomic analysis supports the premise that inhibition of the Warburg effect is involved in shikonin-induced cell death, which is likely further enhanced by dysregulation of glycolysis and the tricarboxylic acid (TCA) cycle, afflicted by ribitol treatment. In conclusion, the present study demonstrates that the metabolite ribitol selectively enhances the cytotoxic effect mediated by shikonin against breast cancer in vitro.
Collapse
Affiliation(s)
- Ravi Doddapaneni
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA
| | | | | | - Qi L. Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Atrium Health, Charlotte, NC 28203, USA
| |
Collapse
|
35
|
Frappaolo A, Zaccagnini G, Giansanti MG. GOLPH3-mTOR Crosstalk and Glycosylation: A Molecular Driver of Cancer Progression. Cells 2025; 14:439. [PMID: 40136688 PMCID: PMC11941073 DOI: 10.3390/cells14060439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/06/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Originally identified in proteomic-based studies of the Golgi, Golgi phosphoprotein 3 (GOLPH3) is a highly conserved protein from yeast to humans. GOLPH3 localizes to the Golgi through the interaction with phosphatidylinositol-4-phosphate and is required for Golgi architecture and vesicular trafficking. Many studies revealed that the overexpression of GOLPH3 is associated with tumor metastasis and a poor prognosis in several cancer types, including breast cancer, glioblastoma multiforme, and colon cancer. The purpose of this review article is to provide the current progress of our understanding of GOLPH3 molecular and cellular functions, which may potentially reveal therapeutic avenues to inhibit its activity. Specifically, recent papers have demonstrated that GOLPH3 protein functions as a cargo adaptor for COP I-coated intra Golgi vesicles and impinges on Golgi glycosylation pathways. In turn, GOLPH3-dependent defects have been associated with malignant phenotypes in cancer cells. Additionally, the oncogenic activity of GOLPH3 has been linked with enhanced signaling downstream of mechanistic target of rapamycin (mTOR) in several cancer types. Consistent with these data, GOLPH3 controls organ growth in Drosophila by associating with mTOR signaling proteins. Finally, compelling evidence demonstrates that GOLPH3 is essential for cytokinesis, a process required for the maintenance of genomic stability.
Collapse
Affiliation(s)
| | | | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy; (A.F.); (G.Z.)
| |
Collapse
|
36
|
Mishan MA, Choo YM, Winkler J, Hamann MT, Karan D. Manzamine A: A promising marine-derived cancer therapeutic for multi-targeted interactions with E2F8, SIX1, AR, GSK-3β, and V-ATPase - A systematic review. Eur J Pharmacol 2025; 990:177295. [PMID: 39863145 DOI: 10.1016/j.ejphar.2025.177295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Manzamine A, a natural compound derived from various sponge genera, features a β-carboline structure and exhibits a range of biological activities, including anti-inflammatory and antimalarial effects. Its potential as an anticancer agent has been explored in several tumor models, both in vitro and in vivo, showing effects through mechanisms such as cytotoxicity, regulation of the cell cycle, inhibition of cell migration, epithelial-to-mesenchymal transition (EMT), autophagy, and apoptosis through multi-target interactions of E2F transcriptional factors, ribosomal S6 kinases, androgen receptor (AR), SIX1, GSK-3β, v-ATPase, and p53/p21/p27 cascades. This systematic review evaluates existing literature on the potential application of this marine alkaloid as a novel cancer therapy, highlighting its promising ability to inhibit cancer cell growth while causing minimal side effects.
