1
|
Li G, Che X, Wang S, Liu D, Xie D, Jiang B, Zheng Z, Zheng X, Wu G. The role of cisplatin in modulating the tumor immune microenvironment and its combination therapy strategies: a new approach to enhance anti-tumor efficacy. Ann Med 2025; 57:2447403. [PMID: 39757995 PMCID: PMC11705547 DOI: 10.1080/07853890.2024.2447403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/27/2024] [Accepted: 11/23/2024] [Indexed: 01/07/2025] Open
Abstract
Cisplatin is a platinum-based drug that is frequently used to treat multiple tumors. The anti-tumor effect of cisplatin is closely related to the tumor immune microenvironment (TIME), which includes several immune cell types, such as the tumor-associated macrophages (TAMs), cytotoxic T-lymphocytes (CTLs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), and natural killer (NK) cells. The interaction between these immune cells can promote tumor survival and chemoresistance, and decrease the efficacy of cisplatin monotherapy. Therefore, various combination treatment strategies have been devised to enhance patient responsiveness to cisplatin therapy. Cisplatin can augment anti-tumor immune responses in combination with immune checkpoint blockers (such as PD-1/PD-L1 or CTLA4 inhibitors), lipid metabolism disruptors (like FASN inhibitors and SCD inhibitors) and nanoparticles (NPs), resulting in better outcomes. Exploring the interaction between cisplatin and the TIME will help identify potential therapeutic targets for improving the treatment outcomes in cancer patients.
Collapse
Affiliation(s)
- Guandu Li
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shijin Wang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Deqian Xie
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bowen Jiang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zunwen Zheng
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xu Zheng
- Department of Cell Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
2
|
IJff M, Mei X, Scutigliani EM, Rodermond HM, van Bochove GGW, Krawczyk PM, Franken NAP, Stalpers LJA, Crezee J, Oei AL. Addition of PARP1-inhibition enhances chemoradiotherapy and thermoradiotherapy when treating cervical cancer in an in vivo mouse model. Int J Hyperthermia 2025; 42:2450514. [PMID: 39837264 DOI: 10.1080/02656736.2025.2450514] [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: 02/01/2024] [Revised: 10/18/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Background: Efficacy of current treatment options for cervical cancer require improvement. Previous in vitro studies have shown the enhancing effects of the addition of PARP1-inhibitors to chemoradiotherapy and thermoradiotherapy. The aim of our present study was to test efficacy of different combinations of treatment modalities radiotherapy, cisplatin, hyperthermia and PARP1-inhibitors using in vitro tumor models, ex vivo treated patient samples and in vivo tumor models. Materials and Methods: In vitro clonogenic survival curves (0-6 Gy) show that PARP1-i (4-5 M Olaparib) enhances both chemoradiotherapy (0.3-0.5 µM cisplatin) and thermoradiotherapy (42 °C for 1 h) in SiHa, CaSki and HeLa cells. A cervical cancer mouse model and freshly obtained in-house developed patient-derived organoids were used to examine the effects of different treatment combinations. For the in vivo study, human cervical cancer (SiHa) cells were injected in the right hind leg of athymic nude mice. In vivo mouse experiments show that PARP1-i enhances thermoradiotherapy or chemoradiotherapy by reduction of tumor volumes. Five cycles of treatment were applied with the following doses per cycle: irradiation 3 Gy, hyperthermia 1 h at 42 °C, cisplatin at 2 mg/kg, and twice PARP1-i at 50 mg/kg. Results: Quadruple treatment, combining radiotherapy, hyperthermia, cisplatin and PARP1-i, was very effective but also lead to severe side effects causing severe weight loss and death. In contrast, thermoradiotherapy or chemoradiotherapy with addition of PARP1-i, were effective without serious side effects. Conclusion: The triple combinations are promising options for potentially more effective treatment of locally advanced cervical cancer without more toxicity.
Collapse
Affiliation(s)
- Marloes IJff
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Xionge Mei
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Enzo M Scutigliani
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Hans M Rodermond
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Gregor G W van Bochove
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Przemek M Krawczyk
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicolaas A P Franken
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Lukas J A Stalpers
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Arlene L Oei
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Raouf AA, El-Kadem AH, Sokar SS, Oraby MA, El-Shitany NA. Cilostazol attenuates cisplatin-induced acute liver injury by targeting the SIRT1/AMPK/PGC-1α signaling pathway, with an impact on miRNA-34a. Eur J Pharmacol 2025; 997:177609. [PMID: 40216180 DOI: 10.1016/j.ejphar.2025.177609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
The dominant chemotherapeutic agent, cisplatin (CP), is widely used to manage various cancer types. Despite its effectiveness, CP use is associated with severe hepatotoxicity. Cilostazol (CSZ), a selective phosphodiesterase III inhibitor, has recently demonstrated remarkable anti-inflammatory and anti-apoptotic properties in different diseases. Additionally, it exhibits hepatoprotective effects against various forms of liver injury. Hence, this study aimed to assess the potential hepatoprotective and ameliorative effects of CSZ on CP-induced acute liver injury (ALI) and to elucidate the underlying molecular mechanisms. To achieve this, ALI was induced by a single injection of CP (20 mg/kg; i.p.) in male Wistar rats pretreated with CSZ (5 or 10 mg/kg) administered orally for one week. The findings revealed that CSZ effectively reversed CP-induced hepatic dysfunction, as evidenced by notable liver function tests and improvements in histological examination. Additionally, CSZ protected against CP-mediated liver oxidative stress by decreasing MDA levels while increasing GSH and GPx levels and enhancing SOD activity. Furthermore, CSZ exhibited a potent anti-inflammatory effect, reducing the expression of pro-inflammatory cytokines, including NF-κB, IL-1β, and TNF-α. Regarding hepatocyte apoptosis, CSZ suppressed Bax immunoexpression and caspase-3 and caspase-9 levels while enhancing Bcl-2 expression, thereby mitigating hepatic cell death. The hepatoprotective effects of CSZ could be attributed to the regulation of the miRNA-34a/AMPK/SIRT1/PGC-1α signaling pathway, leading to the activation of the Nrf2/HO-1-mediated antioxidative defense mechanism. In conclusion, CSZ could be a promising therapeutic agent for preventing CP-induced ALI, potentially improving the quality of life for cancer patients.
Collapse
Affiliation(s)
- Ahmed Amr Raouf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
| | - Aya H El-Kadem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Samia S Sokar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Mamdouh A Oraby
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
| | - Nagla A El-Shitany
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
4
|
Wang JQ, Liu XM, Zhu ZS, Li Z, Xie CZ, Qiao X, Feng YK, Xu JY. Fluoxetine-Conjugated Platinum(IV) Prodrugs Targeting eEF2K and Conquering Multidrug Resistance against Triple-Negative Breast Cancer. J Med Chem 2025; 68:9661-9680. [PMID: 40265570 DOI: 10.1021/acs.jmedchem.5c00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Triple-negative breast cancer (TNBC) poses formidable challenges in the clinic owing to its particularly malignant and aggressive properties. Overexpression of eukaryotic elongation factor-2 kinase (eEF2K) is highly correlated with the poor prognosis of TNBC, representing a promising therapeutic target. Herein, Fluoxetine as eEF2K-inhibitor and chemosensitizer was conjugated with cisplatin/oxaliplatin to present two-in-one prodrugs 8-19. Multievaluation indicated that monosubstituted 8 and disubstituted 12 exhibited 407- and 174-fold higher cytotoxicity than cisplatin against MDA-MB-231 cells by elevating DNA damage-induced apoptosis and eEF2K-triggered autophagy. Moreover, 8 and 12 significantly overcame chemoresistance in A549cisR cells, evidenced by downregulating resistance-related key proteins P-gp, GST-π, ATM, and RAD51. Syngeneic and xenograft mouse models demonstrated that 8 and 12 could effectively inhibit tumor growth and metastasis, and reduce toxicity compared to cisplatin in vivo. Additionally, 8 and 12 stimulated immunomodulation including T-cell proliferation and Th1 cytokine production. All results hold the promise of 8 and 12 as multifunctional chemotherapeutic agents.
Collapse
Affiliation(s)
- Jia-Qian Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xiao-Meng Liu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zi-Shuo Zhu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Li
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Cheng-Zhi Xie
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yu-Kuan Feng
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
5
|
Xiang H, Sun J, Kong L, Wang Y, Qiu X, Zeng J, Li G, He J. NDUFA8 promotes cell viability and inhibits ferroptosis and cisplatin sensitivity by stabilizing Fe-S clusters in cervical cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04237-1. [PMID: 40335672 DOI: 10.1007/s00210-025-04237-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/27/2025] [Indexed: 05/09/2025]
Abstract
Cervical cancer (CC) ranks among the primary causes of cancer fatalities in women, with cisplatin (DDP) resistance significantly impacting clinical outcomes. NADH dehydrogenase (ubiquinone) FA8 (NDUFA8) is significantly upregulated in CC tissues and correlates with lower survival rates, but its role in cisplatin sensitivity in CC is still unclear. NDUFA8 silencing inhibited CC cell viability, promoted ferroptosis, evidenced by increased Fe2+ and lipid ROS levels, along with decreased levels of ATP and reduced activities of complex I, aconitase (ACO), and xanthine oxidase (XO). However, overexpression of NDUFA8 promoted CC cell viability, inhibited ferroptosis, and increased levels of ATP and activities of complex I, ACO, and XO in ferric ammonium citrate (FAC) or rotenone-treated CC cells. NDUFA8 expression showed a negative correlation with the DDP therapy response in CC tissues and cell lines. However, in CC tissues, NDUFA8 expression was positively associated with ACO and XO activities. In in vivo experiments, the overexpression of NDUFA8 diminished the anti-tumor effects of DDP, which was counteracted by FAC. NDUFA8 promotes cell viability and inhibits ferroptosis and DDP sensitivity by stabilizing Fe-S clusters in CC.
