1
|
Chavan PR, Pandey R, Patil BM, Murti K, Kumar N. Unravelling key signaling pathways for the therapeutic targeting of non-small cell lung cancer. Eur J Pharmacol 2025; 998:177494. [PMID: 40090536 DOI: 10.1016/j.ejphar.2025.177494] [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: 12/11/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
Lung cancer (LC) remains the foremost cause of cancer-related mortality across the globe. Non-small cell lung cancer (NSCLC) is a type of LC that exhibits significant heterogeneity at histological and molecular levels. Genetic alterations in upstream signaling molecules activate cascades affecting apoptosis, proliferation, and differentiation. Disruption of these signaling pathways leads to the proliferation of cancer-promoting cells, progression of cancer, and resistance to its treatment. Recent insights into the function of signaling pathways and their fundamental mechanisms in the onset of various diseases could pave the way for new therapeutic approaches. Recently, numerous drug molecules have been created that target these cell signaling pathways and could be used alongside other standard therapies to achieve synergistic effects in mitigating the pathophysiology of NSCLC. Additionally, many researchers have identified several predictive biomarkers, and alterations in transcription factors and related pathways are employed to create new therapeutic strategies for NSCLC. Findings suggest using specific inhibitors to target cellular signaling pathways in tumor progression to treat NSCLC. This review investigates the role of signaling pathways in NSCLC development and explores novel therapeutic strategies to enhance clinical treatment options for NSCLC.
Collapse
Affiliation(s)
- Pavan Ramrao Chavan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Ruchi Pandey
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Baswant Malesh Patil
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Nitesh Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India.
| |
Collapse
|
2
|
Thonusin C, Khuanjing T, Nawara W, Chattipakorn SC, Chattipakorn N. Alpha-7 nicotinic and muscarinic acetylcholine receptor agonists promote a favorable pattern of cardiac metabolic reprogramming in doxorubicin-induced heart failure rats. Arch Biochem Biophys 2025; 769:110427. [PMID: 40252789 DOI: 10.1016/j.abb.2025.110427] [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: 02/23/2025] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Sympathetic hyperactivation and metabolic reprogramming are found in heart failure. Parasympathetic activation by acetylcholine receptor agonists attenuates doxorubicin-induced heart failure by improving mitochondrial function and ameliorating apoptosis and inflammation. However, the effect of these agents on cardiac metabolic reprogramming in doxorubicin-induced heart failure has never been investigated. Male Wistar rats received either vehicle, 6 doses of 3 mg/kg/day of doxorubicin, 6 doses of 3 mg/kg/day of doxorubicin and 3 mg/kg/day of an alpha-7 nicotinic acetylcholine receptor agonist for 30 days, or 6 doses of 3 mg/kg/day of doxorubicin and 12 mg/kg/day of a muscarinic acetylcholine receptor agonist for 30 days. Then, the rats were euthanized to collect heart and serum for metabolomics study. Doxorubicin caused increased glycolysis, increased ketone body utilization, decreased fat utilization, decreased succinate oxidation, and decreased adenosine triphosphate production. Co-treatment with acetylcholine receptor agonist ameliorated an increase in glycolysis, and restored fat utilization, succinate oxidation, and adenosine triphosphate production in the heart. Metabolome alterations in serum were consistent with those in the heart. Our findings highlighted the roles of metabolomics in identifying cardiac metabolic reprogramming and emphasized the potential of acetylcholine receptor agonist in promoting a favorable pattern of cardiac metabolic reprogramming in doxorubicin-induced heart failure.
Collapse
Affiliation(s)
- Chanisa Thonusin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
3
|
Cao X, Zhao L, Zhou J, Ding S, Sun Y, Ma Y, Ma Z, Liu H, Dong T, Luo A, Li Y, Fang B. Dexmedetomidine inhibits ferroptosis through the Akt/GSK3β/Nrf2 axis and alleviates adriamycin-induced cardiotoxicity. Life Sci 2025; 371:123609. [PMID: 40187641 DOI: 10.1016/j.lfs.2025.123609] [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: 01/16/2025] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The cardiotoxicity of Adriamycin(ADR) limits its clinical application, and its molecular mechanism is not very clear. At present, Dexrazoxane (DXZ) is the only approved drug to prevent ADR-induced cardiotoxicity (DIC), but it also has serious adverse reactions. Therefore, it is a key scientific challenge to find a drug with strong myocardial protection, few adverse reactions and no effect on the anti-tumor effect of ADR. In this study, we established the DIC model in rats. Cardiomyocyte hypertrophy and myocardial fibrosis increased significantly, and MDA and LDH increased significantly in serum. Dexmedetomidine (DEX) is a carbohydrate with multiple biological activities that can significantly improve the above DIC process. Echocardiography confirmed that DEX could reverse the changes of ESV, EDV, EF and FS induced by ADR. In vitro, experiments confirmed that DEX reversed the upregulation of ANP, BNP, MHC and Collagen III protein levels induced by ADR. DEX improves DIC by inhibiting ferroptosis. Erastin, a ferroptosis agonist, confirmed that DEX improved DIC by inhibiting ferroptosis. Mechanically, DEX increases the expression of Nrf2 in the nucleus through the Akt/Gsk3β signalling axis, thereby regulating ferroptosis in cardiomyocytes. In addition, DEX can improve DIC while not affecting the anti-tumor effect of ADR.
Collapse
Affiliation(s)
- Xuefeng Cao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China; Department of Anesthesiology, Affiliated Hospital of Chengde Medical College, Chengde, China; Hebei Key Laboratory of Panvascular Diseasesr, Hebei, China
| | - Liang Zhao
- Department of Basic Medicine, Chengde Medical College, Chengde, China; Hebei Key Laboratory of Nerve Injury and Repair, Hebei, China; Hebei Key Laboratory of Panvascular Diseasesr, Hebei, China; Chengde Medical University Discipline Construction Funds, Chengde Medical College, Hebei, China
| | - Jian Zhou
- Department of Basic Medicine, Chengde Medical College, Chengde, China
| | - Shi Ding
- Department of Basic Medicine, Chengde Medical College, Chengde, China
| | - Ying Sun
- Department of Basic Medicine, Chengde Medical College, Chengde, China
| | - Yang Ma
- Department of Anesthesiology, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Zijian Ma
- Department of Anesthesiology, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Hancheng Liu
- Department of Anesthesiology, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Tianxin Dong
- Department of Anesthesiology, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Aijing Luo
- Department of Anesthesiology, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Yan Li
- Department of Anesthesiology, Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
4
|
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 DOI: 10.1038/s41420-025-02554-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [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
|
5
|
Masoumi S, Aleyasin SA, Faghihi S. Albumin nanoparticles-mediated doxorubicin delivery enhances the anti-tumor efficiency in ovarian cancer cells through controlled release. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6885-6900. [PMID: 39692901 DOI: 10.1007/s00210-024-03730-3] [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: 06/28/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
Doxorubicin (DOX) is an anthracycline commonly used as a first-line treatment option for various malignancies, either as a stand-alone treatment or in combination with other chemotherapeutic agents. However, its efficacy in advanced cancer stages requires high doses, resulting in significant cytotoxicity to normal cells and severe side effects. Nanotechnology offers a promising strategy to mitigate these drawbacks through controlled drug release. In this study, bovine serum albumin nanoparticles (BSA-NPs) were synthesized via the desolvation method and successfully loaded with DOX (DOX-BSA-NPs). Characterization using dynamic light scattering, scanning electron microscopy, Fourier-transform infrared spectroscopy, UV-visible spectroscopy, and high-performance liquid chromatography confirmed efficient drug loading. In vitro studies demonstrated that DOX-BSA-NPs enabled sustained drug release and enhanced intracellular delivery. After treatment with DOX-BSA-NPs, ovarian cancer cells showed a twofold increase in cytotoxicity compared to free DOX. Scratch assays further revealed a significant reduction in cancer cell migration and invasion. Additionally, LDH assays and Annexin V-FITC flow cytometry indicated a shift toward apoptosis over necrosis, enhancing the anti-tumor efficacy of DOX. This was supported by increased reactive oxygen species production, upregulation of pro-apoptotic genes, downregulation of anti-apoptotic genes, and elevated caspase 3 and 7 activity, collectively promoting apoptosis. These findings underscore the potential of DOX-BSA-NPs as a superior alternative for targeted and controlled drug delivery, offering enhanced therapeutic efficacy and reduced side effects in ovarian cancer treatment.
Collapse
Affiliation(s)
- Sajjad Masoumi
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Seyed Ahmad Aleyasin
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| | - Shahab Faghihi
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
6
|
Yokoyama T, Saitoh M, Miyazawa K. TGF-β Enhances Doxorubicin Resistance and Anchorage-Independent Growth in Cancer Cells by Inducing ALDH1A1 Expression. Cancer Sci 2025. [PMID: 40414259 DOI: 10.1111/cas.70109] [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: 01/07/2025] [Revised: 04/30/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
The transforming growth factor-β (TGF-β)/Smad signaling pathway promotes malignant transformation through various mechanisms, and its effect on enhancing drug resistance can limit the efficacy of treatment. Here, we showed that pre-stimulation of human lung cancer A549 cells with TGF-β increases resistance to doxorubicin-induced growth inhibition in a Smad3- and Smad4-dependent manner. This effect was suppressed by the aldehyde dehydrogenase (ALDH) inhibitor oxyfedrine, suggesting that ALDH family members are involved in drug resistance. TGF-β upregulated the mRNA and protein expression of ALDH1A1. The TGF-β/Smad3 transcriptional enhancer region on ALDH1A1 was identified by Smad3 ChIP-seq analysis using an open database and by reporter assays. Knockdown of ALDH1A1 in A549 cells suppressed TGF-β-induced doxorubicin resistance, and lentivirus-mediated introduction of ALDH1A1 into A549 SMAD3-KO cells restored drug resistance. We also demonstrated that ALDH1A1 is required and sufficient for TGF-β/Smad3 signaling-induced anchorage-independent growth. The results suggest that the TGF-β/Smad3/4 axis promotes resistance to doxorubicin and anchorage-independent growth by inducing the transcription of ALDH1A1.
Collapse
Affiliation(s)
- Takashi Yokoyama
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Masao Saitoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Chuo, Japan
- Center for Medical Education and Sciences, Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Keiji Miyazawa
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| |
Collapse
|
7
|
Hu R, Duan Z, Wang M, Liu M, Zhang Y, Lu Y, Qian Y, Wei E, Feng J, Guo P, Chen Y. Stable isotope tracing reveals glucose metabolism characteristics of drug-resistant B-cell acute lymphoblastic leukemia. Anal Chim Acta 2025; 1352:343884. [PMID: 40210293 DOI: 10.1016/j.aca.2025.343884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/26/2024] [Accepted: 03/02/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Adult B-cell acute lymphocytic leukemia (B-ALL) is a malignant hematologic tumor characterized by the uncontrolled proliferation of B-cell lymphoblasts in the bone marrow. Despite advances in treatment, including chemotherapy and consolidation therapy, many B-ALL patients experience unfavorable prognoses due to the development of drug resistance. The precise mechanisms governing chemotherapy resistance, particularly those related to metabolic reprogramming within tumors, remain inadequately elucidated. RESULTS Nalm6/DOX cells exhibited significantly elevated levels of glucose, pyruvate, alanine, glutamine, and glycine compared to Nalm6 cells. Conversely, reduced levels of citrate, acetate, and leucine were observed in Nalm6/DOX cells. Upon exposure to the culture medium supplemented with tracer 13C6-glucose, the Nalm6/DOX cells showed an increase in the abundance of 13C-alanine and a decrease in the levels of 13C-lactate, indicating impaired utilization of 13C-pyruvate. Combining β-chloro-alanine (ALTi) with DOX could decrease the drug resistance phenotype of Nalm6/DOX cells. The results demonstrated that glycolysis and tricarboxylic acid cycle were suppressed in Nalm6/DOX cells, while metabolic flux through the alanine and glutamine pathways was increased. Therefore, inhibition of alanine biosynthesis in Nalm6/DOX exhibits the potential to reverse drug resistance. SIGNIFICANCE A new insight into the impact of metabolism on chemotherapy resistance in B-ALL has been gained through the use of stable isotope resolved metabolomics based on nuclear magnetic resonance and ultra-performance liquid chromatography/tandem mass spectrometry. This provides promising ways for the development of innovative therapeutic strategies to alleviate drug resistance and relapse in affected patients.