Collapse
Affiliation(s)
- Mohammad Amir Mishan
- Department of Urology, Brown Cancer Center, 505 S Hancock Street, Louisville, KY, USA
| | - Yeun-Mun Choo
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Jeffery Winkler
- Department of Chemistry, The University of Pennsylvania, Philadelphia, PA, USA
| | - Mark T Hamann
- Department of Drug Discovery and Biomedical Sciences and Public Health, Colleges of Pharmacy and Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Dev Karan
- Department of Urology, Brown Cancer Center, 505 S Hancock Street, Louisville, KY, USA.
| |
Collapse
|
37
|
Thakur GS, Gupta AK, Pal D, Vaishnav Y, Kumar N, Annadurai S, Jain SK. Designing novel cabozantinib analogues as p-glycoprotein inhibitors to target cancer cell resistance using molecular docking study, ADMET screening, bioisosteric approach, and molecular dynamics simulations. Front Chem 2025; 13:1543075. [PMID: 40084274 PMCID: PMC11903459 DOI: 10.3389/fchem.2025.1543075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/04/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction One of the foremost contributors to mortality worldwide is cancer. Chemotherapy remains the principal strategy for cancer treatment. A significant factor leading to the failure of cancer chemotherapy is the phenomenon of multidrug resistance (MDR) in cancer cells. The primary instigator of MDR is the over expression of P-glycoprotein (P-gp), a protein that imparts resistance and facilitates the ATP-dependent efflux of various anticancer agents. Numerous efforts have been made to inhibit P-gp function with the aim of restoring the effectiveness of chemotherapy due to its broad specificity. The main objective has been to create compounds that either serve as direct P-gp inhibitors or interact with cancer therapies to modulate transport. Despite substantial in vitro achievements, there are currently no approved drugs available that can effectively "block" P-gp mediated resistance. Cabozantinib (CBZ), a multi-kinase inhibitor, is utilized in the treatment of various carcinomas. CBZ has been shown to inhibit P-gp efflux activity, thereby reversing P-gp mediated MDR. Consequently, P-gp has emerged as a critical target for research in anti-cancer therapies. Methods The purpose of this study was to computationally identify new andsafer analogues of CBZ using bioisosteric approach, focusing on improved pharmacokinetic properties andreduced toxicity. The physicochemical, medicinal, and ADMET profiles of generated analogues were computed using the ADMETLab 3.0 server. We also predicted the drug likeness (DL) and drug score (DS) of analogues. The molecular docking studies of screened analogues against the protein (PDB ID: 3G5U) were conducted using AutoDock Vina flowing by BIOVIA Discovery Studio for visualizing interactions.Molecular dynamics (MD) simulation of docked ligands was done using Schrödinger suite. Results and Discussion The docking scores for the ligands CBZ01, CBZ06, CBZ11, CBZ13, CBZ25, CBZ34, and CBZ38 ranged from -8.0 to -6.4 kcal/mol against the protein (PDB ID: 3G5U). A molecular dynamics (MD) simulation of CBZ01, CBZ13, and CBZ38 was conducted using the Schrödinger suite, revealing that these complexesmaintained stability throughout the 100 ns simulation. Conclusion An integrated computational approach combining bioisosteric approach, molecular docking, drug likeness calculations, and MD simulations highlights the promise of ligands CBZ01 and CBZ13 as candidates for the development of potential anticancer agents for the treatment of various cancers.
Collapse
Affiliation(s)
- Gajendra Singh Thakur
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ajay Kumar Gupta
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Dipti Pal
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Yogesh Vaishnav
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles’ College of Pharmacy, Udaipur, Rajasthan, India
| | - Sivakumar Annadurai
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Sanmati Kumar Jain
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
38
|
Easwaran VB, Pai KMS, Pai KSR. Mesenchymal Stem Cell-Derived Exosomes in Cancer Resistance Against Therapeutics. Cancers (Basel) 2025; 17:831. [PMID: 40075675 PMCID: PMC11898417 DOI: 10.3390/cancers17050831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are specialized cells that can differentiate into various types of cells. MSCs can be utilized to treat cancer. However, a MSC is considered a double-edged sword, because it can promote tumor progression and support cancer cell growth. Likewise, MSC-derived exosomes (MSC-Exos) carry various intracellular materials and transfer them to other cells. MSC-Exos could also cause tumor progression, including brain cancer, breast cancer, hepatic cancer, lung cancer, and colorectal cancer, and develop resistance against therapies, mainly chemotherapy, radiotherapy, and immunotherapy. An MSC-Exo promotes tumor development and causes drug resistance in various cancer types. The mechanisms involved in cancer drug resistance vary depending on the cancer cell heterogeneity and complexity. In this article, we have explained the various biomarkers and mechanisms involved in the tumor and resistance development through MSC-Exos in different cancer types.