Collapse
Affiliation(s)
- Huaguo Xiang
- Department of Clinical Laboratory, Fuyong People's Hospital of Baoan District, 81 Defeng Road, Fuyong Street, Shenzhen, 518103, China.
| | - Junfang Sun
- Department of Clinical Laboratory, Fuyong People's Hospital of Baoan District, 81 Defeng Road, Fuyong Street, Shenzhen, 518103, China
| | - Lingyue Kong
- Department of Clinical Laboratory, Fuyong People's Hospital of Baoan District, 81 Defeng Road, Fuyong Street, Shenzhen, 518103, China
| | - Yingzhen Wang
- Department of Clinical Laboratory, Fuyong People's Hospital of Baoan District, 81 Defeng Road, Fuyong Street, Shenzhen, 518103, China
| | - Xiaorou Qiu
- Department of Clinical Laboratory, Fuyong People's Hospital of Baoan District, 81 Defeng Road, Fuyong Street, Shenzhen, 518103, China
| | - Jinting Zeng
- Department of Clinical Laboratory, Fuyong People's Hospital of Baoan District, 81 Defeng Road, Fuyong Street, Shenzhen, 518103, China
| | - Guo Li
- Department of Clinical Laboratory, Fuyong People's Hospital of Baoan District, 81 Defeng Road, Fuyong Street, Shenzhen, 518103, China
| | - Jiehong He
- Department of Clinical Laboratory, Fuyong People's Hospital of Baoan District, 81 Defeng Road, Fuyong Street, Shenzhen, 518103, China
| |
Collapse
|
6
|
Iglesias-Jiménez A, Artiaga G, Moreno-Gordaliza E, Milagros Gómez-Gómez M. Metallomic evaluation of selenium nanoparticles and selenomethionine for the attenuation of cisplatin-induced nephrotoxicity. Eur J Pharm Biopharm 2025:114737. [PMID: 40345401 DOI: 10.1016/j.ejpb.2025.114737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Nephrotoxicity is one of the most limiting side effects in oncologic patients treated with cisplatin and is still clinically unresolved. In this work, chitosan-stabilised selenium nanoparticles (Ch-SeNPs) and selenomethionine (SeMet) have been evaluated as nephroprotectors of cisplatin using renal proximal tubule epithelial cells (RPTEC/TERT1) as a model. Moreover, the antineoplastic efficacy of cisplatin co-administered with these selenocompounds has been tested in cervical cancer cells (HeLa). Cell viability, cell localisation of Ch-SeNPs and changes in the morphology and cell ultrastructure, Pt and Se cellular internalisation and cisplatin binding to DNA, and speciation of Pt and Se in the cytosolic extracts were evaluated by MTT assays, transmission electron microscopy coupled to energy dispersive X-ray spectroscopy (TEM-EDS), inductively coupled plasma mass spectrometry (ICP-MS), and both size exclusion chromatography (SEC) and anion exchange chromatography (AEC) coupled to either ICP-MS or UV-Vis. Differences in the pharmacological activity of the two selenospecies were observed. SeMet exerted a moderate protection on kidney cells while reducing their degree of cisplatin intracellular accumulation and DNA binding in both cell lines, but the antitumour effect of cisplatin was not significantly altered. Conversely, Ch-SeNPs did not impair the Pt-drug uptake or DNA binding in any cell type; and even increased its antitumour effect, which might enable using lower doses of cisplatin without loss of anticancer efficacy, which would result in decreased risk of renotoxicity. Furthermore, cells incubated either with SeMet or SeNPs showed higher levels of selenoproteins, which might enhance cellular defences against the reactive oxygen species (ROS) involved in cisplatin renotoxicity. Hence, both selenocompounds are envisioned as potential coadjuvants to reduce the risk of kidney impairment in future treatments with cisplatin.
Collapse
Affiliation(s)
- Alejandro Iglesias-Jiménez
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| | - Gema Artiaga
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| | - Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| | - M Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain.
| |
Collapse
|
7
|
Pal S, Pragti, Kumar A, Mukhopadhyay S. Exploring the effect of Ru(II) arene complexes on cytotoxicity upon co-ligand variation and loading on amine-functionalized silica nanoparticles. Dalton Trans 2025; 54:7449-7457. [PMID: 40226946 DOI: 10.1039/d4dt03536d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
To overcome the undesirable side effects and acquired resistance associated with platinum-based chemotherapeutics, scientists are searching for alternative strategies involving novel metal-based compounds with improved pharmacological properties. Ruthenium complexes have emerged as prospective candidates to combat side effects and improve the selectivity of anticancer agents. In this work, a benzimidazole-based chelating ligand, HL (4-(1H-naphth[2,3-d]imidazol-2-yl)-1,3-benzenediol) with O and N donor atoms, was synthesized and used for complexation with ruthenium to obtain three Ru(II) arene complexes represented by [Ru(η6-p-cym)(L)Cl], [Ru(η6-p-cym)(L)(PPh3)]+ and [Ru(η6-p-cym)(L)(PTA)]+ (where p-cym = p-cymene, PPh3 = triphenylphosphine and PTA = 1,3,5-triaza-7-phosphaadamantane). The synthesized complexes were characterized using spectroscopic techniques. UV-Vis absorption spectroscopy and LC-MS were used to study the stability of the complexes in biological medium. Their lipophilicity was studied by calculating the partition coefficient in n-octanol and water. The complexes showed significant binding with biomolecules like albumin proteins and nucleic acids. All the complexes were found to be cytotoxic, with complex [Ru(η6-p-cym)(L)PPh3]PF6 exhibiting the highest anticancer activity. The mechanism of anticancer activity was attributed to the ability of the complexes to induce apoptosis and generate reactive oxygen species (ROS). The complexes also exhibited antimetastatic properties. Furthermore, complex [Ru(η6-p-cym)(L)PPh3]PF6 was loaded onto amine-functionalized mesoporous silica nanoparticles which led to an increase in its cytotoxic activity.
Collapse
Affiliation(s)
- Srijita Pal
- Department of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore, Simrol, Khandwa Road, Indore, 453552, India.
| | - Pragti
- Department of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore, Simrol, Khandwa Road, Indore, 453552, India.
| | - Amardeep Kumar
- Department of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore, Simrol, Khandwa Road, Indore, 453552, India.
| | - Suman Mukhopadhyay
- Department of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore, Simrol, Khandwa Road, Indore, 453552, India.
| |
Collapse
|
8
|
Mahran SE, Salem SE, Sabry NA, Farid SF. The nephroprotective effect of metformin with cisplatin in bladder cancer: randomized clinical trial. Int Urol Nephrol 2025:10.1007/s11255-025-04505-2. [PMID: 40319155 DOI: 10.1007/s11255-025-04505-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/31/2025] [Indexed: 05/07/2025]
Abstract
PURPOSE Cisplatin-based combination chemotherapy is the mainstay treatment strategy in various forms of carcinomas and sarcomas. However, its dosage and therapeutic efficacy are significantly limited by its nephrotoxicity. Based on metformin renal benefits in different studies, the study aims to determine safety and the potential nephroprotective effect of metformin when used with cisplatin in patients with bladder cancer. METHODS This was a prospective, randomized, parallel, controlled, open-label study in which 78 chemotherapy naïve bladder cancer patients aged 18-65 years and would receive gemcitabine/cisplatin regimen were selected and randomly assigned to treatment or control group in 1:1 allocation. Both groups were receiving cisplatin standard-of-care regimen, whereas metformin (500 mg, twice daily) was added to the treatment group's regimen only. Patients were prospectively followed up for four cycles of gemcitabine/cisplatin with assessment of renal function tests, serum neutrophil gelatinase-associated lipocalin (NGAL), cystatin-c, and metformin's adverse effects. RESULTS Serum creatinine, serum NGAL, and cystatin-C significantly increased in the control group only (P < 0.001). Estimated glomerular filtration rate (eGFR) significantly declines in the control group only (P < 0.001). On the contrary, serum NGAL significantly improved in the treatment group (P = 0.02) with stable and normal mean value of serum creatinine, eGFR, and cystatin-C without a concomitant significant increase in adverse events, such as hypoglycemia, gastrointestinal symptoms, or weight loss compared to the control group. CONCLUSION Metformin prevented renal damage and deterioration in kidney function in cisplatin-treated patients. Therefore, it is a promising agent in reducing cisplatin-induced nephrotoxicity. The study was registered in ClinicalTrials.gov on December, 16, 2023, Identifier Number NCT06215976.
Collapse
Affiliation(s)
- Samah E Mahran
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, P. O. Box: 11562, Cairo, Egypt.
| | - Salem Eid Salem
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Nirmeen A Sabry
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, P. O. Box: 11562, Cairo, Egypt
| | - Samar F Farid
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, P. O. Box: 11562, Cairo, Egypt
| |
Collapse
|
9
|
Shahrahmani F, Badamchizadeh S, Kaihani F, Alavi-Moghadam S, Keshtkari S, Rezaei-Tavirani M, Arjmand R, Larijani B, Arjmand B. Platinum-based chemotherapies-induced nephrotoxicity: mechanisms, potential treatments, and management. Int Urol Nephrol 2025; 57:1563-1583. [PMID: 39630371 DOI: 10.1007/s11255-024-04303-2] [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: 09/25/2024] [Accepted: 11/20/2024] [Indexed: 04/17/2025]
Abstract
Platinum-based chemotherapies are essential in the treatment of several malignancies. However, such medications can damage the kidneys, frequently leading to both acute and chronic kidney disease. Treatment becomes more difficult for such problems. Physicians may alter chemotherapy regimens and utilize kidney-protecting medications to lessen renal damage. New imaging techniques and biomarkers also aid in the early detection of renal issues. To effectively handle the mentioned situation, oncologists, nephrologists, and pharmacists must collaborate. However, additional study is still required to develop customized therapies, discover strategies to minimize kidney injury and produce new platinum medicines. Hereupon, the present review's authors are being sought to address the causes, prospective treatments, and management of nephrotoxicity caused by platinum-based chemotherapy.
Collapse
Affiliation(s)
- Fatemeh Shahrahmani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sana Badamchizadeh
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Keshtkari
- Department of Internal Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Rasta Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Chen J, Xu M, Wu F, Wu N, Li J, Xie Y, Wang R, Xi N, Zhu Y, Xu X, Liu Y. CRKL silencing inhibits melanoma growth and enhances its chemotherapy sensitivity through the PI3K/AKT and NLRP3/GSDMD pathways. Biochem Pharmacol 2025; 235:116840. [PMID: 40024349 DOI: 10.1016/j.bcp.2025.116840] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Great advances have been made in malignant melanoma treatments, whereas drug resistance still limits many drug applications. CRKL has been reported to be overexpressed in various tumors and showed poor prognosis. However, its specific function and mechanism in melanoma remain unclear. In the present study, we investigated the expression of CRKL and its clinical association by bioinformatics and clinical analysis, and then performed a series of in vitro and in vivo experiments to demonstrate its function and mechanism. Results showed that CRKL increased during melanoma progression and was strongly associated with poor prognosis. CRKL silencing effectively inhibited melanoma cell growth and invasion via ERK/MMP9 and PI3K/AKT signaling pathways both in vitro and in vivo. Moreover, CRKL silencing induced pyroptosis in melanoma cells by upregulating the levels of pyroptosis-associated proteins, such as NLRP3, cleaved Caspase-1, and GSDMD-N. Importantly, our study demonstrated that interfering with CRKL expression enhanced the chemotherapy sensitivity of melanoma cells to cisplatin by regulating PI3K/AKT and NLRP3/GSDMD signaling pathways. In conclusion, our study uncovers a novel molecular mechanism by which CRKL functions in melanoma and highlights potential therapeutic strategies for improving chemotherapy sensitivity in melanoma patients.