Collapse
Affiliation(s)
- Rong Hu
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China; Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fujian Medical University, Fuzhou, 350122, China; Institute of Precision Medicine, Fujian Medical University, Fuzhou, 350004, China
| | - Zhengwei Duan
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China; Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Mengyao Wang
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China; Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fujian Medical University, Fuzhou, 350122, China; Institute of Precision Medicine, Fujian Medical University, Fuzhou, 350004, China
| | - Mengting Liu
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China
| | - Yaoxin Zhang
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China
| | - Yanxi Lu
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China
| | - Yuhan Qian
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China
| | - Enjie Wei
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China
| | - Jianghua Feng
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, 361005, China
| | - Pengfei Guo
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, 361005, China
| | - Yang Chen
- Department of Laboratory Medicine, Fujian Medical University, Fuzhou, 350122, China; Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fujian Medical University, Fuzhou, 350122, China; Institute of Precision Medicine, Fujian Medical University, Fuzhou, 350004, China.
| |
Collapse
|
8
|
Bhatt KS, Singh A, Marwaha GS, Ravendranathan N, Sandhu IS, Kim K, Singh E, Frisbee JC, Singh KK. Different Mechanisms in Doxorubicin-Induced Neurotoxicity: Impact of BRCA Mutations. Int J Mol Sci 2025; 26:4736. [PMID: 40429877 PMCID: PMC12111927 DOI: 10.3390/ijms26104736] [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: 04/07/2025] [Revised: 04/30/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
The genotoxic drug doxorubicin (Dox) remains one of the most powerful chemotherapeutic options available for a wide range of cancers including breast, ovarian, and other cancers. However, emerging evidence links Dox treatment with chemotherapy-induced cognitive impairment, a condition that is popularly referred to as Dox-induced neurotoxicity or "chemobrain", which limits the use of the drug. There are no specific treatments for Dox-induced neurotoxicity, only interventions to mitigate the neurotoxic effects of the drug. Accumulating evidence indicates that DNA damage, oxidative stress, dysregulation of autophagy and neurogenesis, inflammation, and apoptosis play central roles in Dox-induced neurotoxicity. Additionally, germline mutations in the tumour suppressor genes breast cancer susceptibility genes 1 and 2 (BRCA1 and BRCA2) increase the risk of breast, ovarian, and related cancers. BRCA1 and BRCA2 are distinct proteins that play crucial, unique roles in homologous recombination-mediated double-stranded break repair. Furthermore, BRCA1 and 2 mitigate oxidative stress in both neural cells and brain microvascular endothelial cells, which suggests that they have a critical role as regulators of pathways central to the development of Dox-induced neurotoxicity. Despite research on the effects of Dox on cognitive function, there is a gap in knowledge about the role of BRCA1 and BRCA2 in Dox-induced neurotoxicity. In this review, we discuss existing findings about the role of different mechanisms and the role of BRCA1 and BRCA2 in Dox-induced neurotoxicity, along with future perspectives.
Collapse
Affiliation(s)
- Kriti S. Bhatt
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (K.S.B.); (A.S.); (G.S.M.); (N.R.); (I.S.S.); (K.K.); (J.C.F.)
| | - Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (K.S.B.); (A.S.); (G.S.M.); (N.R.); (I.S.S.); (K.K.); (J.C.F.)
| | - Gursharan S. Marwaha
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (K.S.B.); (A.S.); (G.S.M.); (N.R.); (I.S.S.); (K.K.); (J.C.F.)
| | - Naresh Ravendranathan
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (K.S.B.); (A.S.); (G.S.M.); (N.R.); (I.S.S.); (K.K.); (J.C.F.)
| | - Inderbir S. Sandhu
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (K.S.B.); (A.S.); (G.S.M.); (N.R.); (I.S.S.); (K.K.); (J.C.F.)
| | - Kristen Kim
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (K.S.B.); (A.S.); (G.S.M.); (N.R.); (I.S.S.); (K.K.); (J.C.F.)
| | - Eesha Singh
- London Central Secondary School, London, ON N6B 2P8, Canada;
| | - Jefferson C. Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (K.S.B.); (A.S.); (G.S.M.); (N.R.); (I.S.S.); (K.K.); (J.C.F.)
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (K.S.B.); (A.S.); (G.S.M.); (N.R.); (I.S.S.); (K.K.); (J.C.F.)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
9
|
Suman I, Jezidžić A, Dobrić D, Domitrović R. Differential Effects of Rutin and Its Aglycone Quercetin on Cytotoxicity and Chemosensitization of HCT 116 Colon Cancer Cells to Anticancer Drugs 5-Fluorouracil and Doxorubicin. BIOLOGY 2025; 14:527. [PMID: 40427716 PMCID: PMC12109564 DOI: 10.3390/biology14050527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/28/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Rutin and quercetin are natural flavonoids with a variety of beneficial health effects, including anticancer activity. In the present study, we compared cytotoxicity and chemosensitization of human colon cancer HCT116 cells to anticancer drugs 5-fluorouracil (5-FU) and doxorubicin (DOX) by both compounds. METHODS The 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT) test was used to determine cell viability. Western blot and immunofluorescence techniques were employed in the detection of expression of proteins involved in oxidative stress, apoptosis, and autophagy. RESULTS Quercetin treatment resulted in reduced cell viability compared to rutin at the same dose, suggesting greater cytotoxicity than rutin against HCT116 cells. Quercetin was also a better chemosensitizer of DOX than rutin, further reducing cell viability. However, rutin was a better chemosensitizer of 5-FU than quercetin. All treatments induced apoptosis, with rutin and DOX inducing intrinsic and 5-FU inducing extrinsic apoptotic cell death. Autophagy was induced in all treatments and played a pro-survival role, with the exception of DOX treatment. Different treatment regimens specifically modulated cancer cell signaling pathways involved in the regulation of oxidative stress, apoptosis, and autophagy. CONCLUSIONS The results of the current study suggest that rutin and quercetin, although structural analogs, act as specific modulators of signaling pathways in cancer cells, differentially affecting cancer cell cytotoxicity and chemosensitization to anticancer drugs, based on the presence of a free hydroxyl group at the C-3 position of the flavonoid backbone at quercetin or rutinose in rutin.
Collapse
Affiliation(s)
- Iva Suman
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (I.S.)
| | - Alberta Jezidžić
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Dorotea Dobrić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (I.S.)
| | - Robert Domitrović
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (I.S.)
| |
Collapse
|
10
|
Liu G, Liu J, Li S, Zhang Y, He R. Exosome-Mediated Chemoresistance in Cancers: Mechanisms, Therapeutic Implications, and Future Directions. Biomolecules 2025; 15:685. [PMID: 40427578 PMCID: PMC12108986 DOI: 10.3390/biom15050685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Chemotherapy resistance represents a formidable obstacle in oncological therapeutics, substantially compromising the efficacy of adjuvant chemotherapy regimens and contributing to unfavorable clinical prognoses. Emerging evidence has elucidated the pivotal involvement of exosomes in the dissemination of chemoresistance phenotypes among tumor cells and within the tumor microenvironment. This review delineates two distinct intra-tumoral resistance mechanisms orchestrated by exosomes: (1) the exosome-mediated sequestration of chemotherapeutic agents coupled with enhanced drug efflux in neoplastic cells, and (2) the horizontal transfer of chemoresistance to drug-sensitive cells through the delivery of bioactive molecular cargo, thereby facilitating the propagation of resistance phenotypes across the tumor population. Furthermore, the review covers current in vivo experimental data focusing on targeted interventions against specific genetic elements and exosomal secretion pathways, demonstrating their potential in mitigating chemotherapy resistance. Additionally, the therapeutic potential of inhibiting exosome-mediated transporter transfer strategy is particularly examined as a promising strategy to overcome tumor resistance mechanisms.
Collapse
Affiliation(s)
| | | | | | - Yumiao Zhang
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| | - Ren He
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| |
Collapse
|
11
|
Li R, Hu Y, Hou Y, Li J, Niu X, Wang M, Wang L. Receptor-mediated membrane fusion drug delivery system based on chitosan derivatives to enhance tumor chemotherapy. Int J Biol Macromol 2025; 311:143869. [PMID: 40348215 DOI: 10.1016/j.ijbiomac.2025.143869] [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: 01/04/2025] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
Tumor chemotherapy drug delivery systems often face significant challenges, including low targeting specificity and lysosomal sequestration, both of which can severely impair therapeutic efficacy. To overcome these limitations, we have developed a novel receptor-mediated membrane fusion (RMF) drug delivery system based on chitosan derivatives. This system can self-assemble into nanoparticles (NPs) and encapsulate doxorubicin (DOX). The physical properties of both unloaded and DOX-loaded NPs were systematically characterized. In vitro experiments demonstrated that the RMF system selectively interacts with tumor cell surfaces, inhibiting cell proliferation and migration. Additionally, the system effectively targets tumor cells, delivers the drug directly into the cytoplasm, thereby bypassing lysosomal sequestration, thus improving targeting efficiency and enhancing drug delivery. In vivo studies further confirmed the superior anticancer efficacy of the RMF system, alongside its excellent systemic safety. In conclusion, this RMF-based strategy offers a promising platform for the precise delivery of chemotherapeutics, addressing the critical limitations of conventional drug delivery systems and significantly enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Ruxiang Li
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Yaqi Hu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Yiyang Hou
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Jingge Li
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Xiaoyuan Niu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Mandi Wang
- Department of Chemistry, University of Wisconsin-Madison, 500 Lincoln Avenue, 161 Bascom Hall, Madison 53706, WI, United States.
| | - Lianyong Wang
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, China.
| |
Collapse
|
12
|
Haffez H, Sanad HH, Ebrahim H, Hassan ZA. Synergistic effects of abietic acid combined with doxorubicin on apoptosis induction in a human colorectal cancer cell line. Sci Rep 2025; 15:16102. [PMID: 40341222 PMCID: PMC12062260 DOI: 10.1038/s41598-025-99616-2] [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: 01/07/2025] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
Cancer is a significant global disease with high mortality and limited therapeutic options. Chemotherapy is a cancer treatment option; however, there are still issues, including severe side effects, inadequate response, and drug resistance. Abietic acid is a natural diterpene with diverse pharmacological properties and can be used for cancer treatment. Therefore, this study aimed to assess the anticancer efficacy of abietic acid in combination with doxorubicin, a highly clinically used chemotherapeutic agent. Biochemical investigations include initial viability assays, combination therapy using isobologram analysis, apoptosis and cell cycle assays, gene expression assay, ELISA analysis of protein expression, DNA fragmentation, and wound healing assays. The data showed that doxorubicin-abietic acid (DOX-AB) is an effective and safe anticancer combination for Caco-2 cells. DOX-AB had a high safety index with minimal cytotoxicity at the combination dose on normal WI-38 fibroblasts cells. DOX-AB significantly decreased the proliferation and viability of Caco-2 cells, with an increase in the apoptosis rate in the late stage and necrosis with cell cycle arrest at the G2/M phase. Significant changes in the expression of modulators related to apoptosis, inflammation, and epigenetics were observed in gene and protein levels. DOX-AB combination had more efficient anticancer activity than doxorubicin alone. This study suggested that the use of abietic acid in combination with doxorubicin is a promising treatment for colorectal cancer because it enhances doxorubicin activity at relatively low doses with minimal cytotoxicity and overcomes multidrug resistance in tumors; these findings merit further investigation.
Collapse
Affiliation(s)
- Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt.
- Center of Scientific Excellence "Helwan Structural Biology Research, (HSBR)", Helwan University, Cairo, 11795, Egypt.
| | - Hend H Sanad
- Health Affairs Directorate, Mansoura Health Administration, Mansura city, , El Dakahlia, Egypt
| | - Hassan Ebrahim
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, P.O. Box 11795, Cairo, Egypt
| | - Zeineb A Hassan
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
13
|
Nan A, Dumitrascu V, Flangea C, Dumitrescu G, Puscasiu D, Vlad T, Popescu R, Vlad C. From Chemical Composition to Antiproliferative Effects Through In Vitro Studies: Honey, an Ancient and Modern Hot Topic Remedy. Nutrients 2025; 17:1595. [PMID: 40362904 PMCID: PMC12074252 DOI: 10.3390/nu17091595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Honey is a natural product which has been used throughout time as a food, spice, and medicine. Its therapeutic use has its origins in direct empirical observations of various beneficial actions in terms of its anti-infectious, anti-inflammatory, and wound-healing effects, to which an antiproliferative effect is added. In the context of malignant transformation, reductions in chronic inflammation, antioxidant action, cell cycle arrest, and apoptosis activation contribute to this antiproliferative effect, achievements attributed mainly to the polyphenols in its composition. A multitude of in vitro studies performed on malignant cell cultures try to elucidate the real mechanism(s) that can scientifically explain this action. In addition, its use as an adjuvant in association with cytostatic therapy demonstrates a promising effect in enhancing its cytotoxic effect, but also in reducing some adverse effects. Highlighting these actions allows for further perspectives to be opened regarding the use of honey for therapeutic and also prophylactic purposes, as a food supplement. Future studies will support the identification of real antiproliferative effects in patients with malignant tumors in terms of actions on the human body as a whole, moving from cell cultures to complex implications.
Collapse
Affiliation(s)
- Alexandru Nan
- Doctoral School “Engineering of Vegetable and Animal Resources”, University of Life Sciences “King Mihai I” from Timişoara, Calea Aradului 119, 300645 Timisoara, Romania;
| | - Victor Dumitrascu
- Department of Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (V.D.); (C.V.)
| | - Corina Flangea
- Department of Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (V.D.); (C.V.)
| | - Gabi Dumitrescu
- Faculty of Bioengineering of Animal Resources, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania
- ANAPATMOL Research Center, “Victor Babes” University of Medicine and Pharmacy, E. Murgu 2, 300041 Timisoara, Romania; (D.P.); (T.V.); (R.P.)
| | - Daniela Puscasiu
- ANAPATMOL Research Center, “Victor Babes” University of Medicine and Pharmacy, E. Murgu 2, 300041 Timisoara, Romania; (D.P.); (T.V.); (R.P.)