Collapse
Affiliation(s)
- Vignesh Balaji Easwaran
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - K Maya S Pai
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - K. Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| |
Collapse
|
39
|
Li SB, Zhao TSY, Ye Z, Zou J, Yuan X, Zhou XL, Liang CQ, Li KZ, Huang LZ. Antitumor effects of BPCO on liver cancer cells. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-13. [PMID: 39985778 DOI: 10.1080/10286020.2025.2467318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
Esculetin is a coumarin compound with anticancer, antioxidant, and anti-inflammatory activities. In this study, we synthesized an esculetin derivative, 6,7-bis(Pentyloxy)-2H-Chromen-2-One (BPCO), through etherification. BPCO inhibited the proliferation of HepG2 cells in a dose- and time-dependent manner. It also inhibited cell migration, promoted apoptosis, and caused cell cycle arrest at the G1 phase. Additionally, BPCO downregulated the expression levels of Bcl-2 and Bcl-XL and upregulated the expression levels of Bax and Bak. This study shows that BPCO inhibits hepatocellular carcinoma cell proliferation and induces apoptosis, providing a basis for further study of BPCO as an antitumor agent.
Collapse
Affiliation(s)
- Shan-Bin Li
- College of Biotechnology, Guilin Medical University, Guilin541199, China
- Key Laboratory of Molecular Medical Engineering, Education Department of Guangxi Zhuang Autonomous Region, Guilin541199, China
| | - Tong-Shi-Yao Zhao
- College of Biotechnology, Guilin Medical University, Guilin541199, China
| | - Zhen Ye
- College of Pharmacy, Guilin Medical University, Guilin541199, China
| | - Jian Zou
- College of Biotechnology, Guilin Medical University, Guilin541199, China
| | - Xi Yuan
- College of Biotechnology, Guilin Medical University, Guilin541199, China
| | - Xian-Li Zhou
- College of Biotechnology, Guilin Medical University, Guilin541199, China
- Key Laboratory of Molecular Medical Engineering, Education Department of Guangxi Zhuang Autonomous Region, Guilin541199, China
| | - Cheng-Qin Liang
- College of Pharmacy, Guilin Medical University, Guilin541199, China
| | - Kang-Zhi Li
- College of Biotechnology, Guilin Medical University, Guilin541199, China
| | - Lan-Zhen Huang
- Science Experiment Center, Guilin Medical University, Guilin541199, China
| |
Collapse
|
40
|
Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XHF, Zheng J. Invasion and metastasis in cancer: molecular insights and therapeutic targets. Signal Transduct Target Ther 2025; 10:57. [PMID: 39979279 PMCID: PMC11842613 DOI: 10.1038/s41392-025-02148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
The progression of malignant tumors leads to the development of secondary tumors in various organs, including bones, the brain, liver, and lungs. This metastatic process severely impacts the prognosis of patients, significantly affecting their quality of life and survival rates. Research efforts have consistently focused on the intricate mechanisms underlying this process and the corresponding clinical management strategies. Consequently, a comprehensive understanding of the biological foundations of tumor metastasis, identification of pivotal signaling pathways, and systematic evaluation of existing and emerging therapeutic strategies are paramount to enhancing the overall diagnostic and treatment capabilities for metastatic tumors. However, current research is primarily focused on metastasis within specific cancer types, leaving significant gaps in our understanding of the complex metastatic cascade, organ-specific tropism mechanisms, and the development of targeted treatments. In this study, we examine the sequential processes of tumor metastasis, elucidate the underlying mechanisms driving organ-tropic metastasis, and systematically analyze therapeutic strategies for metastatic tumors, including those tailored to specific organ involvement. Subsequently, we synthesize the most recent advances in emerging therapeutic technologies for tumor metastasis and analyze the challenges and opportunities encountered in clinical research pertaining to bone metastasis. Our objective is to offer insights that can inform future research and clinical practice in this crucial field.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Graduate School of Biomedical Science, Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lijun Deng