Collapse
Affiliation(s)
- Jiashe Chen
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Mingyuan Xu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Fei Wu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Nanhui Wu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Jie Li
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yongyi Xie
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ruoqi Wang
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ningyuan Xi
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yueyi Zhu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Xiaoxiang Xu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China.
| | - Yeqiang Liu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China.
| |
Collapse
|
11
|
Gerges MN, Donia T, Mohamed TM. Indole-3-Carbinol Mechanisms Combating Chemicals and Drug Toxicities. J Biochem Mol Toxicol 2025; 39:e70280. [PMID: 40269607 DOI: 10.1002/jbt.70280] [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: 04/08/2024] [Revised: 08/20/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
The toxicity of chemicals and drugs is a common crisis worldwide. Therefore, the search for protective compounds is growing. Natural compounds such as indole-3-carbinol (I3C) derived from cruciferous vegetables are preferred since they are safe for humans and the environment. This review focuses on I3C potential role in preventing and repairing damage caused by chemicals and drugs. Interestingly, I3C ameliorates hepatotoxicity induced by carbon tetrachloride (CCl4), diethylnitrosamine (DENA), alcohol, gold nanoparticles, and microbial toxins. Additionally, it inhibits carcinogenesis induced by different chemicals and prevents the deleterious effects of different antineoplastic drugs including cisplatin, doxorubicin (DOX), and trabectidin on normal tissues. Moreover, it reduces fetal malformation and protects against micronuclei formation and calstogenecity induced by cyclophosphamide (CP) in bone marrow cells. It also attenuates methotrexate (MTX)-induced hepatotoxicity, mitigates neurotoxicity caused by thioacetamide and clonidine, and protects against aspirin side effects in gastric mucosa. Furthermore, its nanoparticles inhibit neuronal damage caused by glutamate and rotenone. Thus, I3C prevents the toxicities caused by chemicals in the surrounding environment as well as those of consumed drugs.
Collapse
Affiliation(s)
- Marian N Gerges
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Thoria Donia
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Tarek M Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
12
|
Riediger T, Stammler A, Oldengott J, Walleck S, Glaser T. Support for the Anticipated Binding Mode of a Cytotoxic Dinuclear Copper Complex to Two Neighboring Phosphate Esters of the DNA Backbone. Inorg Chem 2025. [PMID: 40298861 DOI: 10.1021/acs.inorgchem.5c00505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The cytotoxic dinuclear complex [(Htom6-Me){CuII(OAc)}2](OAc) (H2tom6-Me = 2,7-bis(di(6-methylpyridine-2-yl-methyl)aminomethyl)-1,8-naphthalenediol) binds to DNA, interferes with DNA synthesis in PCR at lower concentrations than cisplatin, and kills human cancer cells more efficiently than human stem cells of the same proliferation rate. It is supposed that the OAc- ligands dissociate in aqueous buffer, providing [(Htom6-Me){CuII(OH2)}2]3+, while the fragment [(Htom6-Me)CuII2]3+ binds to two neighboring phosphate diesters of the DNA backbone. Here, we report the hydrolysis of [(Htom6-Me){CuII(OAc)}2](OAc) in phosphate buffer providing the anticipated [(Htom6-Me){CuII(OH2)}2]3+. Moreover, the same reaction in H2O/CH3OH yields the complex [(Htom6-Me){CuII(H2PO4)}2]+. The molecular structures of [(Htom6-Me){CuII(OH2)}2]3+ and [(Htom6-Me){CuII(H2PO4)}2]+ show different coordination modes around the CuII ions demonstrating a significant flexibility of the fragment [(Htom6-Me)CuII2]3+ for binding to exogenous ligands despite its rigid naphthalene backbone. The coupling is weakly antiferromagnetic in [(Htom6-Me){CuII(H2PO4)}2]+, while that in tricationic [(Htom6-Me){CuII(OH2)}2]3+ is weakly ferromagnetic and can be attributed to the orthogonal orientation of the basal planes. UV-vis-NIR spectra of [(Htom6-Me){CuII(H2PO4)}2]+ but also of [(Htom6-Me){CuII(OAc)}2](OAc) in aqueous solution resemble that of [(Htom6-Me){CuII(OH2)}2]3+ demonstrating our earlier assumption that [(Htom6-Me){CuII(OH2)}2]3+ is the active species in buffer solutions for interaction with DNA. Moreover, the exchange of the OAc- ligands by H2PO4- ligands models the anticipated binding of [(Htom6-Me){CuII(OH2)}2]3+ to the phosphates of DNA.
Collapse
Affiliation(s)
- Thomas Riediger
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| | - Anja Stammler
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| | - Jan Oldengott
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| | - Stephan Walleck
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| | - Thorsten Glaser
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| |
Collapse
|
13
|
Mukherjee A, Manna S, Singh A, Ray A, Srivastava S. Investigating Cisplatin Resistance in Squamous Cervical Cancer: Proteomic Insights into DNA Repair Pathways and Omics-Based Drug Repurposing. J Proteome Res 2025. [PMID: 40298920 DOI: 10.1021/acs.jproteome.4c00885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Cisplatin-based chemotherapy is a cornerstone in treating cervical cancer, yet the efficacy is frequently limited by the rapid onset of drug resistance, a major challenge in clinical management. To investigate this, we employed HPV16+ human cervix squamous carcinoma cells, SiHa (CIS/S), and their cisplatin-resistant subline (CIS/R) as a model. Using DIA-based proteomics, we identified 5152 protein groups and over 50,000 peptides with a global FDR <1%. Comparative analysis revealed 123 differentially expressed proteins. Gene Set Enrichment Analysis (GSEA) highlighted proteins involved in DNA damage, metabolism, and repair pathways (RFC4, RFC3, RFC2, DUT, DDX54, CDCA8, CDK7, CHAF1B, and GTF2F1), suggesting a role in developing acquired cisplatin resistance. Pathways related to mitotic spindle assembly and P53 signaling were found to be perturbed in resistant cells. Next, we screened a library of approx. 240 FDA-approved drugs against three protein targets and found four small-molecular ligands as potential hits for further in vitro validation. Cabozantinib and sorafenib gave us positive results in terms of increasing the cisplatin sensitivity of CIS/R cells. In conclusion, our findings provide insights into the molecular mechanisms underpinning cisplatin resistance in cervical cancer and propose novel strategies for combating this resistance through targeted therapies and drug repurposing.
Collapse
Affiliation(s)
- Amrita Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sayan Manna
- Department of Biotechnology, Haldia Institute of Technology, Purba Medinipur, Haldia 721607, West Bengal, India
| | - Avinash Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Adrija Ray
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
14
|
Coelho MP, Farinha PF, Côrte-Real L, Ribeiro N, Luiz H, Pinho JO, Noiva R, Godinho-Santos C, Reis CP, Correia I, Gaspar MM. Liposomal nanoformulations of novel copper-based complexes exhibiting antimelanoma activity - In vitro and in vivo validation. Int J Pharm 2025; 677:125643. [PMID: 40294769 DOI: 10.1016/j.ijpharm.2025.125643] [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: 03/08/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Melanoma stands as the most aggressive form of skin cancer. The lack of effective and safe therapies has led to the investigation of innovative strategies. The present work validates the in vitro and in vivo antimelanoma activity of new copper complexes of 8-hydroxyquinoline (8HQ) derivatives in free or liposomal forms. Firstly, the cytotoxic properties of several copper-based complexes were screened towards human (A375) and murine (B16F10) melanoma cell lines and human dermal fibroblasts or keratinocytes (HaCaT) cell lines. All the complexes presented lower IC50 values (<20 μM) than dacarbazine (DTIC) and temozolomide (TMZ), the positive controls (>80 μM). Aiming to solve low specificity against tumor cells and enhance its targetability to affected sites three metal-based complexes were selected, based on their antiproliferative properties, and incorporated in long blood circulating liposomes. One of them, di-2-(((2-morpholinoethyl)imino)methyl)quinolin-8-olCopper(II), designated as LCR35, was selected for further studies due to the highest incorporation parameters and cytotoxic properties observed. The antiproliferative activity of LCR35 was preserved after its association to liposomes. Moreover, in B16F10 cells this effect was potentiated. Furthermore, cell cycle analysis studies in A375 and B16F10 cell lines were performed to elucidate the mechanism of action of copper-based complex formulations. A cell cycle arrest at G2/M and G0/G1 phases in A375 and B16F10 cells, respectively, both in free and liposomal forms were observed. To validate the therapeutic potential of LCR35 two murine melanoma models were carried out: subcutaneous and metastatic. Pre-clinical studies demonstrated the high therapeutic effect of LCR35, especially after incorporation in liposomes, compared to control group or animals that received LCR35 Free and DTIC. Overall, in vitro and in vivo studies highlight the potential antimelanoma properties of the copper-based complex, LCR35.
Collapse
Affiliation(s)
- Mariana P Coelho
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa 1649 - 003 Lisboa, Portugal; i3N/CENIMAT, Department of Materials Science, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Pedro F Farinha
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa 1649 - 003 Lisboa, Portugal
| | - Leonor Côrte-Real
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa 1049-001 Lisboa, Portugal
| | - Nádia Ribeiro
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa 1049-001 Lisboa, Portugal
| | - Hugo Luiz
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa 1649 - 003 Lisboa, Portugal
| | - Jacinta O Pinho
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa 1649 - 003 Lisboa, Portugal
| | - Rute Noiva
- CIISA - Interdisciplinary Centre of Research in Animal Health, Faculdade de Medicina Veterinaria, Universidade de Lisboa 1300 - 477 Lisboa, Portugal
| | - Catarina Godinho-Santos
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa 1649 - 003 Lisboa, Portugal
| | - Catarina Pinto Reis
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa 1649 - 003 Lisboa, Portugal; IBEB - Institute of Biophysics and Biomedical Engineering, Faculty of Sciences, Universidade de Lisboa 1649-016 Lisboa, Portugal
| | - Isabel Correia
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa 1049-001 Lisboa, Portugal.
| | - Maria Manuela Gaspar
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa 1649 - 003 Lisboa, Portugal; IBEB - Institute of Biophysics and Biomedical Engineering, Faculty of Sciences, Universidade de Lisboa 1649-016 Lisboa, Portugal.
| |
Collapse
|
15
|
Zafar A, Khatoon S, Khan MJ, Abu J, Naeem A. Advancements and limitations in traditional anti-cancer therapies: a comprehensive review of surgery, chemotherapy, radiation therapy, and hormonal therapy. Discov Oncol 2025; 16:607. [PMID: 40272602 PMCID: PMC12021777 DOI: 10.1007/s12672-025-02198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 03/20/2025] [Indexed: 04/25/2025] Open
Abstract
Cancer remains a major global health challenge, consistently ranking as the second leading cause of mortality worldwide. Despite significant advancements in research and technology, the need to deepen our understanding of tumor biology and improve therapeutic strategies persists. This review focuses on the progress and challenges of four traditional cancer treatment modalities: surgery, chemotherapy, radiation therapy, and hormonal therapy. Surgery, the primary method for tumor removal, has evolved with the integration of fluorescence-based technology and robotic systems, enhancing precision and minimizing collateral damage. Radiation therapy has progressed with improved focus, intensity control, and 3D technology, refining both diagnosis and treatment. Chemotherapy has advanced from natural extracts to synthesized derivatives with amplified cytotoxicity against cancer cells. Hormonal therapy has emerged as a crucial strategy for hormone-dependent cancers, restraining growth or inducing regression. Despite these advancements, each approach faces ongoing challenges. Surgery struggles with complete tumor removal due to heterogeneity. Chemotherapy contends with drug resistance and side effects. Radiation therapy grapples with precision issues and limited access in some regions. Hormonal therapy faces resistance development and quality of life impacts. This study provides a comprehensive analysis of the evolution of these traditional anti-cancer therapies, offering insights into their progress and highlighting areas for future research. By examining these modalities, we aim to underscore their relevance in the current oncology landscape and identify opportunities for improvement in cancer treatment strategies.