- Department of Cell and Molecular Biology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Tania Vlad
- ANAPATMOL Research Center, “Victor Babes” University of Medicine and Pharmacy, E. Murgu 2, 300041 Timisoara, Romania; (D.P.); (T.V.); (R.P.)
- Department of Cell and Molecular Biology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Roxana Popescu
- ANAPATMOL Research Center, “Victor Babes” University of Medicine and Pharmacy, E. Murgu 2, 300041 Timisoara, Romania; (D.P.); (T.V.); (R.P.)
- Department of Cell and Molecular Biology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Cristian Vlad
- Department of Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (V.D.); (C.V.)
| |
Collapse
|
14
|
Huang J, Yang J, Yang Y, Lu X, Xu J, Lu S, Pan H, Zhou W, Li W, Chen S. Mitigating Doxorubicin-Induced Cardiotoxicity and Enhancing Anti-Tumor Efficacy with a Metformin-Integrated Self-Assembled Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415227. [PMID: 40052211 PMCID: PMC12061326 DOI: 10.1002/advs.202415227] [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: 11/19/2024] [Revised: 01/06/2025] [Indexed: 05/10/2025]
Abstract
Doxorubicin (Dox) is a potent chemotherapeutic agent commonly used in cancer treatment. However, cardiotoxicity severely limited its clinical application. To address this challenge, a novel self-assembled nanomedicine platform, PMDDH, is developed for the co-delivery of Dox and metformin, an antidiabetic drug with cardioprotective and anti-tumor properties. PMDDH integrates metformin into a polyethyleneimine-based bioactive excipient (PMet), with Dox intercalated into double-stranded DNA and a hyaluronic acid (HA) coating to enhance tumor targeting. The PMDDH significantly improves the pharmacokinetics and tumor-targeting capabilities of Dox, while metformin enhances the drug's anti-tumor activity by downregulating programmed cell death ligand 1 (PD-L1) and activating the AMP-activated protein kinase (AMPK) signaling pathway. Additionally, the DNA component stimulates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, which synergizes with Dox-induced immunogenic cell death (ICD) to promote a robust anti-tumor immune response. PMDDH markedly reduces Dox-induced cardiotoxicity by preserving mitochondrial function, reducing reactive oxygen species (ROS) production, and inducing protective autophagy in cardiomyocytes. These findings position PMDDH as a promising dual-function nanomedicine that enhances the anti-tumor efficacy of Dox while minimizing its systemic toxicity, offering a safer and more effective alternative for cancer therapy.
Collapse
Affiliation(s)
- Jiaxin Huang
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Jieru Yang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Yuanying Yang
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Xiaofeng Lu
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
- Department of CardiologyShanghai General Hospital Jiuquan HospitalNo. 22, West StJiuquanGansu735000China
| | - Juan Xu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
| | - Shan Lu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Hong Pan
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
- Hunan Key Laboratory of The Research and Development of Novel Pharmaceutical PreparationsSchool of Pharmaceutical ScienceChangsha Medical UniversityChangshaHunan410219China
| | - Wenqun Li
- Department of PharmacySecond Xiangya HospitalCentral South UniversityChangshaHunan410011China
| | - Songwen Chen
- Department of CardiologyShanghai General HospitalShanghai Jiao Tong University School of MedicineNo.100, Haining RdShanghai200080China
| |
Collapse
|
15
|
Rakhshani A, Maghsoudian S, Ejarestaghi NM, Yousefi M, Yoosefi S, Asadzadeh N, Fatahi Y, Darbasizadeh B, Nouri Z, Bahadorikhalili S, Shaabani A, Farhadnejad H, Motasadizadeh H. Polyethylene oxide-chitosan-doxorubicin/polycaprolactone-chitosan-curcumin pH-sensitive core/shell nanofibrous mats for the treatment of breast cancer: Fabrication, characterization and in vitro and in vivo evaluation. Int J Biol Macromol 2025; 305:141191. [PMID: 39971028 DOI: 10.1016/j.ijbiomac.2025.141191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/06/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
The main objective of this study was to fabricate a pH-sensitive drug carrier based on coaxial electrospun nanofibrous mats for concurrent local delivery of hydrophilic and hydrophobic anti-cancer drugs to improve the anti-tumor efficacy on breast cancer. Therefore, co-axial electrospinning technique was applied to prepare polyethylene oxide-chitosan/polycaprolactone-chitosan (PEO-CS/PCL-CS) pH-sensitive core-shell nanofibers. Doxorubicin hydrochloride (DOX, hydrophilic anti-cancer) and curcumin (CUR, hydrophobic anticancer) were loaded into core and shell sections of the fabricated pH-sensitive coaxial nanofibers, respectively. Their structure and morphology were analyzed via SEM, TEM, TGA, and FTIR techniques. The results of in vitro release analysis indicated that the release of DOX and CUR from the fabricated nanofibers was strongly depended on pH. The combined effects of the two drugs on MCF-7 cell inhibition, as measured by the MTT assay, revealed that the 1:5 ratio of DOX to CUR resulted in a CI of 0.00492, showing the strongest synergistic effect. The results of in-vivo studies indicated that the PEO-CS-DOX/PCL-CS-CUR pH-sensitive core-shell nanofibers possessed remarkable anti-tumor efficacy. As a result, PEO-CS-DOX/PCL-CS-CUR pH-sensitive core-shell nanofibrous mats with pH-responsive and sustainable and controllable manner could improve the local anti-tumor efficacy on breast cancer via inhibiting the side effects of free DOX and CUR drugs.
Collapse
Affiliation(s)
- Amir Rakhshani
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran university of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Samane Maghsoudian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran university of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Negin Mousavi Ejarestaghi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahzad Yousefi
- Department of Healthcare Emergency Management, Faculty of Medicine, Boston University, Boston, MA, USA; Graduate, Veterinary Medicine School, Āzad University, Tehran, Iran
| | - Sepideh Yoosefi
- Department of Drug and Food Control, Faculty of pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Asadzadeh
- Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran university of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Behzad Darbasizadeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Nouri
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Shaabani
- Department of Polymer and Materials Chemistry, Faculty of Chemistry and Petroleum Sciences, Shahid Beheshti University, Iran
| | - Hassan Farhadnejad
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
16
|
Zhang K, Yuan B, Dai X, Chen W, Zhang C, Qiao Y, Cao W, Chen Y, Duan X, Zhang X, Yang W, Li X, Zhao J, Liu K, Dong Z, Lu J. Selection and identification of DNA aptamer binding VDAC1 for tumor tissue imaging and targeted drug delivery. Int J Biol Macromol 2025; 306:141249. [PMID: 39984095 DOI: 10.1016/j.ijbiomac.2025.141249] [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: 12/25/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Hepatocellular carcinoma (HCC) represents a significant health concern. Identifying novel molecular targets is crucial for clinical diagnosis and targeted treatment of HCC. Aptamers are capable of binding specifically to cancer cells via target protein molecules. Consequently, aptamers are frequently employed to identify novel cancer biomarkers. The invasiveness of tumor cells is closely associated with the recurrence and metastasis of tumors. In this study, the highly invasive Huh7-P3 cells were initially constructed, and subsequently, several aptamers that could specifically recognize Huh7-P3 were developed using cell-based Systematic Evolution of Ligands by Exponential Enrichment (SELEX). The selected aptamer, designated S2-2, demonstrated the capacity to bind to multiple cancer cells. Furthermore, tissue imaging demonstrated that S2-2 exhibited a specific recognition of HCC tissue, while demonstrating no binding to normal tissue. Subsequently, voltage-dependent anion channel 1 (VDAC1) was identified as a potential target for S2-2. Furthermore, Doxorubicin (Dox)-loaded S2-2 was shown to specifically kill target Huh7-P3 cells. In vivo fluorescence imaging revealed that S2-2 was capable of specifically targeting tumors. Importantly, S2-2-Dox enhanced the anti-tumor efficacy of Dox in cell-line-derived xenograft (CDX) model. This study may provide a promising biomarker and molecular target for the clinical diagnosis and targeted therapy of cancers with high VDAC1 expression.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Baoyin Yuan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Xiaoshuo Dai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Wei Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Chengjuan Zhang
- Department of Pathology, Henan Cancer Hospital, Zhengzhou University, Zhengzhou, Henan Province 450003, PR China
| | - Yan Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Wenbo Cao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Yihuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Xiaoxuan Duan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Xiaoyan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Wanjing Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China; State Key Laboratory of Metabolic Disorders and Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China.
| |
Collapse
|
17
|
Fuchs-Tarlovsky V, Alvarez-Altamirano K, Vedrenne-Gutiérrez F. Evaluation and Prognostic Impact of Nutrition in Patients with Acute Leukemia: A Narrative Review. Curr Oncol Rep 2025; 27:625-633. [PMID: 40195210 DOI: 10.1007/s11912-025-01671-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
PURPOSE OF THIS REVIEW Acute leukemia (AL) is a hematological neoplasm with rapid progression that affects nutritional status of patients. Assessing nutrition in patients with haematological malignancies poses challenges due to the rapid progression of the disease, the variety of methods for evaluating malnutrition. In this review we will provide evidence of the need of early malnutrition diagnosis thru timely assessment and the implications of malnutrition in patients evolution in patients with AL. RECENT FINDINGS The prevalence of malnutrition ranging from 15 to 26.5% among patients with AL. It is well known that inflammation and cytokine production have been recognized as potential mechanisms of CCS in hematologic malignancies. Regardless of the mechanism underlying Cancer Cachexia Syndrome (CCS) in AML, patients undergoing conditioning chemotherapy and during transplant or antineoplastic treatment. In addition to this, patients with AL undergoing chemotherapy frequently face two main challenges: oral mucositis (OM) and neutropenic colitis (NC). Both conditions challenge nutrition intake and nutrients absorption which make them more susceptible to have nutritional deficits. Malnutrition in patients with acute leukemia face a higher risk of therapy failure, increased rates of relapse, and higher mortality rates. Nutritional status impact patient's prognosis in many ways, malnutrition increases risk of antineoplastic treatment toxicities, hospital stay, and cost, as well reduces quality of life and this condition worsens patient's prognosis.
Collapse
Affiliation(s)
- Vanessa Fuchs-Tarlovsky
- Servicio de Nutrición Clínica, Hospital General de México, Dr Balmis 148, Colonia Doctores, Delegación Cuauhthemoc, Mexico City, 06720, Mexico.
| | - Karolina Alvarez-Altamirano
- Servicio de Nutrición Clínica, Hospital General de México, Dr Balmis 148, Colonia Doctores, Delegación Cuauhthemoc, Mexico City, 06720, Mexico
| | - Fernand Vedrenne-Gutiérrez
- School of Medicine and Health Sciences, Universidad Anáhuac México, Huixquilucan, Estado de México, México
| |
Collapse
|
18
|
Zhang X, Wang B, Qi X, Qian Z, Gao X, Cheng Y, Wang X. A Glutathione-Responsive System with Prodrug and Sensitization Strategies for Targeted Therapy of Glioma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2501620. [PMID: 40119786 DOI: 10.1002/smll.202501620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/08/2025] [Indexed: 03/24/2025]
Abstract
Glioblastoma represents a highly aggressive form of malignant tumor within the central nervous system. Although chemotherapy remains the primary therapeutic strategy, its efficacy is often limited. To overcome the limitations associated with chemotherapeutic agents, such as high toxicity and non-specific adverse effects, a novel nanoparticle system comprising cRGD-modified and glutathione (GSH)-responsive polymers, and PEG-ss-Dox and apatinib (AP) (PDOX-AP/cRGD-NPs) is developed. PDOX-AP/cRGD-NPs show effective penetration of the blood-brain barrier (BBB), facilitate targeted delivery to brain tumors, and exhibit controlled drug release. PDOX-AP/cRGD-NPs show more effect in reducing the viability of GL-261, U87-MG, and LN-229 cells, inhibiting clonogenicity, and suppressing anti-apoptotic protein expression than PDOX/cRGD-NPs or AP/cRGD-NPs. Additionally, PDOX-AP/cRGD-NPs substantially increase drug uptake, BBB penetration, apoptosis rates, and the proportion of cells in the G2 phase. In vivo experiments further reveal that cRGD-directed nanoparticles exhibit superior accumulation in glioma regions compared to their non-cRGD-modified counterparts. In the interim, PDOX-AP/cRGD-NPs demonstrate significant efficacy in suppressing both ectopic and orthotopic growth of GL-261 gliomas, as well as orthotopic LN-229 gliomas, thereby markedly extending the median survival duration. This study introduces a promising targeted co-delivery system for combination chemotherapy.