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
41
|
Trybus W, Trybus E, Obarzanowski M, Król T. Quinalizarin induces autophagy, apoptosis and mitotic catastrophe in cervical and prostate cancer cells. Sci Rep 2025; 15:5252. [PMID: 39939343 PMCID: PMC11822151 DOI: 10.1038/s41598-025-89847-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/07/2025] [Indexed: 02/14/2025] Open
Abstract
Cancer diseases are a serious health problem for society, and among them cervical and prostate cancer rank high in terms of mortality. One of the reasons is the phenomenon of drug resistance and side effects accompanying conventional chemo- and radiotherapy. This requires continuous development of alternative treatment methods and searching for new compounds with anti-cancer potential. An example is quinalizarin, which was tested for its anti-cancer potential. The MTT test showed cytotoxic activity of quinalizarin against Hela and DU145 cell lines. Morphological analysis showed nuclear changes typical of apoptosis, which was confirmed by the annexin V/PE test, activation of caspases 3/7 and inhibition of Bcl-2 protein expression. Increased permeability of mitochondrial membranes and ROS generation were demonstrated. Inhibition of cell migration, blocking in the G0/G1 phase, increased number of cells with damaged DNA and an increase in markers of mitotic catastrophe, i.e. micro- and multinucleation including the presence of abnormal mitotic figures were also observed. At the same time, increased autophagy was observed, and preincubation of cells with chloroquine inhibited this process, which contributed to the increased cytotoxicity of quinalizarin towards the tested cells. Quinalizarin has a multidirectional effect based on apoptosis and alternative types of cell death.
Collapse
Affiliation(s)
- Wojciech Trybus
- Department of Medical Biology, Jan Kochanowski University of Kielce, Uniwersytecka 7, Kielce, 25-406, Poland.
| | - Ewa Trybus
- Department of Medical Biology, Jan Kochanowski University of Kielce, Uniwersytecka 7, Kielce, 25-406, Poland.
| | - Mateusz Obarzanowski
- Department of Oncology, Medical College, Jan Kochanowski University of Kielce, al. IX Wieków Kielc 19a, Kielce, 25-516, Poland
- Department of Urology, Holy Cross Cancer Center, Stefana Artwińskiego, Kielce, 25-734, Poland
| | - Teodora Król
- Department of Medical Biology, Jan Kochanowski University of Kielce, Uniwersytecka 7, Kielce, 25-406, Poland
| |
Collapse
|
42
|
Radhakrishnan A, Shanmukhan NK, Samuel LC. Pharmacogenomics influence on MDR1-associated cancer resistance and innovative drug delivery approaches: advancing precision oncology. Med Oncol 2025; 42:67. [PMID: 39913003 DOI: 10.1007/s12032-025-02611-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025]
Abstract
Currently, there is a growing concern surrounding the treatment of cancer, a formidable disease. Pharmacogenomics and personalized medicine have emerged as significant areas of interest in cancer management. The efficacy of many cancer drugs is hindered by resistance mechanisms, particularly P-glycoprotein (P-gp) efflux, leading to reduced therapeutic outcomes. Efforts have intensified to inhibit P-gp efflux, thereby enhancing the effectiveness of resistant drugs. P-gp, a member of the ATP-binding cassette (ABC) superfamily, specifically the multidrug resistance (MDR)/transporter associated with antigen processing (TAP) sub-family B, member 1, utilizes energy derived from ATP hydrolysis to drive efflux. This review focuses on genetic polymorphisms associated with P-gp efflux and explores various novel pharmaceutical strategies to address this challenge. These strategies encompass SEDDS/SNEDDS, liposomes, immunoliposomes, solid lipid nanoparticles, lipid core nanocapsules, microemulsions, dendrimers, hydrogels, polymer-drug conjugates, and polymeric nanoparticles. The article aims to elucidate the interplay between pharmacogenomics, P-gp-mediated drug resistance in cancer, and formulation strategies to improve cancer therapy by tailoring formulations to genetically susceptible patients.