Collapse
Affiliation(s)
- Aasma Zafar
- Department of Biosciences, COMSATS University, Islamabad, 45550, Pakistan
| | - Summaiya Khatoon
- College of Health Sciences, QU Health Sector, Qatar University, P.O. Box 2713, Doha, Qatar
| | | | - Junaid Abu
- Aisha bint Hamad al-Attiyah hospital, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Aisha Naeem
- Research and Graduate Studies, QU Health Sector, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
16
|
Anjomshoa M, Amirheidari B, Sahihi M, Janczak J, Forootanfar H, Farsinejad A, Abolhassani Y, Karami-Mohajeri S. In vitro cellular and molecular plus in silico studies of a substituted bipyridine-coordinated Zn(II) ion: cytotoxicity, ROS-induced apoptosis, anti-metastasis, and BAX/BCL2 genes expression. J Biol Inorg Chem 2025:10.1007/s00775-025-02114-z. [PMID: 40253669 DOI: 10.1007/s00775-025-02114-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025]
Abstract
A new dimethyl-substituted bipyridine-Zn(II) complex (2Mebpy-Zn) was synthesized and structurally characterized. Single-crystalline structure of the complex was elucidated as [Zn(2Mebpy)3](ClO4)2∙1.5(dioxane) by X-ray diffraction, where 2Mebpy is 4,4'-dimethyl-2,2'-bipyridine. The three-dimensional electrostatic potential maps (3D ESP) were plotted for [Zn(2Mebpy)3]2+ cation and [Zn(2Mebpy)3](ClO4)2 molecule. In vitro cytotoxicity studies indicated significant cytotoxicity of 2Mebpy-Zn against both breast (MCF-7) and glioblastoma (U-87) cancer cells relative to normal murine embryo cells (NIH/3T3). The results are indicative of a superior selectivity toward MCF-7 over the other cell lines as confirmed by IC50 value of 5.1 ± 0.5 µM after 48 h. Interestingly, MCF-7 and U-87 cells death induced by 2Mebpy-Zn mostly proceed through an apoptotic pathway which probably associates with the overproduction of reactive oxygen species (ROS). The Zn(II) complex suppressed the metastatic affinity of MCF-7 cells by blocking migration as well as formation of colonies. Also, the expression of two opponent apoptosis-relevant genes (BAX and BCL2) measured by real-time polymerase chain reaction (qPCR) experiments indicated that 2Mebpy-Zn could potentially trigger apoptotic cell death. Moreover, 2Mebpy-Zn could cleave hydrolytically the pUC19 DNA without the need to add any external agent. Finally, the binding affinity of two enantiomers of 2Mebpy-Zn toward cancer therapeutic targets, such as anti-apoptotic proteins, estrogen receptor α, tubulin, and topoisomerase II, was studied by in silico molecular docking. In conclusion, 2Mebpy-Zn can be introduced as a potential therapeutic agent in breast cancer and indicates that other metal complexes with bipyridine derivatives can also exhibit promising anticancer effects.
Collapse
Affiliation(s)
- Marzieh Anjomshoa
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical sciences, Kerman, Iran.
| | - Bagher Amirheidari
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical sciences, Kerman, Iran.
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehdi Sahihi
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut de Chimie de Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Jan Janczak
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okólna 2 Str., 50-422, Wrocław, Poland
| | - Hamid Forootanfar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical sciences, Kerman, Iran
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Farsinejad
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasaman Abolhassani
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical sciences, Kerman, Iran
| | - Somayyeh Karami-Mohajeri
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
17
|
Iova V, Tincu RC, Scrobota I, Tudosie MS. Pt(IV) Complexes as Anticancer Drugs and Their Relationship with Oxidative Stress. Biomedicines 2025; 13:981. [PMID: 40299672 PMCID: PMC12024748 DOI: 10.3390/biomedicines13040981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
Despite continuous research, cancer is still a leading cause of death worldwide; therefore, new methods of cancer management improvement are emerging. It is well known that in the pathophysiology of cancer, oxidative stress (OS) is a significant factor. Nevertheless, there is currently no quick or easy way to identify OS in cancer patients using blood tests. Currently, in cancer treatments, Pt(IV) complexes are preferred to Pt(II) complexes in terms of adverse effects, drug resistance, and administration methods. Intracellular reductants convert Pt(IV) complexes to their Pt(II) analogs, which are Pt compounds with anti-carcinogenic effects. Our aim was to find out if Pt(IV) complexes could be used to assess blood oxidative stress indicators and, consequently, monitor the development of cancer. In this review, we analyzed previous research using the PubMed and Google Scholar public databases to verify the potential use of Pt(IV) complexes in cancer management. We found that two main serum antioxidants, glutathione and ascorbic acid, which are easily measured using conventional methods, react favorably with Pt(IV) complexes. Our research results suggest Pt(IV) complexes as therapeutic anticancer drugs and potential diagnosis agents. However, further research must be conducted to verify this hypothesis.
Collapse
Affiliation(s)
- Vlad Iova
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania; (V.I.); (R.C.T.); (M.S.T.)
| | - Radu Ciprian Tincu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania; (V.I.); (R.C.T.); (M.S.T.)
- ICU II Toxicology, Clinical Emergency Hospital, 014461 Bucharest, Romania
| | - Ioana Scrobota
- Department of Dental Medicine, Faculty of Medicine and Pharmacy, University of Oradea, 1st Decembrie Street, 410073 Oradea, Romania
| | - Mihail Silviu Tudosie
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania; (V.I.); (R.C.T.); (M.S.T.)
- ICU II Toxicology, Clinical Emergency Hospital, 014461 Bucharest, Romania
| |
Collapse
|
18
|
Xu Z, Zhang T, Xu Z, Ma Y, Niu Z, Chen J, Zhang M, Shi F. Research Progress and Prospects of Nanozymes in Alleviating Abiotic Stress of Crops. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8694-8714. [PMID: 39936319 DOI: 10.1021/acs.jafc.4c10799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
The continuous destruction of the global ecological environment has led to increased natural disasters and adverse weather, severely affecting crop yields and quality, particularly due to abiotic stress. Nanase, a novel artificial enzyme, simulates various enzyme activities, is renewable, and shows significant potential in promoting crop growth and mitigating abiotic stress. This study reviews the classification of nanoenzymes into carbon-based, metal-based, metal oxide-based, and others based on synthesis materials. The catalytic mechanisms of these nanoenzymes are discussed, encompassing activities, such as oxidases, peroxidases, catalases, and superoxide dismutases. The catalytic mechanisms of nanoenzymes in alleviating salt, drought, high-temperature, low-temperature, heavy metal, and other abiotic stresses in crops are also highlighted. Furthermore, the challenges faced by nanoenzymes are discussed, especially in sustainable agricultural development. This review provides insights into applying nanoenzymes in sustainable agriculture and offers theoretical guidance for mitigating abiotic stress in crops.
Collapse
Affiliation(s)
- Zhenghong Xu
- College of Life Sciences, Shihezi University, Shihezi 832003, China
| | - Tongtong Zhang
- College of Life Sciences, Shihezi University, Shihezi 832003, China
| | - Zhihua Xu
- College of Life Sciences, Shihezi University, Shihezi 832003, China
| | - Yu Ma
- College of Life Sciences, Shihezi University, Shihezi 832003, China
| | - Zhihan Niu
- College of Life Sciences, Shihezi University, Shihezi 832003, China
| | - Jiaqi Chen
- College of Life Sciences, Shihezi University, Shihezi 832003, China
| | - Min Zhang
- College of Life Sciences, Shihezi University, Shihezi 832003, China
| | - Feng Shi
- College of Life Sciences, Shihezi University, Shihezi 832003, China
| |
Collapse
|
19
|
Xie WH, Xiao WW, Chang H, Xu MJ, Hu YH, Zhou TC, Zhong Q, Chen CY, Lu LX, Wang QX, Zhu YJ, Yang J, Shi XY, Kang HL, Wei JW, Huang R, Peng HH, Yuan Y, Wu SH, Jiang XH, Liu YJ, Wen BX, Gao YH. Four cycles of docetaxel plus cisplatin as neoadjuvant chemotherapy followed by concurrent chemoradiotherapy in stage N2-3 nasopharyngeal carcinoma: phase 3 multicentre randomised controlled trial. BMJ 2025; 389:e081557. [PMID: 40233976 PMCID: PMC11997745 DOI: 10.1136/bmj-2024-081557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/17/2025]
Abstract
OBJECTIVE To compare the effects of four cycles of docetaxel with cisplatin as a neoadjuvant chemotherapy followed by concurrent chemoradiotherapy with concurrent chemoradiotherapy alone by assessing reductions in distant metastasis and improvements in survival in patients with stage N2-3nasopharyngeal carcinoma. DESIGN Phase 3, multicentre, randomised controlled trial. SETTING Six sites in China from 23 February 2016 to 18 February 2019. PARTICIPANTS 186 participants aged ≤70 years with a diagnosis of untreated stage T1-4N2-3M0 nasopharyngeal carcinoma. INTERVENTION Participants were prospectively enrolled and randomly allocated to either the neoadjuvant chemotherapy plus concurrent chemoradiotherapy group (four cycles of neoadjuvant chemotherapy (docetaxel 75 mg/m2 on day 1 and cisplatin 37.5 mg/m2 on days 2-3, every 3 weeks) followed by concurrent chemoradiotherapy (intensity modulated radiotherapy plus weekly cisplatin 40 mg/m2) or the concurrent chemoradiotherapy only group, in a 1:1 ratio. MAIN OUTCOME MEASURES Five year distant metastasis-free survival and overall survival were analysed using the intention-to-treat approach. RESULTS 93 participants were assigned to each of the neoadjuvant chemotherapy plus concurrent chemoradiotherapy and concurrent chemoradiotherapy only groups. After a median follow-up time of 76.9 (interquartile range 65.4-85.9) months, the neoadjuvant chemotherapy plus concurrent chemoradiotherapy group had superior five year distant metastasis-free survival (91.3% (95% confidence interval (CI) 85.4% to 97.2%) versus 78.2% (69.8% to 86.6%); hazard ratio 0.41 (95% CI 0.19 to 0.87); P=0.02) and five year overall survival (90.3% (84.2% to 96.4%) versus 82.6% (75.0% to 90.2%); hazard ratio 0.38 (0.18 to 0.82); P=0.01). Grade 3/4 acute toxicities were observed in 60 (65%) and 46 (51%) patients in the neoadjuvant chemotherapy plus concurrent chemoradiotherapy and concurrent chemoradiotherapy only groups, respectively (P=0.05). The higher acute toxicity observed in the neoadjuvant chemotherapy plus concurrent chemoradiotherapy group was primarily due to grade 3/4 neutropenia (43 (47%) v 10 (11%); P<0.001). No significant difference in any late toxicity was observed between the two groups, and participants in the neoadjuvant chemotherapy plus concurrent chemoradiotherapy group tended to have a better quality of life five years after enrolment. CONCLUSIONS Four cycles of docetaxel plus cisplatin neoadjuvant chemotherapy with concurrent chemoradiotherapy can effectively reduce distant metastasis and improve survival for patients with stage N2-3 nasopharyngeal carcinoma with manageable toxicities. TRIAL REGISTRATION ClinicalTrials.gov NCT02512315.