Collapse
Affiliation(s)
- Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Xin Qi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| |
Collapse
|
19
|
Skaličková M, Abramenko N, Charnavets T, Vellieux F, Leischner Fialová J, Kučnirová K, Kejík Z, Masařík M, Martásek P, Pacak K, Pacák T, Jakubek M. Interaction of Selected Anthracycline and Tetracycline Chemotherapeutics with Poly(I:C) Molecules. ACS OMEGA 2025; 10:15935-15946. [PMID: 40321536 PMCID: PMC12044458 DOI: 10.1021/acsomega.4c05483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 02/11/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025]
Abstract
Despite the natural ability of the immune system to recognize cancer and, in some patients, even to eliminate it, cancer cells have acquired numerous evading mechanisms. With the increasing knowledge and focus shifting from targeting rapidly proliferating cells with chemotherapy to modulating the immune system, there have been recent efforts to integrate (e.g., simultaneously or sequentially) various therapeutic approaches. Combining the oncolytic activity of some chemotherapeutics with immunostimulatory molecules, so-called chemoimmunotherapy, is an attractive strategy. An example of such an immunostimulatory molecule is polyinosinic:polycytidylic acid [Poly(I:C)], a synthetic analogue of double-stranded RNA characterized by rapid nuclease degradation hampering its biological activity. This study investigated the possible interactions of tetracycline and anthracycline chemotherapeutics with different commercial Poly(I:C) molecules and protection against nuclease degradation. Fluorescence spectroscopy and circular dichroism revealed an interaction of all of the selected chemotherapeutics with Poly(I:C)s and the ability of doxycycline and minocycline to prolong the resistance to RNase cleavage, respectively. The partial protection was observed in vitro as well.
Collapse
Affiliation(s)
- Markéta Skaličková
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Nikita Abramenko
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Tatsiana Charnavets
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, 252
50 Vestec, Czech
Republic
| | - Frédéric Vellieux
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | | | - Kateřina Kučnirová
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Zdeněk Kejík
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Michal Masařík
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
- Department
of Physiology, Faculty of Medicine, Masaryk
University, Kamenice 5, 625 00 Brno, Czech Republic
- Department
of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625
00, Czech Republic
| | - Pavel Martásek
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Karel Pacak
- Section on
Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute
of Child Health and Human Development, National
Institutes of Health, Building 10, Room 1-3140, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Tomáš Pacák
- TumorSHOT, Italská 2581/67, Vinohrady,
Praha 2, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| |
Collapse
|
20
|
Bazavar MR, Helali H, Boukani LM, Yousefi B, Valizadeh A. Enhancing Doxorubicin Sensitivity in Osteosarcoma Cancer Cells: Unveiling the Role of Resveratrol-Induced Oxidative DNA Damage. Drug Res (Stuttg) 2025. [PMID: 40273920 DOI: 10.1055/a-2567-9916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Our current research aims to investigate how Resveratrol influences DOX-induced apoptosis in Saos-2 cells resulting from DNA damage.Saos-2 cells were cultured with DOX, and an MTT assay was conducted to evaluate cell viability. The expression levels of DNA damage markers were evaluated using qRT-PCR and western blotting methods. Apoptosis was also investigated by flow cytometry.In a dose-dependent way, DOX produced a profuse suppression of cell proliferation. This study investigates the effect of Resveratrol on DOX-induced apoptosis due to DNA damage in Saos-2 cells (P<0.05). There was an increase in H2AX, ATR, ATM, Rad51, and p53 expression, possibly contributing to subsequent apoptosis. Furthermore, Resv enhanced the apoptosis caused by DOX in Saos-2 cells.The findings from the current research provide an understanding of how Resv plays a role in potentially treating osteosarcoma by enhancing DOX-induced apoptosis.
Collapse
Affiliation(s)
- Mohammad Reza Bazavar
- Department of Orthopedic Surgery, School of Medicine and Shohada Educational Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Helali
- Department of Orthopedic Surgery, School of Medicine and Shohada Educational Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Bahman Yousefi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Valizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Zhao S, Bai B, Zhu B, Cui Y, Deng W, Xie Z, Wang S, Wang X, Mao Y, Lu Y, Huang Y, You T, Sun W, Shen X, Lu X. Hyaluronic acid-modified doxorubicin-covalent organic framework nanoparticles triggered pyroptosis in combinations with immune checkpoint blockade for the treatment of breast cancer. Int J Biol Macromol 2025; 310:143265. [PMID: 40274165 DOI: 10.1016/j.ijbiomac.2025.143265] [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: 02/19/2025] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive malignancy, and the current treatment strategies have poor efficacy. Pyroptosis, a type of immunogenic cell death, significantly enhances antitumor immunity by triggering the release of numerous intracellular components. Here, we prepared a covalent-organic framework (COF) loaded with doxorubicin (DOX) and coated it with hyaluronic acid (HA) as a therapeutic delivery system (HA@DOX-COF) for the treatment of TNBC. Mechanistically, upon cellular uptake, HA@DOX-COF activated pyroptosis through the caspase-3/GSDME pathway. Moreover, HA@DOX-COF induced a pyroptosis-induced antitumor immune response and further augmented the efficacy of immune checkpoint blockade (ICB). Our results demonstrated that the combination of HA@DOX-COF and an anti-PD-1 antibody markedly inhibited tumor progression in a TNBC murine model. Overall, our work offers a potential approach for pyroptosis-induced immunotherapy for TNBC, which may increase the efficacy of ICB-based immunotherapy.
Collapse
Affiliation(s)
- Shengsheng Zhao
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Binglong Bai
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Bingzi Zhu
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yuekai Cui
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wenhai Deng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zuoliang Xie
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shuaibin Wang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiang Wang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yiwen Mao
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongyong Lu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yingpeng Huang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Tao You
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xian Shen
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xufeng Lu
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
22
|
Singh S, Singh V, Singh R, Gouri V, Koch B, Samant M. Synergistic combination of doxorubicin with fisetin for the treatment of lymphoma. Eur J Pharmacol 2025; 992:177361. [PMID: 39929420 DOI: 10.1016/j.ejphar.2025.177361] [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: 09/18/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/25/2025]
Abstract
Lymphoma is a common cancer of the lymphatic system, and its treatment presents considerable clinical difficulties due to the constraints of existing medicines. Anticancer drug such as Doxorubicin (DOX) is an effective chemotherapeutic drug that is frequently used to treat lymphoma and other cancers; however, it is linked with considerable toxicities. Fisetin, a naturally occurring flavonoid, exhibits anticancer properties and has the potential to augment the therapeutic effects of DOX. This study explores the synergistic effects of combining DOX with fisetin in the treatment of lymphoma. The combination of DOX and fisetin significantly inhibits cell viability, induced membrane blabbing, chromatin condensation, and promoted apoptosis compared to monotherapies. The study also showed that the synergistic effect of fisetin along with DOX significantly promotes apoptosis in DL cells through intracellular ROS generation, mitochondrial aggregation at the periphery of the nucleus and, increased p53, Bax, cytochrome c, caspase 3, caspase 9, and cleaved caspase 9 expression. Additionally, combination therapy not only increased the mean survival of the treated group animals but also reduced the tumor burden. While histopathological parameters have shown overall improvement in combination therapy. This study proposes a novel combinational therapy for the treatment of lymphoma and requires further clinical investigation.
Collapse
Affiliation(s)
- Sumeet Singh
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Virendra Singh
- Genotoxicology and Cancer Biology Laboratory, Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Ranjeet Singh
- Department of Zoology, Soban Singh Jeena University (Bageshwar Campus), Almora, Uttarakhand, India; Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Vinita Gouri
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India; Department of Zoology, Kumaun University, Nainital, Uttarakhand, India
| | - Biplob Koch
- Genotoxicology and Cancer Biology Laboratory, Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Mukesh Samant
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India.
| |
Collapse
|
23
|
Tani T, Oikawa M, Ohara H, Yaegashi D, Sato Y, Yokokawa T, Miura S, Misaka T, Yoshihisa A, Ishida T, Takeishi Y. Subclinical B-type Natriuretic Peptide Elevation 24 Months After Anthracycline-Containing Chemotherapy. Int Heart J 2025; 66:279-284. [PMID: 40090710 DOI: 10.1536/ihj.24-293] [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] [Indexed: 03/18/2025]
Abstract
The incidence of anthracycline-induced cardiotoxicity typically occurs within the first year after chemotherapy, but the changes in cardiac function and biomarkers beyond this initial year have not been adequately investigated. We analyzed 105 consecutive patients followed for 24 months after anthracycline-containing chemotherapy at Fukushima Medical University Hospital from June 2018 to April 2021. Echocardiography and blood tests for cardiac troponin I and B-type natriuretic peptide (BNP) were conducted at baseline, and 3, 6, 12, and 24 months after chemotherapy initiation. In the whole patient cohort, BNP levels increased from 10.5 [6.3-18.3] pg/mL at baseline to 19.2 [12.1-34.5] pg/mL at 24 months after chemotherapy (P < 0.01). Based on BNP levels at 24 months, the patients were divided into 2 groups: a BNP-elevated group (n = 57) and a BNP-normal group (n = 48). In the BNP-elevated group, time-course changes revealed that BNP levels remained stable until 12 months, but increased at 24 months. Multivariate logistic analysis identified age, total anthracycline dose, and baseline BNP levels as predicting factors for elevated BNP levels at 24 months. Subclinical BNP elevation was observed at 24 months of follow-up after initiation of anthracycline-containing chemotherapy.
Collapse
Affiliation(s)
- Tetsuya Tani
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Masayoshi Oikawa
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Himika Ohara
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Daiki Yaegashi
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Yu Sato
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Tetsuro Yokokawa
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Shunsuke Miura
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Tomofumi Misaka
- Department of Cardiovascular Medicine, Fukushima Medical University
- Department of Community Cardiovascular Medicine, Fukushima Medical University
| | - Akiomi Yoshihisa
- Department of Cardiovascular Medicine, Fukushima Medical University
- Department of Clinical Laboratory Sciences, Fukushima Medical University
| | - Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University
| | | |
Collapse
|
24
|
Xiang L, Xiang X, Jiao Q, Luo Y, Zeng G, Zhang W, Qin Y, Chen Y. Inhibition of HSC proliferation and hepatic fibrogenesis with Erythrocyte membrane coated Doxorubicin/Black phosphorus nanosheets. Int J Pharm 2025; 673:125403. [PMID: 40015579 DOI: 10.1016/j.ijpharm.2025.125403] [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: 11/12/2024] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Rapid proliferation underlies the abnormal expansion of activated hepatic stellate cells (aHSCs) and thereby contributes to the development and progression of liver fibrosis, so inhibition of HSC proliferation serves as a good antifibrotic strategy. As a potent topoisomerase II inhibitor, doxorubicin (DOX), an antineoplastic drug, exhibits a significant antifibrotic activity in vitro via retarding the growth of aHSCs and reversing their myofibroblastic phenotype, but its severe hepatotoxicity, cardiotoxicity, and renal toxicity limit its wide clinical application. Therefore, enhancing the specificity and efficacy of DOX in targeting aHSCs to improve its therapeutic index and minimize its adverse effects has become a key point for the success of DOX in antifibrotic treatment. In this study aimed at liver fibrosis treatment, we combined the excellent drug-loading capability and good biocompatibility of black phosphorus nanosheets (BPNSs), the protective and camouflaging properties of red blood cell membrane encapsulation, and the HSCs-targetability provided by the surface modification with vitamin A derivatives, into the construction of HSCs-targeted BP/DOX nanovesicles (BP/DOX@RMV-VA). The obtained DOX nanovesicles exhibited a uniform particle size and spheroid morphology, excellent diffusion property and stability, and high DOX loading. Specifically, they demonstrated outstanding biosafety, effective HSCs-targetability both in vivo and in vitro, and markedly improved pharmacokinetic profile of DOX. BP/DOX@RMV-VA produced strong antiproliferative and MF-phenotype reverting activity both in cultured aHSCs and in mice chronically injured by CCl4. And accordingly, the administration of BP/DOX@RMV-VA to CCl4-injured mice effectively suppressed the expansion of aHSCs and fibrogenesis, and significantly improved liver structure and function without causing detectable cardiotoxicity. These results highly suggest the therapeutic potential of BP/DOX@RMV-VA in treating liver fibrosis and other fibrosis-associated liver diseases.
Collapse
Affiliation(s)
- Li Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Xianjing Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Yu Luo
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Guodong Zeng
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Wenhui Zhang
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Yuting Qin
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China
| | - Yuping Chen
- School of Pharmaceutical Sciences, University of South China, Hengyang, Hunan 410001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China; MOE Key Laboratory of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China.
| |
Collapse
|
25
|
Chmur K, Brzeski J, Reghukmar S, Tesmar A, Sikorski A, Inkielewicz-Stępniak I, Wyrzykowski D. Structural, Physicochemical, and Biological Insights into Novel (Acetylacetonate)(Oxydiacetato)Oxidovanadium(IV) Complexes with N-Containing Aromatic Compounds. Chemistry 2025; 31:e202404496. [PMID: 39911089 DOI: 10.1002/chem.202404496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/07/2025]
Abstract
The crystal structures of three novel oxidovanadium(IV) salts containing the [VO(acac)(oda)]- anion (acac=acetylacetonate, oda=oxydiacetate) and quinolinium ([QH]+), isoquinolinium ([(isoQ)H+) and acridinium ([(acr)H+)] counterions, of molecular formulas [QH][VO(acac)(oda)] (1), [(isoQ)H][VO(acac)(oda)](H2O) (2) and [(acr)H][VO(acac)(oda)](H2O)2 (3) are reported. Notably, these complexes represent the first structurally characterized salts comprising the heteroligand [VO(acac)(oda)]- complex anion. A comprehensive physicochemical characterization of the complexes in both solid state and solution is provided, with general discussions on the role of nitrogen-containing heterocyclic compounds on the structure of the resulting ternary oxidovanadium(IV) complexes. The experimental results were complemented by density functional theory (DFT) calculations to evaluate possible ligand conformations in the coordination sphere of V(IV). In addition, the nature of the bonds involved in the chelation of the vanadium(IV) cation is considered and a detailed assessment of the electron spin density is presented. Finally, the cytotoxic activity of the compounds was tested against breast (MDA-MB-231 and MCF-7) cancer cell lines to assess their potential use as chemotherapeutic agents in cancer therapy.