Collapse
Affiliation(s)
- Arun Radhakrishnan
- Department of Pharmaceutics, JKKN College of Pharmacy, Kumarapalayam, Tamil Nadu, 638183, India.
| | - Nikhitha K Shanmukhan
- Department of Pharmaceutics, JKKN College of Pharmacy, Kumarapalayam, Tamil Nadu, 638183, India
| | - Linda Christabel Samuel
- Department of Conservative Dentistry and Endodontics, JKKN Dental College and Hospitals, Kumarapalayam, 638183, India
| |
Collapse
|
43
|
Wang M, Zhang Y, Ni S, Sun M, Wu Q, Wu X, Chen Q, Wang S. The anti-cancer activity of Dioscin: an update and future perspective. Med Oncol 2025; 42:63. [PMID: 39899128 PMCID: PMC11790812 DOI: 10.1007/s12032-024-02572-6] [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: 10/05/2024] [Accepted: 11/18/2024] [Indexed: 02/04/2025]
Abstract
Natural drugs have the advantages of multi-pathway, multi-target, low toxicity, and high efficiency, which make them widely used and effective in anti-tumor therapy. Dioscin is a steroidal saponin compound that can be extracted from Dioscaceae plants. In recent years, it has been found that Dioscin has potent anti-tumor effects, can inhibit tumor cell proliferation, induce apoptosis and autophagy, inhibits tumor cell metastasis, reverses multidrug resistance, and increases sensitivity to anticancer drugs, and thus inhibit tumor progression. Meanwhile, the construction of Dioscin nanocarriers can improve the efficiency of drug use, reduce drug toxicity, realize the precise delivery of drugs, and improve the bioavailability of Dioscin. In this paper, the anticancer mechanism and targets of Dioscin in recent years were reviewed, thereby providing new ideas and a theoretical basis for further research and promotion of Dioscin.
Collapse
Affiliation(s)
- MengYue Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - YaNan Zhang
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
- Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - SongLin Ni
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Mo Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - QiaoLan Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - XiaoLin Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Qian Chen
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
- Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| | - ShiJun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
44
|
Ferdousmakan S, Mansourian D, Seyedi Asl FS, Fathi Z, Maleki-Sheikhabadi F, Afjadi MN, Zalpoor H. Autophagy induced by metabolic processes leads to solid tumor cell metastatic dormancy and recurrence. Med Oncol 2025; 42:62. [PMID: 39899220 DOI: 10.1007/s12032-025-02607-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
A crucial cellular mechanism that has a complex impact on the biology of cancer, particularly in solid tumors, is autophagy. This review explores how metabolic processes trigger autophagy, which helps metastatic tumor cells go dormant and recur. During metastasis, tumor cells frequently encounter severe stressors, such as low oxygen levels and nutritional deprivation, which causes them to activate autophagy as a survival tactic. This process allows cancer stem cells (CSCs) to withstand severe conditions while also preserving their features. After years of dormancy, dormant disseminated tumor cells (DTCs) may reappear as aggressive metastatic cancers. The capacity of autophagy to promote resistance to treatments and avoid immune detection is intimately related to this phenomenon. According to recent research, autophagy promotes processes, such as the epithelial-to-mesenchymal transition (EMT) and helps build a pre-metastatic niche, which makes treatment strategies more challenging. Autophagy may be a promising therapeutic target because of its dual function as a tumor suppressor in early-stage cancer and a survival promoter in advanced stages. To effectively treat metastatic diseases, it is crucial to comprehend how metabolic processes interact with autophagy and affect tumor behavior. In order to find novel therapeutic approaches that can interfere with these processes and improve patient outcomes, this study highlights the critical need for additional investigation into the mechanisms by which autophagy controls tumor dormancy and recurrence.