Collapse
Affiliation(s)
- Wei-Hao Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Wei-Wei Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Hui Chang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Ming-Jun Xu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yong-Hong Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Tong-Chong Zhou
- Guangzhou Institute of Cancer Research, Guangzhou, China
- Department of Radiation Oncology, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiong Zhong
- Department of Oncology, Ganzhou People's Hospital, Ganzhou, China
| | - Chun-Yan Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Li-Xia Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Qiao-Xuan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Yu-Jia Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Jing Yang
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xing-Yuan Shi
- Guangzhou Institute of Cancer Research, Guangzhou, China
- Department of Radiation Oncology, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Radiation Oncology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hua-Long Kang
- Department of Oncology, Ganzhou People's Hospital, Ganzhou, China
| | - Jia-Wang Wei
- Department of Oncology, Ganzhou People's Hospital, Ganzhou, China
| | - Rong Huang
- Department of Oncology, Ganzhou People's Hospital, Ganzhou, China
| | - Hai-Hua Peng
- Guangzhou Institute of Cancer Research, Guangzhou, China
- Department of Radiation Oncology, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yan Yuan
- Guangzhou Institute of Cancer Research, Guangzhou, China
- Department of Internal Medicine, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shi-Hai Wu
- Department of Radiation Oncology, Second Clinical Medicine College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Xin-Hua Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiography, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| | - Ya-Jie Liu
- Department of Radiation Oncology, Second Clinical Medicine College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
- Department of Radiation Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bi-Xiu Wen
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuan-Hong Gao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Centre, Guangzhou, China
| |
Collapse
|
20
|
Xu E, Yu Z, Zheng J. Lycopene improves cisplatin induced hepatointestinal injury in rats by modulating the microbe-gut-liver axis. Food Funct 2025; 16:3064-3074. [PMID: 40143795 DOI: 10.1039/d4fo06059h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Cisplatin (CIS) is a commonly used antitumor drug in clinics, but its application is limited due to hepatotoxicity, nephrotoxicity and gastrointestinal toxicity. In recent years, a large number of studies have shown that the imbalance of intestinal flora is one of the important factors in the malignant development of diseases. Therefore, improving organ function by regulating intestinal flora may be an important strategy to prevent the side effects of chemotherapy drugs. Lycopene (LYC) is found in a wide range of red foods and has antioxidant, anti-inflammatory and immune-enhancing effects. So the purpose of this study was to explore its effect on hepatointestinal injury caused by chemotherapy drugs. The results of this study showed that CIS could significantly restore body weight, diet, water intake, and AST, ALT and other physiological and biochemical indexes of rats. HE staining, projective electron microscopy and TUNEL results showed that LYC alleviated morphological and ultrastructural damage of the liver and intestine. Then, ELISA results showed that LYC can reduce cell apoptosis by increasing the antioxidant capacity and reducing inflammatory response. Secondly, 16sRNA and metabolome results showed that LYC enriched beneficial bacteria (Firmicutes and Proteobacteria), reduced harmful bacteria (E. coli, etc.), enhanced metabolic pathway changes such as taurine and hypotaurine metabolism, and alleviated organ damage caused by CIS. Finally, network pharmacology, molecular docking and immunohistochemistry showed that LYC could reduce CIS induced hepatocyte inflammation and apoptosis by activating the PI3K/AKT pathway. In summary, LYC alleviates the toxic side effects of chemotherapy drugs by regulating the PI3K/AKT pathway and the intestinal microbiota-metabolite-liver axis.
Collapse
Affiliation(s)
- Enshuang Xu
- Department of Veterinary Surgery, College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163000, China.
| | - Zhiying Yu
- Department of Veterinary Surgery, College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163000, China.
| | - Jiasan Zheng
- Department of Veterinary Surgery, College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163000, China.
| |
Collapse
|
21
|
Liu H, Zou J, Li X, Ge Y, He W. Drug delivery for platinum therapeutics. J Control Release 2025; 380:503-523. [PMID: 39923853 DOI: 10.1016/j.jconrel.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 01/18/2025] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
Cancer remains a severe threat to human health. Platinum drugs, such as cisplatin (CDDP), oxaliplatin, and carboplatin, are extensively utilized for treating various cancers and have become the primary drugs in first-line treatments for numerous solid tumors due to their effective anticancer properties. However, their side effects, including drug resistance, nephrotoxicity and ototoxicity, limit the clinical application. Therefore, there is an urgent need to develop targeted delivery and controlled release systems for platinum drugs to address the disadvantages, enhancing tumor accumulation and improving therapeutic effects. In this review, we first review the progress of platinum drugs, their anticancer mechanism, clinical applications and limitations. Then, we comprehensively summarize the platinum-based delivery using drug carriers and responsive strategies. We especially highlight the platinum-delivery formulations in ongoing clinical trials. Finally, we provide perspectives for this field. The review could provide an increasingly in-depth understanding of platinum therapeutics and motivate increasing delivery tactics to overcome the limitations of platinum application.
Collapse
Affiliation(s)
- Hui Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Yizhi Ge
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, PR China.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, PR China.
| |
Collapse
|
22
|
Munir N, Gürbüz N, Chaudhry GES, Özdemir İ, Sarfraz M, Şen B, Aygün M, Zafar MN. Synergistic Antiproliferative Activity of Newly Synthesized Benzimidazole-Based Silver(I) Complexes on MCF-7 and T47D Cell Lines, CT-DNA Interactions Supported by Computational Studies. ACS OMEGA 2025; 10:13278-13295. [PMID: 40224434 PMCID: PMC11983176 DOI: 10.1021/acsomega.4c11048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/15/2025]
Abstract
This article reports the synthesis, characterization, and antitumor properties of newly synthesized benzimidazole-based Ag(I)-(BNHCs) complexes from their proligands. All of the compounds underwent comprehensive characterization using techniques such as 1H, COSY, 13C NMR, IR spectroscopy, electrospray ionization (ESI)-mass, elemental, and single-crystal X-ray diffraction (XRD) analysis. Density functional theory (DFT) studies were carried out to observe the electronic effects of bound ligands to modulate the selectivity and reactivity of silver complexes. Time-dependent DFT (TD-DFT) studies assessed the optical properties of synthesized complexes and were further highlighted by orbital contributions with oscillator strengths. All compounds were tested against breast cancer MCF-7 and T47D cell lines. The synergistic effects of benzimidazole-incorporated aryl constituent structuring silver complexes were also observed. Nearly all silver complexes have been found to be promising anticancer agents with the added benefit of low cytotoxic effects toward normal cells. Intriguingly, [AgL 4 (Cl)] exhibited the best cytotoxic activity among our screened complexes as IC50 values for both MCF-7 and T47D were 9 ± 1.04 and 11 ± 1.41, respectively. The apoptosis mode of cell death was confirmed by phosphatidylserine exposure and annexin V/PI staining imaging method. CT-DNA interactions of the most active silver complex ([AgL 4 (Cl)]) and its proligand (HL 4 (Cl)) were carried out to support the mode of compound-DNA interaction. Strong DNA binding affinities (K b) with compounds through electrostatic and intercalation modes induced structural changes in DNA. Moreover, molecular docking studies were carried out to comprehend the possible interactions of compounds with various receptors such as EGFR (epidermal growth factor receptor), VEGFR2 (vascular endothelial growth factor receptors), FGFR (fibroblast growth factor receptor), and SRC (proto-oncogene tyrosine kinase protein) of tyrosine kinase family serves as crucial receptors in breast cancer.
Collapse
Affiliation(s)
- Naima Munir
- Department
of Chemistry, Quaid-I-Azam University, 45320 Islamabad, Pakistan
- Catalysis
Research and Application Centre, Inonü
University, 44210 Malatya, Turkiye
| | - Navin Gürbüz
- Catalysis
Research and Application Centre, Inonü
University, 44210 Malatya, Turkiye
- Faculty
of Science and Art, Department of Chemistry, İnönü University, 44210 Malatya, Turkiye
| | - Gul-e-Saba Chaudhry
- Institute
of Climate Adaptation and Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Terengganu, Malaysia
| | - İsmail Özdemir
- Catalysis
Research and Application Centre, Inonü
University, 44210 Malatya, Turkiye
- Faculty
of Science and Art, Department of Chemistry, İnönü University, 44210 Malatya, Turkiye
| | - Muhammad Sarfraz
- Institute
of chemistry, Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan
| | - Betül Şen
- Department
of Physics, Faculty of Arts and Sciences, Dokuz Eylül University, 35150 Izmir, Turkiye
| | - Muhittin Aygün
- Department
of Physics, Faculty of Arts and Sciences, Dokuz Eylül University, 35150 Izmir, Turkiye
| | | |
Collapse
|
23
|
Bai D, Nowak M, Lu D, Wang Q, Fitzgerald M, Zhang H, MacDonald R, Xu Z, Luo L. The outcast of medicine: metals in medicine--from traditional mineral medicine to metallodrugs. Front Pharmacol 2025; 16:1542560. [PMID: 40260378 PMCID: PMC12010122 DOI: 10.3389/fphar.2025.1542560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/07/2025] [Indexed: 04/23/2025] Open
Abstract
Metals have long held a significant role in the human body and have been utilized as mineral medicines for thousands of years. The modern advancement of metals in pharmacology, particularly as metallodrugs, has become crucial in disease treatment. As the machanism of metallodurgsare increasingly uncovered, some metallodrugs are already approved by FDA and widely used in treating antitumor, antidiabetes, and antibacterial. Therefore, a thorough understanding of metallodrug development is essential for advancing future study. This review offers an in-depth examination of the evolution of mineral medicines and the applications of metallodrugs within contemporary medicine. We specifically aim to summarize the historical trajectory of metals and mineral medicines in Traditional Chinese Mineral Medicine by analyzing key historical texts and representative mineral medicines. Additionally, we discuss recent advancements in understanding metallodrugs' mechanisms, such as protein interactions, enzyme inhibition, DNA interactions, reactive oxygen species (ROS) generation, and cellular structure targeting. Furthermore, we address the challenges in metallodrug development and propose potential solutions. Lastly, we outline future directions for metallodrugs to enhance their efficacy and effectiveness. The progression of metallodrugs has broadened their applications and contributed significantly to patient health, creating good healthcare solutions for the global population.