Collapse
Affiliation(s)
- Katarzyna Chmur
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Jakub Brzeski
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
- QSAR Lab Ltd, Trzy Lipy 3, 80-172, Gdańsk, Poland
| | - Swathy Reghukmar
- Department of Pharmaceutical Pathophysiology, Medical University of Gdansk, Dębinki 7, Building 27, 80-211, Gdańsk, Poland
| | - Aleksandra Tesmar
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Artur Sikorski
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Iwona Inkielewicz-Stępniak
- Department of Pharmaceutical Pathophysiology, Medical University of Gdansk, Dębinki 7, Building 27, 80-211, Gdańsk, Poland
| | - Dariusz Wyrzykowski
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| |
Collapse
|
26
|
Zhang D, Xu T, Gao X, Qu Y, Su X. Methyltransferase-like 3-mediated RNA N 6-methyladenosine contributes to immune dysregulation: diagnostic biomarker and therapeutic target. Front Immunol 2025; 16:1523503. [PMID: 40196133 PMCID: PMC11973086 DOI: 10.3389/fimmu.2025.1523503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/11/2025] [Indexed: 04/09/2025] Open
Abstract
Methyltransferase-like 3 (METTL3) plays a crucial role in post-transcriptional gene regulation. Substantial evidence links METTL3 to various immune dysfunctions, such as the suppression of antiviral immunity during viral infections and the disruption of immune tolerance in conditions like autoimmune diseases, myeloid leukemia, skin cancers, and anticancer immunotherapy. However, a thorough review and analysis of this evidence is currently missing, which limits the understanding of METTL3's mechanisms and significance in immune dysfunctions. This review aims to elucidate the roles and mechanisms of METTL3 in these immune issues, highlighting its connections and proposing new insights into its modulation of immune responses. Analysis results in this review suggest that METTL3 hampers antiviral immunity, worsens viral replication and infection, and disrupts immune tolerance; conversely, regulating METTL3 enhances antiviral immunity and facilitates viral clearance. Moreover, clinical data corroborates these findings, showing that METTL3 overexpression is associated with increased susceptibility to viral infections and autoimmune conditions. This review establishes a theoretical basis for considering METTL3 as a novel regulator, an important diagnostic biomarker, and a potential target for treating immune dysfunctions.
Collapse
Affiliation(s)
- Deshuang Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ting Xu
- Department of Pediatrics, School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaoxue Gao
- Department of Pediatrics, School of Clinical Medicine & The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yi Qu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaojuan Su
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Wang X, Liu G, Pu X, Ren T, Zhang F, Shen M, Zhu Y, Kros A, Yang J. Combating cisplatin-resistant lung cancer using a coiled-coil lipopeptides modified membrane fused drug delivery system. J Control Release 2025; 379:45-58. [PMID: 39756686 DOI: 10.1016/j.jconrel.2025.01.004] [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: 09/06/2024] [Revised: 12/05/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Drug resistance to chemotherapy in treating cancers becomes an increasingly serious challenge, which leads to treatment failure and poor patient survival. Drug-resistant cancer cells normally reduce intracellular accumulation of drugs by controlling drug uptake and promoting drug efflux, which severely limits the efficacy of chemotherapy. To overcome this problem, a membrane fused drug delivery system (MF-DDS) was constructed to treat cisplatin (DDP)-resistant lung cancer (A549-DDP) by delivering DDP via membrane fusion using a complementary coiled-coil forming peptides (CP8K4/CP8E4). The lipopeptide CP8K4 was pre-incubated firstly and decorated on the surface of A549-DDP cells, and then the cells interacted with the lipopeptide CP8E4 modified on the lipid bilayer (LB) coated PLGA nanoparticles loading DDP (PLGA-DDP@LB-CP8E4), leaded to the direct cytosolic DDP delivery and cancer cell death. Compared with free DDP, this MF-DDS achieved a 13.42-folds reduced IC50 value of A549-DDP cells in vitro, and tumor size was down-regulated, showing only 1/5.26 of the original weight in vivo. Meanwhile, the anti-drug resistant mechanism was explored, where the MF-DDS inhibited the expression of efflux protein genes, including MRP1, MRP2, and ABCG2, leading to increased intracellular drug accumulations. Altogether, this MF-DDS effectively delivered DDP into DDP-resistant cancer cells, making it a promising and improved pharmacological therapeutic approach for drug-resistant tumor treatment.
Collapse
Affiliation(s)
- Xi Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Guiquan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Xueyu Pu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Tangjun Ren
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Fan Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - MengJie Shen
- Leiden Institute of Chemistry-Supramolecular and Biomaterial Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Alexander Kros
- Leiden Institute of Chemistry-Supramolecular and Biomaterial Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, the Netherlands.
| | - Jian Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
28
|
Kashyap A, Choudhury K, Mazumdar P, Choudhury D. Interaction of tautomers of doxorubicin with guanine-cytosine base pair: a density functional theory study. J Mol Model 2025; 31:105. [PMID: 40029510 DOI: 10.1007/s00894-025-06331-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: 01/06/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
CONTEXT Anthracycline anticancer antibiotics from Streptomyces peucetius show high affinity for nucleobases. This study uses quantum mechanical density functional theory (DFT) to investigate interactions between doxorubicin (DOX) tautomers and the guanine-cytosine (GC) base pair. Intermolecular distances and interaction energies reveal structural relationships and stabilization. Interaction energy studies show that DOX-GC has greater binding affinity and greater stability in the aqueous phase as compared to that in gaseous phase. Interestingly, the tautomer which show greater affinity for GC in the gas phase is different from the one in the aqueous phase. Reduced density gradient (RDG) scatter plots and quantum theory of atoms in molecules (QTAIM) confirm the presence of hydrogen bonds and its strength. Natural bond orbital (NBO) analysis elucidates donor-acceptor orbital interactions. These findings provide an understanding of the intermolecular interactions between DOX tautomers and the GC base pair, which is likely to provide insight into the molecular basis for DOX's anticancer activity and therapeutic efficacy. METHODS DFT calculations were performed using the B3LYP functional with a 6-31G(d,p) basis set in the Gaussian 09 package, including solvent effects through the integral equation formalism polarizable continuum model (IEF-PCM). Topological analysis and quantum theory of atoms in molecules (QTAIM) studies were conducted using the Multiwfn program, while non-covalent interactions were analysed using visual molecular dynamics (VMD) software.
Collapse
Affiliation(s)
- Angarag Kashyap
- Department of Chemistry, B Borooah College, Ulubari, Guwahati, 781007, Assam, India
- Department of Chemistry, Gauhati University, Guwahati, 781014, India
| | - Kripangkar Choudhury
- Department of Chemistry, B Borooah College, Ulubari, Guwahati, 781007, Assam, India
- Department of Chemistry, Gauhati University, Guwahati, 781014, India
| | - Pradyumna Mazumdar
- Department of Chemistry, B Borooah College, Ulubari, Guwahati, 781007, Assam, India
| | - Diganta Choudhury
- Department of Chemistry, B Borooah College, Ulubari, Guwahati, 781007, Assam, India.
| |
Collapse
|
29
|
Son Phan K, Nghi Do H, Thuy Doan B, Thu Huong Le T, Thu Trang Mai T, Bao Ngoc Nguyen Q, Nham Dong T, Hung Bui Ha B, Dung Dang V, Dang LH, Quyen Tran N, Thu Ha P. The Influence of Cyanine 5.5 and Doxorubicin on Cell Cycle Arrest, Magnetic Resonance, and Near-Infrared Fluorescence Optical Imaging for Fe 3O 4-Encapsulated PLA-TPGS Nanoparticles. ChemMedChem 2025; 20:e202400586. [PMID: 39568159 DOI: 10.1002/cmdc.202400586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/22/2024]
Abstract
The combination of magnetic resonance imaging (MRI)/near-infrared (NIR) fluorescence signals and chemotherapy agents has been developed for cancer diagnosis and treatment. In this work, we investigated the impacts of Cyanine 5.5 and Doxorubicin on cell cycle arrest, magnetic resonance, and NIR fluorescence optical imaging for Fe3O4-encapsulated nanosystems based on poly(lactide)-tocopheryl polyethylene glycol succinate (PLA-TPGS) copolymer. Although Cyanine 5.5 and Fe3O4 nanoparticles (NPs) are less cytotoxic than Doxorubicin, they present a cytostatic effect, inducing cell cycle arrest at the G2/M phase in human brain adenocarcinoma (CCF-STTG1) cells. For MRI applications, the permeability of the PLA-TPGS copolymer coating layer to water molecules might lengthen the translational diffusion time (τ D ${{{{\bf\tau}}}_{{\bf D}}}$ ), causing the higher relaxivity ratio (r2/r1) compared to bare Fe3O4 NPs under an applied magnetic field (7 Tesla). Notably, the chemical structures of Cyanine 5.5 and Doxorubicin significantly contribute to the enhancement of the T2 relaxivities of Fe3O4 NPs through π-π and ρ-π conjugation. Furthermore, the radiance ratio and signal-to-noise ratio enhancement and a slight blue shift in the optimal excitation and emission wavelengths were recorded. These findings show the potential for in vivo MRI and NIR bioimaging experiments of the nanoparticles.
Collapse
Affiliation(s)
- Ke Son Phan
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Huu Nghi Do
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Bich Thuy Doan
- The Institute I-CLeHS Institute of Chemistry for Life and Health Sciences, ENSCP Chimie ParisTech, PSL Université, CNRS UMR, 8060, Paris, France
| | - Thi Thu Huong Le
- Faculty of Natural Resources and Environment, Vietnam National University of Agriculture, Trau Quy, Gia Lam District, Hanoi, Vietnam
| | - Thi Thu Trang Mai
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Quynh Bao Ngoc Nguyen
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Thi Nham Dong
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Bao Hung Bui Ha
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Viet Dung Dang
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Le Hang Dang
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, HCMC, VietNam
| | - Ngoc Quyen Tran
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, HCMC, VietNam
| | - Phuong Thu Ha
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| |
Collapse
|
30
|
Abu Saleem E, Lafi Z, Shalan N, Alshaer W, Hamadneh I. Formation and evaluation of doxorubicin and cromoglycate metal-organic framework for anti-cancer activity. Nanomedicine (Lond) 2025; 20:467-479. [PMID: 39888613 PMCID: PMC11875491 DOI: 10.1080/17435889.2025.2459059] [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: 09/28/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
AIMS We develop and evaluate copper-based metal-organic frameworks (Cu-MOFs) incorporating cromolyn as a linker to enhance structural stability, drug delivery efficiency, and therapeutic potential, particularly for breast cancer treatment. MATERIALS & METHODS Two Cu-MOF formulations were synthesized: Cu-MOFs-BDC-DOX (using terephthalic acid) and Cu-MOFs-CROMO-DOX (using cromolyn as a linker). Characterization was performed using SEM/TEM for morphology, and FTIR, XRD, and TGA to confirm structural integrity. Drug encapsulation efficiency and release profiles were assessed, followed by in vitro cytotoxicity, cell migration, and colony formation assays using MDA-MB-231 breast cancer cells. RESULTS Both formulations demonstrated a high encapsulation efficiency (83-91%) and sustained drug release over 48 h at pH 7.4. Cu-MOFs-CROMO-DOX exhibited superior cytotoxicity with an IC50 of 0.88 ± 0.07 µM compared to 7.1 ± 0.11 µM for Cu-MOFs-BDC-DOX. Both formulations inhibit cancer cell migration and colony formation in a dose-dependent manner. CONCLUSIONS The Cu-MOFs-CROMO-DOX formulation demonstrated enhanced therapeutic potential, outperforming its counterpart in targeting breast cancer cells. This study highlights the promise of MOF-based nanocarriers in overcoming the limitations of conventional chemotherapy, offering a pathway to more effective and targeted cancer treatments with reduced side effects.
Collapse
Affiliation(s)
- Ebaa Abu Saleem
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Naeem Shalan
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Walhan Alshaer
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Imad Hamadneh
- Department of Chemistry, The University of Jordan, Amman, Jordan
| |
Collapse
|
31
|
Goyal K, Babu MA, Afzal M, Rekha A, Ali H, Gupta S, Singh RP, Mishra A, Singh H, Agrawal M, Rana M, Imran M, Khan A. Exploring the therapeutic promise of fisetin: molecular mechanisms and clinical aspects in lung cancer. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2025:jcim-2024-0444. [PMID: 40013371 DOI: 10.1515/jcim-2024-0444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025]
Abstract
Fisetin, a flavonol belonging to the flavonoid subclass, is a ubiquitous dietary flavonoid present in fruits and vegetables, including fruit peels, and has proven potential for anticancer activity, especially for lung cancer - a leading cause of cancer-related deaths globally. The current paper provides the most detailed and elaborate list of the various roles of fisetin in experimentally induced lung cancer cells, and these roles include the promotion of apoptosis, inhibition of cell proliferation, migration, and invasion, as well as the regulation of autophagy. Among the molecular targets, some identified pathways, such as PI3K/Akt, MAPK, and NF-κB, that fisetin affects are crucial for tumor formation, so it can be considered a potential chemopreventive agent. Moreover, fisetin improves the effectiveness of conventional treatments as a chemo- and radiosensitizer and minimizes side effects. However, the overall utility of fisetin for clinical use is now somewhat restricted by its poor solubility and short half-life. It is predicted that the future development of nanotechnologies for drug delivery, such as nanoparticle encapsulation, might help solve these difficulties. Further Preclinical and clinical investigations are required to uniformly determine the safety, efficacy, and standard dosage of fisetin for consumption in lung cancer therapy.