Collapse
Affiliation(s)
- Saeid Ferdousmakan
- Department of Pharmacy Practice, Nargund College of Pharmacy, Bangalore, 560085, India
| | - Dorrin Mansourian
- Faculty of Pharmacy, Eastern Mediterranean University, Gazimagusa TRNC via Mersin 10, Mersin, Turkey
| | | | - Zeinab Fathi
- Medical School, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
45
|
Hamdy NM, Zaki MB, Abdelmaksoud NM, Ismail RA, Abd-Elmawla MA, Rizk NI, Fathi D, Abulsoud AI. Insights into the genetic and epigenetic mechanisms governing X-chromosome-linked-miRNAs expression in cancer; a step-toward ncRNA precision. Int J Biol Macromol 2025; 289:138773. [PMID: 39675615 DOI: 10.1016/j.ijbiomac.2024.138773] [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: 12/04/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
Sex chromosomes play a significant role in establishing sex-specific differences in gene expression, thereby contributing to phenotypic diversity and susceptibility to various diseases. MicroRNAs (miRNAs), which are small non-coding RNAs encoded by both the X and Y chromosomes, exhibit sex-specific regulatory characteristics. Computational analysis has identified several X-linked miRNAs differentially expressed in sex-specific cancers. This review aims to elucidate the genetic and epigenetic mechanisms that govern the sex-specific expression of X- and Y-linked miRNAs, with particular attention to their functional role in regulating diverse cellular processes in different cancer pathways. In addition, this review provides a comprehensive understanding of the targeted therapeutic interventions and critical insights into the potential clinical implications of targeting sex-specific miRNAs. In conclusion, this review opens new horizons for further research to effectively translate these findings into viable treatment options.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo 11566, Egypt.
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | | | - Rehab A Ismail
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Kasr Al-Ainy, Cairo 11562, Egypt
| | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo 11786, Egypt
| | - Doaa Fathi
- Department of Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21526, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al Azhar University, Nasr City, Cairo 11231, Egypt
| |
Collapse
|
46
|
Chen C, Wang M, Tu D, Cao J, Zhang C, Bai D. Roles of anoikis in hepatocellular carcinoma: mechanisms and therapeutic potential. Med Oncol 2025; 42:58. [PMID: 39885089 DOI: 10.1007/s12032-025-02612-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver cancer, is a highly aggressive malignancy with limited viable therapeutic options. For early HCC, resection surgery is currently the most effective treatment. However, in advanced stages, resection alone does not sufficiently address the disease, so finding a method with a better prognosis is necessary. Anoikis, known as matrix detachment-induced apoptosis or detachment-induced cell death, is crucial for tissue development and homeostasis. Cancer cells develop means to evade anoikis, e.g. anoikis resistance, thereby allowing for cells to survive under anchorage-independent conditions. HCC cells often acquire resistance to anoikis, allowing them to survive after detaching from the extracellular matrix and contributing to tumor spread. This review discusses the mechanisms of anoikis in HCC, exploring the potential of drug-induced anoikis and targeting anoikis resistance as promising therapeutic strategies for treating HCC, analyzing the value of anoikis in the immune of HCC, and propose potential pathways in oncotherapy, which can provide background knowledge for subsequent related research.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Mengyao Wang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Daoyuan Tu
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Jun Cao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Chi Zhang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Dousheng Bai
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, China.