Collapse
Affiliation(s)
- Donghan Bai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Michal Nowak
- Faculty of Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Dajun Lu
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Qiaochu Wang
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC, United States
| | | | - Hui Zhang
- Institute of Traditional Chinese Medicine, European University of Chinese Medicine, Horsens, Denmark
| | - Remy MacDonald
- Department of Statistics, George Mason University, Virginia, VA, United States
| | - Ziwen Xu
- Department of Nursing, The University of Melbourne, Parkville, VIC, Australia
| | - Lu Luo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Wang Y, Lv H, Shen L, Chen Z. Miltirone enhances the chemosensitivity of gastric cancer cells to cisplatin by suppressing the PI3K/AKT signaling pathway. Front Pharmacol 2025; 16:1553791. [PMID: 40260390 PMCID: PMC12009761 DOI: 10.3389/fphar.2025.1553791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025] Open
Abstract
Background Gastric cancer (GC) is one of the most common malignant tumors with poor survival. Although cisplatin is a first-line chemotherapy drug for GC, it still has the potential to develop drug resistance and side effects. Miltirone, extracted from Chinese herb Salvia miltiorrhiza Bunge, has been reported to significantly inhibit some types of cancer. However, its effects on GC have not been studied, the possible anti-tumor effects of miltirone in combination with cisplatin in GC patients have not been explored. Materials and methods Human GC cell lines AGS, HGC27, MKN45 and MGC803 cells were treated with miltirone and cisplatin individually or combinatorially. Cell proliferation assay, flow cytometric assay, colony formation assay and Western blot were employed to evaluate the cytotoxic effects under these treatments. Wound healing and transwell assays were used to examine the effects of miltirone and/or cisplatin on GC cell migration and invasion. RNA-seq analysis was used to determine miltirone's potential target genes in AGS cells. GO analysis and molecular docking assay were used to determine the pathways affected by miltirone. Next, we examined changes in the selected pathway proteins. The in vivo animal model was verified the results of the in vitro experiments. Results Miltirone inhibited cell growth, migration, and invasion, as well as induced apoptosis in GC cells. In combinatorial treatments, miltirone synergistically enhanced cytotoxicity of cisplatin in GC cells. Moreover, the expression levels of 606 genes appeared to be significantly modulated by miltirone via RNA-seq analyses, and PI3K/AKT signaling pathway was found to refer to miltirone activity. Furthermore, miltirone together with cisplatin treatment significantly reduced the expression levels of p-PI3K, p-Akt, p-mTOR, while the total levels of PI3K and Akt remained unchanged. In addition, compared with the control group, the tumors growth was significantly suppressed in groups treated with the two agents alone or in combination, and even more so in the combination group in vivo. Discussion Miltirone inhibited the proliferation of GC cells and significantly potentiates the anticancer activities of cisplatin by downregulating the PI3K/AKT signaling pathway. Combination therapy of miltirone and cisplatin represents a novel potential treatment of gastric cancer.
Collapse
Affiliation(s)
- Yiping Wang
- Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hang Lv
- Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Shen
- Institute of Basic Theory of TCM, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhe Chen
- Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
25
|
Nair M, Samidurai A, Das A, Kakar SS, Kukreja RC. Ovarian cancer and the heart: pathophysiology, chemotherapy-induced cardiotoxicity, and new therapeutic strategies. J Ovarian Res 2025; 18:72. [PMID: 40188339 PMCID: PMC11971845 DOI: 10.1186/s13048-025-01636-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/21/2025] [Indexed: 04/07/2025] Open
Abstract
Ovarian Cancer (OC) is recognized as the most lethal gynecologic malignancy, characterized by numerous genetic mutations that trigger uncontrolled cellular growth and replication. Emerging evidence suggests that non-coding RNAs including miRNAs and lncRNAs significantly influence OC through their multiple roles including tumor initiation, progression, metastasis, immune evasion, and chemoresistance, making them promising diagnostic markers and therapeutic targets. The primary approach to treating OC typically involves cytoreductive surgery followed by chemotherapy. However, the chemotherapeutic agents, particularly the anthracyclines such as doxorubicin (DOX), are known for their cardiotoxic effects, which can range from acute to chronic, potentially leading to heart failure and death. To enhance the overall treatment response and to minimize cardiotoxicity, alternative strategies have been explored. These include the use of liposomal doxorubicin (DOXIL) as a substitute for DOX, various radiotherapies, immunotherapies, and the co-administration of angiotensin-converting enzyme inhibitors and/or beta-blockers. Phosphodiesterase-5 inhibitors (PDE5i) have also demonstrated efficacy in reducing cardiotoxicity linked to cancer treatments and in promoting apoptosis in cancer cells across multiple cancer types. Although there is no current clinical trial directly examining the impact of PDE5i on reducing cardiotoxicity in OC, however emerging therapies such as Withaferin A, PARP inhibitors, and nanoparticle combination therapy show promise. Additional research is essential to develop treatments that are both effective against OC and less harmful to the heart.
Collapse
Affiliation(s)
- Megha Nair
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Arun Samidurai
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Anindita Das
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Sham S Kakar
- Department of Physiology, University of Louisville, Louisville, KY, USA
| | - Rakesh C Kukreja
- Department of Internal Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
| |
Collapse
|
26
|
Mihajlović E, Biancalana L, Mojić M, Jelača S, Chiaverini L, Zacchini S, Mijatović S, Maksimović-Ivanić D, Marchetti F. Anticancer activity promoted by ligand diversity in diiron thiocarbyne complexes. Eur J Med Chem 2025; 287:117364. [PMID: 39923532 DOI: 10.1016/j.ejmech.2025.117364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Mononuclear iron (II) complexes have been intensively investigated with the aim of developing efficacious anticancer drugs that can overcome the serious limitations associated with the platinum complexes currently employed in chemotherapy. Combining a promising antitumor potential with appropriate physicochemical properties, such as aqueous stability and a balanced hydrophilic/lipophilic character, is essential for clinical progression. We prepared six highly functionalized diiron(I) complexes from the μ-thiocarbyne precursor [Fe2Cp2(CO)2(μ-CO)(μ-CSMe)]CF3SO3, 1 (Cp = η5-C5H5), through the substitution of one carbonyl ligand with isocyanides (2-4) and the subsequent substitution of a second CO with N- or P-ligands (5-7). All products 2-7 were structurally characterized using IR and multinuclear NMR spectroscopy. One compound from series (7) was also characterized by single crystal X-ray diffraction. Complexes 2-7 exhibit outstanding stability in physiological-like solutions, with 92-97 % of the compounds unchanged after storing in DMEM at 37 °C for 24 h, and substantial amphiphilicity, with most of Log Pow values falling in the range -1 to +1. Complexes 3, 4, 5 and 7 exhibited cytotoxic activity against human (HCT 116, MCF-7, A2780) and murine (CT26, 4T1, B16-F1, B16-F10) cancer cell lines with IC50 values up to the nanomolar range, along with moderate selectivity toward the malignant phenotype. The induction of cell differentiation, senescence, and apoptotic cell death with cell-specific redox response were in the background of cytotoxic activity. However, limited tumor volume reduction and observed systemic toxicity in vivo indicated the need for additional structure-activity relationship studies to optimize the compounds anticancer profile.
Collapse
Affiliation(s)
- Ekatarina Mihajlović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia
| | - Lorenzo Biancalana
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, I-56124 Pisa, Italy.
| | - Marija Mojić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia
| | - Sanja Jelača
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia
| | - Lorenzo Chiaverini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, I-56124 Pisa, Italy
| | - Stefano Zacchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Via Piero Gobetti 85, I-40129 Bologna, Italy
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia.
| | - Fabio Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, I-56124 Pisa, Italy.
| |
Collapse
|
27
|
Sun H, Wang D, Zheng Y, Ye Y. Elucidating the Molecular Mechanisms of Hederagenin-Regulated Mitophagy in Cervical Cancer SiHa Cells through an Integrative Approach Combining Proteomics and Advanced Network Association Algorithm. J Proteome Res 2025; 24:2081-2095. [PMID: 40135937 PMCID: PMC11976847 DOI: 10.1021/acs.jproteome.5c00022] [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: 01/09/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Hederagenin (Hed), a natural triterpenoid, exhibits antitumor potential in cervical cancer. The present study was designed to explore Hed's regulatory mechanisms on mitophagy in SiHa cervical cancer cells, employing tandem mass tag (TMT) proteomics and an advanced network association algorithm (NAA). Our findings revealed that Hed decreased SiHa cell viability, induced apoptosis, and altered mitochondrial membrane potential. Notably, Hed inhibited mitophagic flux under both normoxic and hypoxic conditions. Through TMT proteomics analysis and innovative NAA, we identified a close association between the HIF-1 signaling pathway and mitophagy. Network analysis further suggested that Hed acts on a target network centered on SRC, STAT3, AKT1, and HIF1A. Western blot analysis confirmed the expression and phosphorylation status of these targets in response to Hed. This study elucidates the molecular mechanisms underlying Hed's regulation of mitophagy in SiHa cells, offering novel insights and potential therapeutic targets for cervical cancer treatment.
Collapse
Affiliation(s)
- Hao Sun
- Pharmacy
Department, Women’s Hospital, Zhejiang
University School of Medicine, Hangzhou 310006, China
| | - Dan Wang
- Pharmacy
Department, Zhejiang Hospital, Hangzhou 310030, China
| | - Yongquan Zheng
- Pharmacy
Department, Women’s Hospital, Zhejiang
University School of Medicine, Hangzhou 310006, China
| | - Yiqing Ye
- Pharmacy
Department, Women’s Hospital, Zhejiang
University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
28
|
Xu LB, Smith ER, Koutouratsas V, Chen ZS, Xu XX. The Persistent Power of the Taxane/Platin Chemotherapy. Cancers (Basel) 2025; 17:1208. [PMID: 40227809 PMCID: PMC11987835 DOI: 10.3390/cancers17071208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
The cancer chemotherapy regimen of a taxane and platinum combination was developed more than forty years ago, yet remains the cornerstone of treatment for several major cancer types today. Although many new agents targeting cancer genes and pathways have been developed and evaluated, none have been sufficient to replace the long-established taxane/platinum combination. This leads us to ponder why, after four decades of colossal efforts, multiple discoveries, and tremendous advances in understanding gene mutations and biology, the development of conceptually superior targeted therapies has not yet achieved overwhelming success in replacing cytotoxic chemotherapy. The concept of targeted therapy is based on the idea that blocking the altered pathway(s) crucial for cancer development (and maintenance), the disturbance in cellular signaling, metabolism, and functions will make the targeted cancer cells unfit and trigger programmed cell death in cancer cells, but without the significant side effects that limit chemotherapy. We propose that the lack of anticipated triumphs of targeted therapy stems from the desensitization of programmed cell death pathways during neoplastic transformation and malignant progression of cancer cells. This renders the targeting drugs largely ineffective at killing cancer cells and mostly insufficient in clinical implements. Recent advances in understanding suggest that, in contrast to targeted therapies, taxanes and platinum agents kill cancer cells by physical rupturing nuclear membranes rather than triggering apoptosis, making their effect independent of the intrinsic cellular programmed cell death mechanism. This new recognition of the non-programmed cell death mechanism in the success of chemotherapeutic agents, such as taxanes and platinum, may inspire oncologists and cancer researchers to focus their efforts more productively on developing effective non-programmed cell death cancer therapies to replace or significantly improve the application of the current standard taxane/platinum regimens.