Collapse
Affiliation(s)
- Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, India
| | - M Arockia Babu
- School of Pharmaceutical Sciences, GLA University, Mathura, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Arcot Rekha
- D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Saurabh Gupta
- Department of Pharmacology, Chameli Devi Institute of Pharmacy, Indore, Madhya Pradesh, India
| | | | - Anurag Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Himmat Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Mohit Agrawal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, 158223 College of Pharmacy, Northern Border University , Rafha, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abida Khan
- Department of Pharmaceutical Chemistry, 158223 College of Pharmacy, Northern Border University , Rafha, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
32
|
He H, Deng X, Wang Z, Chen J. Recent progress in the development of peptide-drug conjugates (PDCs) for cancer therapy. Eur J Med Chem 2025; 284:117204. [PMID: 39731788 DOI: 10.1016/j.ejmech.2024.117204] [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: 11/04/2024] [Revised: 12/10/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024]
Abstract
Peptide-drug conjugates (PDCs) are emerging therapeutic agents composed of peptides, linkers, and payloads, which possess favorable targeting capability and can deliver enough payloads to the tumor sites with minimized impact on healthy tissues. However, only a few PDCs have been approved for clinical use so far. To advance the research on PDCs, this review summarizes the approved PDCs, and PDCs in clinical and preclinical stages based on the payload types. Additionally, the biological activity and pharmacokinetic properties of preclinical PDCs are detailedly described. Lastly, the challenges and future development directions of PDCs are discussed. This review aims to inspire insights into the development of PDCs for cancer treatment.
Collapse
Affiliation(s)
- Haiqi He
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Deng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinic Al Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhijie Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Shenzhen Key Laboratory of Viral Oncology, Ministry of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
33
|
Lim JX, Yong YK, Dewi FRP, Chan SY, Lim V. Nanoscale strategies: doxorubicin resistance challenges and enhancing cancer therapy with advanced nanotechnological approaches. Drug Deliv Transl Res 2025:10.1007/s13346-025-01790-3. [PMID: 39955406 DOI: 10.1007/s13346-025-01790-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2025] [Indexed: 02/17/2025]
Abstract
Doxorubicin (DOX), an anthracycline, is widely used in cancer treatment by interfering RNA and DNA synthesis. Its broad antitumour spectrum makes it an effective therapy for a wide array of cancers. However, the prevailing drug-resistant cancer has proven to be a significant drawback to the success of the conventional chemotherapy regime and DOX has been identified as a major hurdle. Furthermore, the clinical application of DOX has been limited by rapid breakdown, increased toxicity, and decreased half-time life, highlighting an urgent need for more innovative delivery methods. Although advancements have been made, achieving a complete cure for cancer remains elusive. The development of nanoparticles offers a promising avenue for the precise delivery of DOX into the tumour microenvironment, aiming to increase the drug concentration at the target site while reducing side effects. Despite the good aspects of this technology, the classical nanoparticles struggle with issues such as premature drug leakage, low bioavailability, and insufficient penetration into tumours due to an inadequate enhanced permeability and retention (EPR) effect. Recent advancements have focused on creating stimuli-responsive nanoparticles and employing various chemosensitisers, including natural compounds and nucleic acids, fortifying the efficacy of DOX against resistant cancers. The efforts to refine nanoparticle targeting precision to improve DOX delivery are reviewed. This includes using receptor-mediated endocytosis systems to maximise the internalisation of drugs. The potential benefits and drawbacks of these novel techniques constitute significant areas of ongoing study, pointing to a promising path forward in addressing the challenges posed by drug-resistant cancers.
Collapse
Affiliation(s)
- Jian Xin Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Firli Rahmah Primula Dewi
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Siok Yee Chan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Pulau Pinang, Malaysia
| | - Vuanghao Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Penang, Malaysia.
| |
Collapse
|
34
|
Lim YG, Park HG, Park K. Facile One-Pot Preparation of Self-Assembled Hyaluronate/Doxorubicin Nanoaggregates for Cancer Therapy. Biomimetics (Basel) 2025; 10:91. [PMID: 39997114 PMCID: PMC11853142 DOI: 10.3390/biomimetics10020091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Hyaluronic acid (HA)-based delivery systems for doxorubicin (DOX) have been developed to selectively target cancer cells and enhance their therapeutic effects while reducing systemic side effects. However, conventional methods for preparing HA-based drug delivery systems are often limited by multistep synthetic processes, time-consuming purification, and the use of crosslinkers or surfactants, which can cause undesired toxicities. To resolve these issues, we developed a facile one-pot method to prepare self-assembled sodium hyaluronate/doxorubicin (HA/DOX) nanoaggregates by mixing HA and DOX. The self-assembled HA/DOX nanoaggregates were formed via cation-π interactions between the aromatic moiety of DOX and Na+ ions in HA as well as electrostatic interactions between HA and DOX. The optimized HA/DOX nanoaggregates with a [DOX]/[HA] molar ratio of 5 had an average particle size of approximately 250 nm and a sphere-like shape. In vitro studies revealed that HA/DOX nanoaggregates effectively targeted CD44-overexpressing cancer cells, selectively delivering DOX into the cell nuclei more efficiently than free DOX and resulting in enhanced cytotoxic effects. Annexin V and transferase dUTP nick-end labeling assays confirmed that HA/DOX nanoaggregates induced apoptosis via DNA fragmentation more effectively than free DOX.
Collapse
Affiliation(s)
| | | | - Kyeongsoon Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Gyeonggi, Republic of Korea; (Y.G.L.); (H.G.P.)
| |
Collapse
|
35
|
Iacob N, Palade P, Comanescu C, Crisan O, Toderascu LI, Socol G, Schinteie G, Kuncser V. Comprehensive Methodology for Evaluating the Drug Loading of Iron Oxide Nanoparticles Using Combined Magnetometry and Mössbauer Spectroscopy. Molecules 2025; 30:676. [PMID: 39942780 PMCID: PMC11820844 DOI: 10.3390/molecules30030676] [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: 12/17/2024] [Revised: 01/12/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
A methodology for the quantitative estimation of the drug loading of iron oxide-based magnetic nanoparticles by corroborating magnetometry and Mössbauer spectroscopy investigations is reported. The proposed methodology is exemplified in the case of two series of nanoparticles, namely Fe3O4 nanoparticles covered with citric acid molecules and further functionalized with doxorubicin, and Fe3O4 nanoparticles covered with L-Cysteine molecules and further functionalized with doxorubicin. The general idea of the proposed methodology is to probe the real magnetic structure of the magnetic core via low-temperature Mössbauer spectroscopy for the correct estimation of the spontaneous magnetization of the magnetic core. It subsequently uses the ratio between the spontaneous magnetization of the covered nanoparticles and that of the magnetic core for the reliable and nondestructive evaluation of the nanoparticle loading by organic molecules. Although the methodology is exemplified in the case of magnetite-based nanoparticles, it can be successfully considered for a large class of medicine-loaded Fe-containing magnetic nanoparticles where 57Fe Mössbauer spectroscopy can be applied.
Collapse
Affiliation(s)
- Nicusor Iacob
- National Institute of Materials Physics, 077125 Magurele, Romania; (N.I.); (P.P.); (C.C.); (O.C.)
| | - Petru Palade
- National Institute of Materials Physics, 077125 Magurele, Romania; (N.I.); (P.P.); (C.C.); (O.C.)
| | - Cezar Comanescu
- National Institute of Materials Physics, 077125 Magurele, Romania; (N.I.); (P.P.); (C.C.); (O.C.)
- Faculty of Physics, University of Bucharest, 077125 Magurele, Romania
| | - Ovidiu Crisan
- National Institute of Materials Physics, 077125 Magurele, Romania; (N.I.); (P.P.); (C.C.); (O.C.)
| | - Luiza Izabela Toderascu
- National Institute for Laser, Plasma and Radiation Physics, 077125 Magurele, Romania; (L.I.T.); (G.S.)
- Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Gabriel Socol
- National Institute for Laser, Plasma and Radiation Physics, 077125 Magurele, Romania; (L.I.T.); (G.S.)
| | - Gabriel Schinteie
- National Institute of Materials Physics, 077125 Magurele, Romania; (N.I.); (P.P.); (C.C.); (O.C.)
| | - Victor Kuncser
- National Institute of Materials Physics, 077125 Magurele, Romania; (N.I.); (P.P.); (C.C.); (O.C.)
| |
Collapse
|
36
|
Bie N, Li S, Liang Q, Zheng W, Xu S, Liu H, Zhang X, Wei Z, Yong T, Yang X, Gan L. Tumor-Repopulating Cell-Derived Microparticle-Based Therapeutics Amplify the Antitumor Effect through Synergistic Inhibition of Chemoresistance and Immune Evasion. Mol Pharm 2025; 22:733-746. [PMID: 39772575 DOI: 10.1021/acs.molpharmaceut.4c00709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Traditional chemotherapy often encounters failure attributed to drug resistance mediated by tumor-repopulating cells (TRCs) and chemotherapy-triggered immune suppression. The effective inhibition of TRCs and the mitigation of drug-induced immune suppression are pivotal for the successful chemotherapy. Here, TRC-derived microparticles (3D-MPs), characterized by excellent tumor-targeting and high TRC uptake properties, are utilized to deliver metformin and the chemotherapeutic drug doxorubicin ((DOX+Met)@3D-MPs). (DOX+Met)@3D-MPs efficiently enhance tumor accumulation and are highly internalized in tumor cells and TRCs. Additionally, (DOX+Met)@3D-MPs significantly decrease the chemotherapy-triggered upregulation in P-glycoprotein expression to enhance intracellular doxorubicin retention, resulting in increased chemotherapy sensitivity and immunogenic cell death in tumor cells and TRCs for improved antitumor immunity. Importantly, (DOX+Met)@3D-MPs also remarkably reduce chemotherapy-induced PD-L1 expression, efficiently alleviating immune suppression facilitated by the PD-L1/PD-1 axis to further enhance immunological response against malignancy. These results underscore the (DOX+Met)@3D-MPs' potential as a viable platform for augmenting the efficacy of antitumor therapies.
Collapse
Affiliation(s)
- Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingle Liang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wenxia Zheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Haojie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaojuan Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
37
|
Guo S, Zhang Y, Wang Y, Guo T, Zhu J, Chang L, Ling W, Westover KD, Zhou Z, Wei X. DCR2-targeted ultrasound nanobubbles loaded with verteporfin promote M2 macrophage polarization to overcome doxorubicin resistance in breast cancer. CHEMICAL ENGINEERING JOURNAL 2025; 505:159277. [DOI: 10.1016/j.cej.2025.159277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
|
38
|
Wernhart S, Rassaf T. Exercise, cancer, and the cardiovascular system: clinical effects and mechanistic insights. Basic Res Cardiol 2025; 120:35-55. [PMID: 38353711 PMCID: PMC11790717 DOI: 10.1007/s00395-024-01034-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/21/2024] [Accepted: 01/21/2024] [Indexed: 03/05/2024]
Abstract
Cardiovascular diseases and cancer are the leading causes of death in the Western world and share common risk factors. Reduced cardiorespiratory fitness (CRF) is a major determinant of cardiovascular morbidity and cancer survival. In this review we discuss cancer- induced disturbances of parenchymal, cellular, and mitochondrial function, which limit CRF and may be antagonized and attenuated through exercise training. We show the impact of CRF on cancer survival and its attenuating effects on cardiotoxicity of cancer-related treatment. Tailored exercise programs are not yet available for each tumor entity as several trials were performed in heterogeneous populations without adequate cardiopulmonary exercise testing (CPET) prior to exercise prescription and with a wide variation of exercise modalities. There is emerging evidence that exercise may be a crucial pillar in cancer treatment and a tool to mitigate cardiotoxic treatment effects. We discuss modalities of aerobic exercise and resistance training and their potential to improve CRF in cancer patients and provide an example of a periodization model for exercise training in cancer.