| |
Collapse
|
47
|
Stasiak P, Sopel J, Lipowicz JM, Rawłuszko-Wieczorek AA, Korbecki J, Januchowski R. The Role of Elacridar, a P-gp Inhibitor, in the Re-Sensitization of PAC-Resistant Ovarian Cancer Cell Lines to Cytotoxic Drugs in 2D and 3D Cell Culture Models. Int J Mol Sci 2025; 26:1124. [PMID: 39940891 PMCID: PMC11817197 DOI: 10.3390/ijms26031124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Chemotherapy resistance is a significant barrier to effective cancer treatment. A key mechanism of resistance at the single-cell level is the overexpression of drug transporters in the ABC family, particularly P-glycoprotein (P-gp), which leads to multidrug resistance (MDR). Inhibitors of these transporters can help re-sensitize cancer cells to chemotherapeutics. This study evaluated elacridar (GG918 and GF120918), a potent third-generation P-gp inhibitor, for its ability to reverse MDR in paclitaxel (PAC)-resistant ovarian cancer cell lines. Sensitive and PAC-resistant cells were cultured in two-dimensional (2D) and three-dimensional (3D) models. MDR1 gene expression was analyzed using Q-PCR, and P-gp protein expression was examined via Western blot and immunofluorescence. Drug sensitivity was evaluated with MTT assays, and P-gp activity was analyzed by flow cytometry and fluorescence microscopy. Elacridar effectively inhibited P-gp activity and increased sensitivity to PAC and doxorubicin (DOX) in 2D cultures but not cisplatin (CIS). In 3D spheroids, P-gp activity inhibition was observed via Calcein-AM staining. However, no re-sensitization to PAC occurred and limited improvement was observed for DOX. These findings suggest that elacridar effectively inhibits P-gp in both 2D and 3D conditions. However, its ability to overcome drug resistance in 3D models is limited, highlighting the complexity of tissue-specific resistance mechanisms.
Collapse
Affiliation(s)
- Piotr Stasiak
- Institute of Biological Sciences, University of Zielona Góra, 65-417 Zielona Góra, Poland
- The Doctoral School of Exact and Technical Sciences, University of Zielona Góra, 65-417 Zielona Góra, Poland
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.S.); (J.K.); (R.J.)
| | - Justyna Sopel
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.S.); (J.K.); (R.J.)
| | - Julia Maria Lipowicz
- Department of Histology and Embryology, Doctoral School, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | | | - Jan Korbecki
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.S.); (J.K.); (R.J.)
| | - Radosław Januchowski
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.S.); (J.K.); (R.J.)
| |
Collapse
|
48
|
Ghai S, Shrestha R, Su KH. HSF1 at the crossroads of chemoresistance: from current insights to future horizons in cell death mechanisms. Front Cell Dev Biol 2025; 12:1500880. [PMID: 39850800 PMCID: PMC11754285 DOI: 10.3389/fcell.2024.1500880] [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: 09/24/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025] Open
Abstract
Heat Shock Factor 1 (HSF1) is a major transcriptional factor regulating the heat shock response and has become a potential target for overcoming cancer chemoresistance. This review comprehensively examines HSF1's role in chemoresistance and its potential as a therapeutic target in cancer. We explore the complex, intricate mechanism that regulates the activation of HSF1, HSF1's function in promoting resistance to chemotherapy, and the strategies used to manipulate HSF1 for therapeutic benefit. In addition, we discuss emerging research implicating HSF1's roles in autophagy, apoptosis, DNA damage repair, drug efflux, and thus chemoresistance. This article highlights the significance of HSF1 in cancer chemoresistance and its potential as a target for enhancing cancer treatment efficacy.