Collapse
Affiliation(s)
- Lucy B. Xu
- Department of Biology, University of Miami, Miami, FL 33136, USA;
| | - Elizabeth R. Smith
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Vasili Koutouratsas
- College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (V.K.)
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (V.K.)
| | - Xiang-Xi Xu
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
29
|
Yang J, Yue G, Fan Z, Zhang N, Nie S, Li J, Ji Y. FOXA1 Targets NEK2 to Mediate Cisplatin Resistance in Lung Adenocarcinoma Cells by Activating DNA Damage Repair. Drug Dev Res 2025; 86:e70087. [PMID: 40233258 DOI: 10.1002/ddr.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
Lung adenocarcinoma (LUAD) is one of the main causes of death in cancer patients, as its hidden course is difficult to uncover, resulting in many patients being diagnosed as advanced. Late-stage LUAD patients are prone to develop resistance to cisplatin. This study aims to explore the potential molecular regulatory mechanism of NEK2 on cisplatin resistance in LUAD cells. The expression levels of NEK2 and FOXA1 in LUAD tissues were analyzed based on bioinformatics methods. qRT-PCR analysis was carried out to measure the mRNA expression levels of NEK2 and FOXA1 in LUAD cells. CCK8 detected and calculated cell viability and IC50 values for each group of cells. Gene set enrichment analysis (GSEA) analyzed signaling pathways enriched by the NEK2 gene in LUAD. Dual luciferase and CHIP experiments were conducted to verify the binding relationship between NEK2 and FOXA1. Comet assay was utilized to analyze the level of DNA damage in LUAD cells. Western blot (WB) measured the expression levels of DNA damage-related proteins (γ-H2AX, p-ATM). The experimental results showed that FOXA1 and NEK2 were highly expressed in LUAD tissues and cells. GSEA analysis showed that NEK2 was enriched in DNA damage-related pathways, and silencing NEK2 could reduce the vitality of LUAD cisplatin-resistant cells, lower the IC50 value of cells to cisplatin, and increase their DNA damage levels. FOXA1 can target the promoter region that binds to NEK2, and it can activate NEK2 through transcription to promote DNA damage repair and cisplatin resistance in cisplatin-resistant LUAD cells. This study confirms that FOXA1 can target NEK2 to promote DNA damage repair and cisplatin resistance in LUAD cells, providing a new valuable therapeutic target for the treatment of LUAD and the control of chemotherapy drug resistance.
Collapse
Affiliation(s)
- Junhong Yang
- Department of Medical Oncology, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Guangcheng Yue
- Department of Thoracic Surgery, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Zhiguo Fan
- Department of Medical Oncology, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Ning Zhang
- Department of Medical Oncology, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Shiwei Nie
- Department of Thoracic Surgery, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Jing Li
- Department of Medical Oncology, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| | - Yuanyuan Ji
- Department of Medical Oncology, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, 455000, China
| |
Collapse
|
30
|
Xie DM, Li ZY, Ren BK, Gong R, Yang D, Huang S. Tanshinone II A Facilitates Chemosensitivity of Osteosarcoma Cells to Cisplatin via Activation of p38 MAPK Pathway. Chin J Integr Med 2025; 31:326-335. [PMID: 39499413 DOI: 10.1007/s11655-024-4118-5] [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: 04/19/2024] [Indexed: 11/07/2024]
Abstract
OBJECTIVE To examine the mechanism of action of tanshinone II A (Tan II A) in promoting chemosensitization of osteosarcoma cells to cisplatin (DDP). METHODS The effects of different concentrations of Tan II A (0-80 µ mol/L) and DDP (0-2 µ mol/L) on the proliferation of osteosarcoma cell lines (U2R, U2OS, 143B, and HOS) at different times were examined using the cell counting kit-8 and colony formation assays. Migration and invasion of U2R and U2OS cells were detected after 24 h treatment with 30 µ mol/L Tan II A, 0.5 µ mol/L DDP alone, and a combination of 10 µ mol/L Tan II A and 0.25 µ mol/L DDP using the transwell assay. After 48 h of treatment of U2R and U2OS cells with predetermined concentrations of each group of drugs, the cell cycle was analyzed using a cell cycle detection kit and flow cytometry. After 48 h treatment, apoptosis of U2R and U2OS cells was detected using annexin V-FITC apoptosis detection kit and flow cytometry. U2R cells were inoculated into the unilateral axilla of nude mice and then the mice were randomly divided into 4 groups of 6 nude mice each. The 4 groups were treated with equal volume of Tan II A (15 mg/kg), DDP (3 mg/kg), Tan II A (7.5 mg/kg) + DDP (1.5 mg/kg), and normal saline, respectively. The body weight of the nude mice was weighed, and the tumor volume and weight were measured. Cell-related gene and signaling pathway expression were detected by RNA sequencing and Kyoto Encyclopedia of Genes and Genomes pathway analysis. p38 MAPK signaling pathway proteins and apoptotic protein expressions were detected by Western blot. RESULTS In vitro studies have shown that Tan II A, DDP and the combination of Tan II A and DDP inhibit the proliferation, migration and invasion of osteosarcoma cells. The inhibitory effect was more pronounced in the Tan II A and DDP combined treatment group (P<0.05 or P<0.01). Osteosarcoma cells underwent significantly cell-cycle arrest and cell apoptosis by Tan II A-DDP combination treatment (P<0.05 or P<0.01). In vivo studies demonstrated that the Tan II A-DD combination treatment group significantly inhibited tumor growth compared to the Tan II A and DDP single drug group (P<0.01). Additionally, we found that the combination of Tan II A and DDP treatment enhanced the p38 MAPK signaling pathway. Western blot assays showed higher p-p38, cleaved caspase-3, and Bax and lower caspase-3, and Bcl-2 expressions with the combination of Tan II A and DDP treatment compared to the single drug treatment (P<0.01). CONCLUSION Tan II A synergizes with DDP by activating the p38/MAPK pathway to upregulate cleaved caspase-3 and Bax pro-apoptotic gene expressions, and downregulate caspase-3 and Bcl-2 inhibitory apoptotic gene expressions, thereby enhancing the chemosensitivity of osteosarcoma cells to DDP.
Collapse
Affiliation(s)
- Da-Ming Xie
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, 330006, China
| | - Zhi-Yun Li
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Bing-Kai Ren
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Rui Gong
- Department of Clinical Medicine, Jiangxi Health Vocational College, Nanchang, 330052, China
| | - Dong Yang
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, 330006, China
| | - Sheng Huang
- Department of Orthopaedics, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
- Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Nanchang, 330006, China.
| |
Collapse
|
31
|
Miller EJ, Galsky MD. An evaluation of nivolumab plus gemcitabine and cisplatin in the treatment of advanced urothelial carcinoma. Expert Rev Anticancer Ther 2025; 25:319-326. [PMID: 40016118 DOI: 10.1080/14737140.2025.2473645] [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: 11/13/2024] [Revised: 02/10/2025] [Accepted: 02/25/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION For decades, first-line treatment for advanced/metastatic urothelial cancer has been platinum-based chemotherapy. However, many patients do not respond to platinum-based chemotherapy alone, and the vast majority do not have durable responses. While immune checkpoint blockade has demonstrated benefit in this setting, initial trials of concurrent chemotherapy and immune checkpoint blockade did not demonstrate improvements in overall survival. AREAS COVERED The recent CheckMate 901 trial compared gemcitabine, cisplatin, plus nivolumab to gemcitabine and cisplatin alone for first-line treatment of advanced/metastatic urothelial cancer. This was the first trial to demonstrate significant benefit in the combined chemotherapy and immune checkpoint blockade arm in advanced/metastatic urothelial cancer, most significantly showing an improvement in the primary outcomes of progression-free survival and overall survival, and the exploratory outcomes of objective response rate, complete response rate, and duration of complete response. EXPERT OPINION The combination of gemcitabine, cisplatin, plus nivolumab represents a new first-line treatment option for metastatic urothelial cancer. This article details the clinical benefit observed and how this establishes proof-of-concept for prior hypotheses related to the importance of the specific chemotherapy regimen combined with immune checkpoint blockade, revolving around immunomodulatory mechanisms of action of cisplatin, and synergy of these mechanisms with immunotherapy.
Collapse
Affiliation(s)
- Eric J Miller
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Galsky
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
32
|
Kim JH, Han JY, Kwon JH, Kim MG. Cardioprotective effects of PARP inhibitors for platinum-agent induced cardiotoxicity. Invest New Drugs 2025; 43:243-249. [PMID: 39962008 DOI: 10.1007/s10637-025-01509-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/28/2025] [Indexed: 05/03/2025]
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors may have cardioprotective properties. This study aimed to evaluate the potential cardioprotective effects of PARP inhibitors in patients with epithelial ovarian cancer treated with platinum-based chemotherapeutic agents. A retrospective cohort study was conducted using the Health Insurance Review & Assessment Service claims database from January 2007 to July 2022. Eligible patients were those diagnosed with ovarian, primary peritoneal, or fallopian tube cancer who received platinum-based chemotherapy after 2017. Propensity score matching was employed to adjust for potential confounders, and logistic regression and Cox proportional hazards regression analyses were utilized to estimate the odds ratios, hazard ratios, and 95% confidence intervals (CIs) for the occurrence of cardiac adverse events, including myocardial infarction, cardiomyopathy, and heart failure. A total of 7,253 eligible patients were included in the study, of which 233 (3.2%) used PARP inhibitors. After propensity score matching, no significant cardioprotective effect was observed in the PARP inhibitor-exposed group compared to the non-exposed group (adjusted odds ratio, 0.753; 95% CI 0.275-2.059; adjusted hazard ratio, 0.601; 95% CI 0.228-1.584). Although no statistically significant cardioprotective effect of PARP inhibitors was found in this study, there was a directional trend suggesting that patients with gynecologic malignancies treated with platinum-based chemotherapy could potentially benefit from PARP inhibitors. Further research with larger sample sizes and longer follow-up periods is warranted to elucidate the role of PARP inhibitors in mitigating cardiac adverse events in this patient population.