Collapse
Affiliation(s)
- Simon Wernhart
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Tienush Rassaf
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
39
|
Pal C. Small Molecules Targeting Mitochondria: A Mechanistic Approach to Combating Doxorubicin-Induced Cardiotoxicity. Cardiovasc Toxicol 2025; 25:216-247. [PMID: 39495464 DOI: 10.1007/s12012-024-09941-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Doxorubicin (Dox) is a commonly used chemotherapy drug effective against a range of cancers, but its clinical application is greatly limited by dose-dependent and cumulative cardiotoxicity. Mitochondrial dysfunction is recognized as a key factor in Dox-induced cardiotoxicity, leading to oxidative stress, disrupted calcium balance, and activation of apoptotic pathways. Recent research has emphasized the potential of small molecules that specifically target mitochondria to alleviate these harmful effects. This review provides a comprehensive analysis of small molecules that offer cardioprotection by preserving mitochondrial function in the context of doxorubicin-induced cardiotoxicity (DIC). The mechanisms of action include the reduction of reactive oxygen species (ROS) production, stabilization of mitochondrial membrane potential, enhancement of mitochondrial biogenesis, and modulation of key signaling pathways involved in cell survival and apoptosis. By targeting mitochondria, these small molecules present a promising therapeutic strategy to prevent or reduce the cardiotoxic effects associated with Dox treatment. This review not only discusses the mechanistic actions of these agents but also emphasizes their potential in improving cardiovascular outcomes for cancer patients. Gaining insight into these mechanisms can help in creating more effective strategies to safeguard the heart during chemotherapy, allowing for the ongoing use of Dox with a lower risk to the patient's cardiovascular health. This review highlights the critical role of mitochondria-targeted therapies as a promising approach in addressing DIC.
Collapse
Affiliation(s)
- Chinmay Pal
- Department of Chemistry, Gobardanga Hindu College, North 24 Parganas, West Bengal, 743273, India.
| |
Collapse
|
40
|
Zhuang F, Huang S, Liu L. MALSU1-mediated regulation of mitochondrial function governs proliferation and doxorubicin resistance in triple-negative breast cancer cells. Mol Cell Biochem 2025; 480:1197-1207. [PMID: 38896203 DOI: 10.1007/s11010-024-05053-6] [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: 01/21/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
Triple-negative breast cancer (TNBC) poses a formidable challenge in oncology due to its aggressive nature and limited treatment options. Although doxorubicin, a widely used chemotherapeutic agent, shows efficacy in TNBC treatment, acquired resistance remains a significant obstacle. Our study explores the role of MALSU1, a regulator of mitochondrial translation, in TNBC and its impact on cell proliferation and doxorubicin resistance. We observed increased MALSU1 expression in TNBC, correlating with poor patient prognosis. MALSU1 knockdown in TNBC cells significantly reduced proliferation, indicating its pivotal role in sustaining cell growth. Mechanistically, MALSU1 depletion resulted in decreased activities of mitochondrial respiratory chain complexes, cellular ATP levels, and mitochondrial respiration. Notably, exogenous addition of normal mitochondria restored proliferation and mitochondrial respiration in MALSU1-depleted TNBC cells. Importantly, MALSU1 knockdown enhanced the sensitivity of doxorubicin-resistant TNBC cells to doxorubicin treatment. Furthermore, pharmacological inhibition of mitochondrial translation using tigecycline and chloramphenicol mimicked the effects of MALSU1 knockdown, suggesting mitochondrial translation as a potential therapeutic target. Taken together, our findings not only elucidate the intricate role of MALSU1 in TNBC biology and doxorubicin resistance but also lay the groundwork for future investigations targeting MALSU1 and/or mitochondrial translation as a promising avenue for developing innovative therapeutic strategies against TNBC.
Collapse
Affiliation(s)
- Feifei Zhuang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Shaoyan Huang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Lei Liu
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China.
| |
Collapse
|
41
|
Song B, Wu M, Qin L, Liang W, Wang X. Smart Design of Targeted Drug Delivery System for Precise Drug Delivery and Visual Treatment of Brain Gliomas. Adv Healthc Mater 2025; 14:e2402967. [PMID: 39707642 DOI: 10.1002/adhm.202402967] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/04/2024] [Indexed: 12/23/2024]
Abstract
In the treatment of glioma, which is one of the malignant tumors, although chemotherapy is used as the most common treatment method, it often suffers from low bioavailability. Therefore, improving the precision and efficiency of drugs is crucial in treating gliomas and a great challenge. Here, an advanced drug delivery system is reported for gliomas (CZQD@HA@DOX), which aggregates multiple features such as the susceptible imaging tracer property due to the use of CZQD and the targeting of HA to the receptor cluster 44 (CD44) of glioma cells, which provides the system with the functions of targeted enrichment and precise drug delivery at the tumor site. The pH-responsive drug delivery system has not only an excellent encapsulation rate but also a high drug loading capacity, and the doxorubicin loaded on it can be released centrally at the tumor microenvironment site and causes an increase of reactive oxygen species in the mitochondria and trigger oxidative stress, which leads to high expression of Bax apoptotic proteins, ultimately activating the mitochondrial pathway-mediated apoptotic process in glioma cells. Overall, this drug delivery system has great potential for application in precision targeted therapy and visual tracer imaging of gliomas.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Mengru Wu
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Lijing Qin
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| |
Collapse
|
42
|
Dong T, Li J, Liang X, Wang W, Chen M, Yang G, Wu D. Cannabidiol Ameliorates Doxorubicin-Induced Myocardial Injury via Activating Hippo Pathway. Drug Des Devel Ther 2025; 19:569-583. [PMID: 39876987 PMCID: PMC11774276 DOI: 10.2147/dddt.s497323] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Background Doxorubicin (DOX) is a chemotherapeutic agent widely used for cancer treatment and has non-negligible cardiotoxicity. Some previous studies have reported that cannabidiol (CBD) has cardioprotective effects. In this study, we evaluated the protective effects of CBD against DOX-induced cardiomyocyte injury, and explored the downstream molecular mechanism. Methods and Materials GSE193861, containing healthy myocardial tissues and myocardial tissues with DOX-induced injury, was analyzed to screen for the involved proteins and pathways. Molecular docking was performed to identify candidate drugs. After H9c2 cells were treated with DOX and CBD, their viability, oxidative stress, and apoptosis were assessed. After YAP depletion, the role of the Hippo pathway in CBD function was investigated. C57BL/6 mice were treated with DOX to establish an in vivo model, and CBD and verteporfin (VP) were used to treat the mice. Histological analyses and immunofluorescence were used to evaluate myocardial tissue injury, and apoptosis and oxidative stress of the myocardial tissues were also analyzed. Western blotting was used to investigate the regulatory effects of CBD on the Hippo and apoptosis-related pathways. Results Bioinformatic analysis suggested that the Hippo pathway was a crucial pathway involved in DOX-induced myocardial injury. Molecular docking showed that CBD targeted multiple regulators of the Hippo pathway. CBD showed cardioprotective effects against DOX-induced myocardial injury both in vitro and in vivo and regulated Hippo pathway activity in cardiomyocytes. After inactivation of the Hippo pathway by YAP knockdown or VP intervention, the protective effects of CBD were reversed. Conclusion For the first time, we revealed that CBD is likely to reduce DOX-induced myocardial injury by regulating the Hippo signaling pathway.
Collapse
Affiliation(s)
- Tianwei Dong
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, 154000, People’s Republic of China
| | - Jinlian Li
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, 154007, People’s Republic of China
| | - Xinfang Liang
- Department of Cardiology, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, 154002, People’s Republic of China
| | - Wang Wang
- Department of Cardiology, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, 154002, People’s Republic of China
| | - Meichi Chen
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, 154000, People’s Republic of China
| | - Guangyuan Yang
- Cardiovascular Medicine, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, 154002, People’s Republic of China
| | - Dongmei Wu
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, 154000, People’s Republic of China
| |
Collapse
|
43
|
Lee J, Kim Y, Jung HI, Lim J, Kwak BS. Channel-assembling tumor microenvironment on-chip for evaluating anticancer drug efficacy. J Control Release 2025; 377:376-384. [PMID: 39566854 DOI: 10.1016/j.jconrel.2024.11.030] [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: 09/02/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Organ-on-a-chip is an advanced system for evaluating drug response in diseases. It simulates the in vivo tumor microenvironment, aiding in the understanding of drug mechanisms and tumor responses. It mimics the structure of the tumor microenvironment and the dynamic conditions within the body. As a result, it holds the potential for applications in precision and personalized medicine. However, there are still limitations in sequential development processes and complex structures, resulting in time-consuming molecular interference during system development. In this study, we developed a channel-assembling tumor microenvironment-on-chip (CATOC) system to overcome these limitations. CATOC was easily segmented into blood vessels and a tumor microenvironment-on-chip, which can be independently developed. The tumor microenvironment-on-chip consists of two independent channels for evaluating drug responses in different types of tumor microenvironments. Each fully developed system was physically interconnected to create a CATOC. Interconnected CATOC was used to validate chemical and targeted anticancer drug responses in different subtypes of the breast tumor microenvironment. We also emphasized the significance of on-chip experiments by observing the drug response of tumor spheroids on CATOC and scaffold-free platforms.
Collapse
Affiliation(s)
- Jaehun Lee
- Yonsei University, School of Mechanical Engineering, 50 Yonsei-ro, Seadaemun-gu, Seoul 03722, Republic of Korea; Dongguk University, College of Medicine, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do 10326, Republic of Korea
| | - Youngwon Kim
- Yonsei University, School of Mechanical Engineering, 50 Yonsei-ro, Seadaemun-gu, Seoul 03722, Republic of Korea; Dongguk University, College of Medicine, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do 10326, Republic of Korea
| | - Hyo-Il Jung
- Yonsei University, School of Mechanical Engineering, 50 Yonsei-ro, Seadaemun-gu, Seoul 03722, Republic of Korea; The DABOM Inc., 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jiseok Lim
- MediSphere Inc., Gyeongsan-si, Gyeongsanbuk-do, Republic of Korea; Yeungnam University, School of Mechanical Engineering, 280 Daehak-ro, Gyeongsan-si, Gyeongsangbuk-do 38541, Republic of Korea.
| | - Bong Seop Kwak
- Dongguk University, College of Medicine, 32 Dongguk-ro, Ilsandong-gu, Goyangsi, Gyeonggi-do 10326, Republic of Korea; MediSphere Inc., Gyeongsan-si, Gyeongsanbuk-do, Republic of Korea.
| |
Collapse
|
44
|
Zhang W, Yang X, Qu Z, Ding P, Kong X, Wang X, Liu Q, Zhang X, Lu Y, Wang J, Chen Z, Fang Y. DNA tetrahedral nanoparticles: Co-delivery of siOTUD6B/DOX against triple-negative breast cancer. J Control Release 2025; 377:197-211. [PMID: 39549731 DOI: 10.1016/j.jconrel.2024.11.025] [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: 04/14/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited targeted therapeutic options. Recently, the deubiquitinizing enzyme ovarian tumor domain-containing 6B (OTUD6B) has been reported to play a potential role in TNBC progression. Therefore, this study investigates the role and underlying molecular mechanisms of OTUD6B in vitro and xenograft models of TNBC. Specifically, we examined the therapeutic effects of siOTUD6B and doxorubicin (DOX) co-delivery using synthesized tetrahedral DNA nanoparticles (Tds) on tumor growth and progression. Additionally, the uptake and efficacy of the siOTUD6B/DOX@Td in TNBC cells were evaluated. Notably, the siOTUD6B/DOX@Td nanoparticle demonstrated efficient cellular uptake by TNBC cells, resulting in OTUD6B knockdown and controlled release of DOX. Additionally, siOTUD6B/DOX@Td treatment enhanced apoptosis rates increased DOX sensitivity, and inhibited TNBC cell growth, migration, and metastasis. Moreover, in vivo experiments confirmed that siOTUD6B/DOX@Td treatment inhibited tumor growth and metastasis without damaging the primary organs. Mechanistically, OTUD6B regulates TNBC progression by stabilizing murine double minute 2 (MDM2) and degrading forkhead box O3a (FOXO3a). Conclusively, this study demonstrates the potential applicability of DNA nanoparticles loaded with DOX and siOTUD6B for TNBC treatment.
Collapse
Affiliation(s)
- Wenxiang Zhang
- Department of Breast Surgical 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
| | - Xue Yang
- Department of Breast Surgical 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
| | - Zheng Qu
- Department of Breast Surgical 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
| | - Peikai Ding
- Department of Breast Surgical 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
| | - Xiangyi Kong
- Department of Breast Surgical 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
| | - Xiangyu Wang
- Department of Breast Surgical 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
| | - Qiang Liu
- Department of Breast Surgical 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
| | - Xingsong Zhang
- Pooling Medical Research Institutes of 100Biotech, Beijing 100006, China
| | - Ye Lu
- Department of Breast Surgical 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
| | - Jing Wang
- Department of Breast Surgical 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.
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing 100006, China.
| | - Yi Fang
- Department of Breast Surgical 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
|
45
|
Lu C, Wei J, Gao C, Sun M, Dong D, Mu Z. Molecular signaling pathways in doxorubicin-induced nephrotoxicity and potential therapeutic agents. Int Immunopharmacol 2025; 144:113373. [PMID: 39566381 DOI: 10.1016/j.intimp.2024.113373] [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: 08/11/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024]
Abstract
Doxorubicin (DOX), an anthracycline chemotherapeutic agent, is extensively utilized in the clinical management of both solid and hematological malignancies. Nevertheless, the clinical application of this treatment is significantly limited by adverse reactions and toxicity that may arise during or after administration. Its cytotoxic effects are multifaceted, with cardiotoxicity being the most prevalent side effect. Furthermore, it has the potential to adversely affect other organs, including the brain, kidneys, liver, and so on. Notably, it has been reported that DOX may cause renal failure in patients and there is currently no effective treatment for DOX-induced kidney damage, which has raised a high concern about DOX-induced nephrotoxicity (DIN). Although the precise molecular mechanisms underlying DIN remain incompletely elucidated, prior research has indicated that reactive oxygen species (ROS) are pivotal in this process, triggering a cascade of detrimental pathways including apoptosis, inflammation, dysregulated autophagic flux, and fibrosis. In light of these mechanisms, decades of research have uncovered several DIN-associated signaling pathways and found multiple potential therapeutic agents targeting them. Thus, this review intends to delineate the DIN associated signaling pathways, including AMPK, JAKs/STATs, TRPC6/RhoA/ROCK1, YAP/TEAD, SIRTs, Wnt/β-catenin, TGF-β/Smad, MAPK, Nrf2/ARE, NF-κB, and PI3K/AKT, and to summarize their potential regulatory agents, which provide a reference for the development of novel medicines against DIN.