Collapse
Affiliation(s)
| | | | - Kuo-Hui Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, United States
| |
Collapse
|
49
|
Rodrigues JAO, Kiran NS, Chatterjee A, Prajapati BG, Dhas N, Dos Santos AO, de Sousa FF, Souto EB. Metallodrugs: Synthesis, mechanism of action and nanoencapsulation for targeted chemotherapy. Biochem Pharmacol 2025; 231:116644. [PMID: 39577705 DOI: 10.1016/j.bcp.2024.116644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
As a multifactorial and heterogeneous disease, cancer has a high mortality rate, and the search for more effective treatments is an enormous challenge. Metal coordination compounds open a range of possibilities that conventional organic and biological molecules can no longer fulfil due to increasing drug resistance. Metallodrugs still have tremendous potential to help overcome drug resistance and find new cures in medicine, considering that at least 25 metallic elements participate in healthy functioning of the human body. Transition metal ions, such as copper, zinc and iron, are incorporated into catalytic proteins, the so-called metalloenzymes, which participate in various chemical reactions necessary for life. The interaction of metal complexes in different pathways with the structural richness of deoxyribonucleic acid encouraged to seek to understand the mechanisms of action and overcome the obstacles encountered for a promising future of these drugs. The success of platinum-based metallodrugs is one of the great inspirations for the search of new metallodrugs, although the approval of these molecules has been slow in recent years due to the risk of systemic toxicity and insufficient understanding of their mechanisms. To overcome the clinical limitations encountered in some metallodrugs, nanoencapsulation has been proposed as a new approach to improve therapeutic index in chemotherapy. The remarkable selectivity of nanoencapsulated metallodrugs and their enhanced capacity to bypass various biological barriers allow site-specific targeting. In this review, we present the advances in the development and use of the most relevant metallodrugs, and new delivery approaches, in the fight against cancer.
Collapse
Affiliation(s)
- Jessica A O Rodrigues
- Center for Social Sciences, Health and Technology, Federal University of Maranhão (UFMA), 65900-410 Imperatriz, MA, Brazil.
| | - Neelakanta S Kiran
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Ankita Chatterjee
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Bhupendra G Prajapati
- Shree. S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana 384012, Gujarat, India; Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Adenilson O Dos Santos
- Center for Social Sciences, Health and Technology, Federal University of Maranhão (UFMA), 65900-410 Imperatriz, MA, Brazil
| | - Francisco F de Sousa
- Center for Social Sciences, Health and Technology, Federal University of Maranhão (UFMA), 65900-410 Imperatriz, MA, Brazil; Institute of Exact and Natural Sciences, Federal University of Pará (UFPA), 66075-110, Belem, PA, Brazil
| | - Eliana B Souto
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland.
| |
Collapse
|
50
|
Shen M, Ma Z, Zhu J, Wen G, Jin H, An J, Tuo B, Liu X, Li T. CDK4/6 inhibitors in HR-positive breast cancer immunotherapy. Bioorg Chem 2025; 154:108095. [PMID: 39732088 DOI: 10.1016/j.bioorg.2024.108095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024]
Abstract
Breast cancer is the most prevalent malignant tumour among women. Approximately 70 % of patients are hormone receptor (HR)-positive and undergo endocrine therapy as the main form of treatment; however, the efficacy of this type of therapy is limited by some factors, such as drug resistance and complex tumour microenvironments. Using network pharmacology and molecular docking, this study examined how CDK4/6 inhibitors enhance the effects of immunotherapy for HR-positive breast cancer, focusing on their effects on the tumour microenvironment (TME) and immune cell activity. CDK4/6 inhibitor influence crucial signalling pathways, including TP53 and EGFR, underscoring their importance in modulating immune responses and combating drug resistance. Our analysis revealed that the combination of CDK4/6 inhibitors with other immunotherapies, such as immune checkpoint inhibitors, might play an important role in improving the effectiveness of treatment in patients with cancer. This study provides new perspectives on treatment options for HR-positive breast cancers, promoting ongoing research and improvements in therapeutic approaches.
Collapse
Affiliation(s)
- Mimi Shen
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaxing Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|