Collapse
Affiliation(s)
- Changxu Lu
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China; Department of Urology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Can Gao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China.
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China.
| | - Zhongyi Mu
- Department of Urology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
46
|
Ge W, Zhang X, Lin J, Wang Y, Zhang X, Duan Y, Dai X, Zhang J, Zhang Y, Jiang M, Qiang H, Zhao Z, Zhang X, Sun D. Rnd3 protects against doxorubicin-induced cardiotoxicity through inhibition of PANoptosis in a Rock1/Drp1/mitochondrial fission-dependent manner. Cell Death Dis 2025; 16:2. [PMID: 39755713 DOI: 10.1038/s41419-024-07322-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
Doxorubicin, a representative drug of the anthracycline class, is widely used in cancer treatment. However, Doxorubicin-induced cardiotoxicity (DIC) presents a significant challenge in its clinical application. Mitochondrial dysfunction plays a central role in DIC, primarily through disrupting mitochondrial dynamics. This study aimed to investigate the impact of Rnd3 (a Rho family GTPase 3) on DIC, with a focus on mitochondrial dynamics. Cardiomyocyte-specific Rnd3 transgenic mice (Rnd3-Tg) and Rnd3LSP/LSP mice (N-Tg) were established for in vivo experiments, and adenoviruses harboring Rnd3 (Ad-Rnd3) or negative control (Ad-Control) were injected in the myocardium for in vitro experiments. The DIC model was established using wild-type, N-Tg, and Rnd3-Tg mice, with subsequent intraperitoneal injection of Dox for 4 weeks. The molecular mechanism was explored through RNA sequencing, immunofluorescence staining, co-immunoprecipitation assay, and protein-protein docking. Dox administration induced significant mitochondrial injury and cardiac dysfunction, which was ameliorated by Rnd3 overexpression. Further, the augmentation of Rnd3 expression mitigated mitochondrial fragmentation which is mediated by dynamin-related protein 1 (Drp1), thereby ameliorating the PANoptosis (pyroptosis, apoptosis, and necroptosis) response induced by Dox. Mechanically, the interaction between Rnd3 and Rho-associated kinase 1 (Rock1) may impede Rock1-induced Drp1 phosphorylation at Ser616, thus inhibiting mitochondrial fission and dysfunction. Interestingly, Rock1 knockdown nullified the effects of Rnd3 on cardiomyocytes PANoptosis, as well as Dox-induced cardiac remodeling and dysfunction elicited by Rnd3. Rnd3 enhances cardiac resilience against DIC by stabilizing mitochondrial dynamics and reducing PANoptosis. Our findings suggest that the Rnd3/Rock1/Drp1 signaling pathway represents a novel target for mitigating DIC, and modulating Rnd3 expression could be a strategic approach to safeguarding cardiac function in patients undergoing Dox treatment. The graphical abstract illustrated the cardioprotective role of Rnd3 in DIC. Rnd3 directly binds to Rock1 in cytoplasm and ameliorates mitochondrial fission by inhibiting Drp1 phosphorylation at ser616, thereby alleviating PANoptosis (apoptosis, pyroptosis, and necroptosis) in DIC.
Collapse
Affiliation(s)
- Wen Ge
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaohua Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yangyang Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yu Duan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xinchun Dai
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huanhuan Qiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhijing Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
47
|
Zhai Y, Zhang W, Wang J, Kong Y, Rong R, Lang T, Zheng C, Wang Y, Yu Y, Zhu HH, Cai Y, Zhang P, Li Y. Interleukin 15-Presenting Nanovesicles with Doxorubicin-Loaded Ferritin Cores for Cancer Immunochemotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409194. [PMID: 39625860 PMCID: PMC11789581 DOI: 10.1002/advs.202409194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/13/2024] [Indexed: 01/30/2025]
Abstract
Interleukin 15 (IL15) is crucial for fostering the survival and proliferation of nature killer (NK) cells and cytotoxic T lymphocytes (CTLs), playing a pivotal role in tumor control. However, IL15 supplementary therapy encounters challenges such as systemic inflammation and non-specific stimulation of cancer cells. Herein, a nanovesicle termed DoxFILN, comprising a membrane presenting IL15/IL15 receptor α complexes (IL15c) and a core of doxorubicin-loaded ferritin (Dox-Fn) are reported. The DoxFILN significantly enhances the densities and activities of intratumoral CTLs and NK cells. Mechanistically, DoxFILN undergoes deshelling in the acidic tumor microenvironment, releasing Dox-Fn and membrane-bound IL15c. Dox-Fn selectively target transferrin receptors on cancerous cells, facilitating intracellular Dox release and inducing immunogenic cell death. Concurrently, membrane-bound IL15c recognizes and activates IL15 receptor β/γc heterodimers, leading to a remarkable increase in the proliferation and activation of CTLs (16-fold and 28-fold) and NK cells (37-fold and 50-fold). The IL15-displaying nanovesicle introduced here holds promise as a potential platform for immunochemotherapy in the treatment of cancer.
Collapse
Affiliation(s)
- Yihui Zhai
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wen Zhang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- China State Institute of Pharmaceutical IndustryShanghai201203China
| | - Jinming Wang
- State Key Laboratory of Oncogenes and Related GenesRenji‐Med‐X Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200127China
| | - Ying Kong
- Yantai Institute of Materia MedicaShandong264000China
| | - Rong Rong
- Yantai Institute of Materia MedicaShandong264000China
| | - Tianqun Lang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Chao Zheng
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- China State Institute of Pharmaceutical IndustryShanghai201203China
| | - Yanke Wang
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai201210China
| | - Yang Yu
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai201210China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related GenesRenji‐Med‐X Stem Cell Research CenterDepartment of UrologyRen Ji HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200127China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine & Advanced PreparationsYantai Institute of Materia MedicaShandong264000China
| | - Pengcheng Zhang
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and DevicesShanghaiTech UniversityShanghai201210China
- Shanghai Clinical Research and Trial CenterShanghai201203China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine & Advanced PreparationsYantai Institute of Materia MedicaShandong264000China
| |
Collapse
|
48
|
Bisht A, Avinash D, Sahu KK, Patel P, Das Gupta G, Kurmi BD. A comprehensive review on doxorubicin: mechanisms, toxicity, clinical trials, combination therapies and nanoformulations in breast cancer. Drug Deliv Transl Res 2025; 15:102-133. [PMID: 38884850 DOI: 10.1007/s13346-024-01648-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Doxorubicin is a key treatment for breast cancer, but its effectiveness often comes with significant side effects. Its actions include DNA intercalation, topoisomerase II inhibition, and reactive oxygen species generation, leading to DNA damage and cell death. However, it can also cause heart problems and low blood cell counts. Current trials aim to improve doxorubicin therapy by adjusting doses, using different administration methods, and combining it with targeted treatments or immunotherapy. Nanoformulations show promise in enhancing doxorubicin's effectiveness by improving drug delivery, reducing side effects, and overcoming drug resistance. This review summarizes recent progress and difficulties in using doxorubicin for breast cancer, highlighting its mechanisms, side effects, ongoing trials, and the potential impact of nanoformulations. Understanding these different aspects is crucial in optimizing doxorubicin's use and improving outcomes for breast cancer patients. This review examines the toxicity of doxorubicin, a drug used in breast cancer treatment, and discusses strategies to mitigate adverse effects, such as cardioprotective agents and liposomal formulations. It also discusses clinical trials evaluating doxorubicin-based regimens, the evolving landscape of combination therapies, and the potential of nanoformulations to optimize delivery and reduce systemic toxicity. The review also discusses the potential of liposomes, nanoparticles, and polymeric micelles to enhance drug accumulation within tumor tissues while sparing healthy organs.
Collapse
Affiliation(s)
- Anjali Bisht
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Dubey Avinash
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, 17 km Stone, NH-2, Chaumuhan, Mathura, 281406, UP, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
49
|
Tayal R, Mannan A, Singh S, Dhiman S, Singh TG. Unveiling the Complexities: Exploring Mechanisms of Anthracyclineinduced Cardiotoxicity. Curr Cardiol Rev 2025; 21:42-77. [PMID: 39484769 PMCID: PMC12060933 DOI: 10.2174/011573403x322928241021100631] [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: 04/12/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 11/03/2024] Open
Abstract
The coexistence of cancer and heart disease, both prominent causes of illness and death, is further exacerbated by the detrimental impact of chemotherapy. Anthracycline-induced cardiotoxicity is an unfortunate side effect of highly effective therapy in treating different types of cancer; it presents a significant challenge for both clinicians and patients due to the considerable risk of cardiotoxicity. Despite significant progress in understanding these mechanisms, challenges persist in identifying effective preventive and therapeutic strategies, rendering it a subject of continued research even after three decades of intensive global investigation. The molecular targets and signaling pathways explored provide insights for developing targeted therapies, emphasizing the need for continued research to bridge the gap between preclinical understanding and clinical applications. This review provides a comprehensive exploration of the intricate mechanisms underlying anthracycline-induced cardiotoxicity, elucidating the interplay of various signaling pathways leading to adverse cellular events, including cardiotoxicity and death. It highlights the extensive involvement of pathways associated with oxidative stress, inflammation, apoptosis, and cellular stress responses, offering insights into potential and unexplored targets for therapeutic intervention in mitigating anthracycline-induced cardiac complications. A comprehensive understanding of the interplay between anthracyclines and these complexes signaling pathways is crucial for developing strategies to prevent or mitigate the associated cardiotoxicity. Further research is needed to outline the specific contributions of these pathways and identify potential therapeutic targets to improve the safety and efficacy of anthracycline-based cancer treatment. Ultimately, advancements in understanding anthracycline-induced cardiotoxicity mechanisms will facilitate the development of more efficacious preventive and treatment approaches, thereby improving outcomes for cancer patients undergoing anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- Rohit Tayal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
50
|
Tuncbilek Z, Cakmak NK, Tas A, Ayan D, Silig Y. PEGylated Titanium Dioxide Nanoparticle-bound Doxorubicin and Paclitaxel Drugs Affect Prostate Cancer Cells and Alter the Expression of DUSP Family Genes. Anticancer Agents Med Chem 2025; 25:257-271. [PMID: 39473102 DOI: 10.2174/0118715206330115241015092548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/24/2024] [Accepted: 09/09/2024] [Indexed: 03/25/2025]
Abstract
BACKGROUND Prostate cancer (PC) is among the cancer types with high incidence and mortality. New and effective strategies are being sought for the treatment of deadly cancers, such as PC. In this context, the use of nanocarrier systems containing titanium dioxide (TiO2) can improve treatment outcomes and increase the effectiveness of anticancer drugs. OBJECTIVE This study aimed to evaluate the cytotoxic activity of doxorubicin (DOX) and paclitaxel (PTX) drugs on the PC cell line by attaching them to PEGylated TiO2 nanoparticles and to examine their effect on the expression levels of dual-specificity phosphatase (DUSP) genes. METHODS Free DOX and PTX drugs, DOX and PTX compounds bound to the pegylated TiO2 system were applied to DU-145 cells, a PC cell line, under in vitro conditions, and MTT analysis was performed. Additionally, the IC50 values of these compounds were analyzed. In addition, the expression levels of DUSP1, DUSP2, DUSP4, DUSP6, and DUSP10 genes were measured using RT-PCR. Additionally, bioinformatics and molecular docking analyses were performed on DUSP proteins. RESULTS The cytotoxic activity of PTX compound bound to PEGylated TiO2 was found to be higher than that of DOX compound bound to PEGylated TiO2. Additionally, when the expression levels were compared to the control group, the expression levels of DUSPs were found to be lower in the drugs of the drug carrier systems. CONCLUSION Accordingly, it was predicted that the PEGylated TiO2 nano-based carrier could be effective in PC.
Collapse
Affiliation(s)
- Zuhal Tuncbilek
- Department of Chemistry and Chemical Technologies, Yildizeli Vocational School, Sivas Cumhuriyet University, Sivas, 58500, Türkiye
| | - Nese Keklikcioglu Cakmak
- Department of Chemical Engineering, Faculty of Engineering, Sivas Cumhuriyet University, Sivas, 58140, Türkiye
| | - Ayca Tas
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Sivas Cumhuriyet University, Sivas, 58140, Türkiye
| | - Durmus Ayan
- Department of Medical Biochemistry, Faculty of Medicine, Nigde Omer Halisdemir University, Niğde, 51240, Türkiye
| | - Yavuz Silig
- Department of Medical Biochemistry, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, 58140, Türkiye
| |
Collapse
|