Collapse
Affiliation(s)
- Jae Hyun Kim
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju, Republic of Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Ja-Young Han
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Jae-Hee Kwon
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Myeong Gyu Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 ewhayeodae- gil seodaemun-gu, Seoul, 03760, Republic of Korea.
| |
Collapse
|
33
|
Khalil EM, Rady MI, Darwish SF, Abd-Allah ER. Nano Spirulina platensis countered cisplatin-induced repro-toxicity by reversing the expression of altered steroid hormones and downregulation of the StAR gene. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4053-4070. [PMID: 39414699 PMCID: PMC11978726 DOI: 10.1007/s00210-024-03483-z] [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: 09/04/2024] [Accepted: 09/21/2024] [Indexed: 10/18/2024]
Abstract
Cisplatin is a commonly utilized chemotherapy medication for treating different sarcomas and carcinomas. Its ability interferes with cancer cells' DNA repair pathways and postpones unfavorable outcomes in cancer patients. The current investigation's goal was to ascertain if nano Spirulina platensis (NSP) might shield rat testicles from cisplatin damage by assessing the expression of the StAR and SOD genes, sex hormones, 17ß-hydroxysteroid dehydrogenase(17ß-HSD), sperm profile picture, oxidative condition of testes, testicular histology, and DNA damage. Four equal and random groups of 28 adult male Wistar rats were created; the control group was given saline for 8 weeks. An extraction of NSP at a concentration of 2500 mg/kg body weight was administered orally for 8 weeks to the NSP group. For the first 4 weeks, the cisplatin group was intraperitoneally injected with 2 mg/kg/body weight of cisplatin, and for the next 4 weeks, they were given a dosage of 4 mg/kg/body weight. The cisplatin + NSP group was given both NSP and cisplatin. The results of the experiment showed that intake of NSP and cisplatin improved sperm profile; re-established the balance of oxidizing agents and antioxidant state; enhanced testicular histology; promoted the histometric parameters of seminiferous tubules including epithelial height, their diameter, and Johnsen's score, decreasing DNA breakage in testicular tissue; increased testosterone level; decreased 17ß-HSD concentration; and upregulated both the StAR and SOD gene expression in testicles compared to rats exposed to cisplatin alone. These results demonstrate that NSP is a promising agent for improving cisplatin-induced testicular injury and infertility.
Collapse
Affiliation(s)
- Eman M Khalil
- Department of Zoology, Faculty of Science (Girls), Al-Azhar University, Nasr City, Egypt
| | - Mohamed I Rady
- Department of Zoology, Faculty of Science (Boys), Al-Azhar University, Nasr City, Egypt
| | - Samah F Darwish
- Biotechnology Research Unit, Animal Reproduction Research Institute, Giza, Egypt
| | - Entsar R Abd-Allah
- Department of Zoology, Faculty of Science (Girls), Al-Azhar University, Nasr City, Egypt.
| |
Collapse
|
34
|
Wang H, Wang X, Shen W, Zhou Y, Cui J, Li H, Yu J. CircRNA (circ)_0007823 Contributes to Triple-Negative Breast Cancer Progression and Cisplatin Resistance via the miR-182-5p/FOXO1 Pathway. Biochem Genet 2025; 63:1330-1342. [PMID: 38557813 DOI: 10.1007/s10528-024-10783-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 03/13/2024] [Indexed: 04/04/2024]
Abstract
Cisplatin (DDP) is used for the clinical management of triple-negative breast cancer (TNBC). However, the development of drug resistance limits its therapeutic efficacy. Circular RNAs (circRNAs) are known to be involved in tumor DDP resistance. In our previous study, we reported that circ_0007823 expression is downregulated and correlated with adverse prognosis in TNBC. However, its association with DDP resistance remains unclear. This study aimed to determine the role of circ_0007823 and miR-182-5p in DDP-resistant TNBC and explore the underlying mechanisms. First, expression profiles circ_0007823, microRNA (miR)-182-5p, and forkhead box O1 (FOXO1) in TNBC cells were determined. Additionally, biological characteristics of cells, including apoptosis, cell cycle, proliferation, and migration, were analyzed using various assays. Luciferase reporter and rescue assays were used to determine the correlations among circ_0007823, miR-182-5p, and FOXO1 expression. MiR-182-5p was overexpressed in DDP-resistant TNBC cells. MiR-182-5p knockdown suppressed the invasiveness and increased the apoptosis of drug-resistant cells, contributing to G1 arrest and S phase reduction. Mechanistically, circ_0007823 targeted miR-182-5p, and its overexpression drastically reduced the promotional effects of the miR-182-5p mimic on the aggression and transfer ability of drug-resistant cells. Furthermore, FOXO1 overexpression increased the sensitivity of cells to DDP and reduced their malignant progression. Therefore, FOXO1 was established as the downstream target of miR-182-5p that may be used to treat DDP-resistant TNBC. In summary, circ_0007823 overexpression attenuated DDP resistance in TNBC via the miR-182-5p-FOXO1 axis, indicating the therapeutic potential of circ_0007823 DDP-resistant TNBC treatment.
Collapse
Affiliation(s)
- Haofeng Wang
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, No. 786, Yuyuan Road, Changning District, Shanghai, 200050, China
| | - Xiaojie Wang
- Department of Pathology, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, China
| | - Weida Shen
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, No. 786, Yuyuan Road, Changning District, Shanghai, 200050, China
| | - Yingzi Zhou
- Department of Pathology, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, China
| | - Jing Cui
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, No. 786, Yuyuan Road, Changning District, Shanghai, 200050, China
| | - Haichuan Li
- Department of Laboratory, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, 200050, China
| | - Jinling Yu
- Department of Breast Surgery, Shanghai Changning Maternity and Infant Health Hospital, East China Normal University, No. 786, Yuyuan Road, Changning District, Shanghai, 200050, China.
| |
Collapse
|
35
|
Jin SK, Baek KH. Unraveling the role of deubiquitinating enzymes on cisplatin resistance in several cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189297. [PMID: 40058507 DOI: 10.1016/j.bbcan.2025.189297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
The use of platinum-based drugs in cancer treatment is one of the most common methods in chemotherapy. Especially, cisplatin induces cell death by interrupting DNA synthesis by binding to the DNA bases, thereby leading to the apoptosis via multiple pathways. However, the major hurdle in chemotherapy is drug resistance. To overcome drug resistance, the ubiquitin-proteasome system (UPS) has emerged as a potential therapeutic target. The UPS is a pivotal signaling pathway that regulates the majority of cellular proteins by attaching ubiquitin to substrates, leading to proteasomal degradation. Conversely, deubiquitinating enzymes (DUBs) remove tagged ubiquitin from the substrate and inhibit degradation, thereby maintaining proteostasis. Recently, studies have been conducted to identify the substrates of DUBs and investigated the cellular mechanisms, and now the development of therapeutics using DUB inhibitors is in clinical trials. However, the mechanism of the DUB response to cisplatin remains still unclear. In this review, we summarize the research reported on the function of DUBs responding to cisplatin.
Collapse
Affiliation(s)
- Sun-Kyu Jin
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea.
| |
Collapse
|
36
|
Alanazi ST, Salama SA, Althobaiti MM, Bakhsh A, Aljehani NM, Alanazi E, Alanazi MT, Musa A. Theaflavin alleviates cisplatin-induced nephrotoxicity: Targeting SIRT1/p53/FOXO3a/Nrf2 signaling and the NF-kB inflammatory cascade. Food Chem Toxicol 2025; 198:115334. [PMID: 39978533 DOI: 10.1016/j.fct.2025.115334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/09/2025] [Accepted: 02/18/2025] [Indexed: 02/22/2025]
Abstract
Cisplatin is a widely used chemotherapeutic agent. Nevertheless, a significant fraction of cisplatin-treated patients develops nephrotoxicity which limits cisplatin therapeutic implementation. The current work was devoted to investigate the potential nephroprotective impact of theaflavin against the cisplatin-induced nephrotoxicity using male Wistar rats as a mammalian model. The results indicated that theaflavin significantly improved the renal histopathological picture and glomerular filtration rate, along with reduced renal injury marker KIM-1, urinary albumin/creatinine ratio, serum creatinine, and urea. Mechanistically, theaflavin upregulated protein level of SIRT1 and downregulated the acetylated forms of the inflammatory transcription factor (TF) NF-kB, the antioxidant TF FOXO3a, and the pro-apoptotic TF p53 in the cisplatin-treated rats. Additionally, it upregulated the antioxidant TF Nrf2. In the same context, it suppressed the inflammatory responses, oxidative stress, and apoptosis. NF-kB nuclear translocation and levels of its responsive gene products IL-6 and TNF-α were suppressed. Lipids and DNA oxidation were reduced, and level of the antioxidant GSH and activity of the antioxidant enzymes SOD, GPx, and CAT were increased. The apoptotic markers caspase-3, BAX, and Bcl2 were modulated. Collectively, these findings highlight the nephroprotective competency of theaflavin against cisplatin-induced nephrotoxicity and underscore modulations of SIRT1, p53, FOXO3a, Nrf2, and NF-kB as potential targets.
Collapse
Affiliation(s)
- Samyah T Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| | - Musaad M Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Afnan Bakhsh
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Najla M Aljehani
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, 13316, Saudi Arabia
| | - Ebtisam Alanazi
- Cardiac Center King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Maha T Alanazi
- Medical Imaging, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Arafa Musa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| |
Collapse
|
37
|
Razuwika R, Sookai S, Aronson R, Kaur M, Munro OQ. A Cytotoxic Indazole-based Gold(III) Carboxamide Pincer Complex Targeting DNA Through Dual Binding Modes of Groove Binding and Alkylation. Chemistry 2025:e202404345. [PMID: 40162557 DOI: 10.1002/chem.202404345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/02/2025]
Abstract
Gold(III) complexes have garnered increasing attention in drug delivery due to their structural and mechanistic similarities to cisplatin. This study investigates an indazole-based gold(III) carboxamide pincer complex, [N2·N6-bis(1-methyl-1H-indazol-3-yl)pyridine-2·6-dicarboxamide]gold(III) chloride (AuL), for its potential as an anticancer agent. Speciation analysis at physiological pH revealed that AuL predominantly exists as a neutral chlorinated species. The complex exhibited strong cytotoxicity against the MCF-7 breast cancer cell line, with an impressive IC50 value of 9 µM, while showing no significant activity against the HT-29 colon cancer cell line. Comprehensive analysis using electrophoresis, viscometry, ultraviolet-visible spectroscopy (UV-Vis), circular dichroism (CD), linear dichroism (LD) spectroscopy, and biomolecular simulations demonstrated that AuL binds to DNA via a dual mechanism, specifically minor groove binding and alkylation, with binding constants Ka1 = 1.48 × 109 M-1 and Ka2 = 6.59 × 105 M-1, respectively. Our data indicate that AuL initially binds to the minor groove of DNA, at which point a nucleobase substitutes the Cl ion, resulting in AuL binding directly to the DNA bases. In conclusion, the dual binding mode of AuL with DNA underscores its potential as a promising anticancer agent, opening new avenues for drug discovery and the development of metal-based therapeutics.
Collapse
Affiliation(s)
- Rufaro Razuwika
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smuts Avenue, Johannesburg, 2050, South Africa
| | - Sheldon Sookai
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smuts Avenue, Johannesburg, 2050, South Africa
| | - Ruth Aronson
- School of Molecular and Cell Biology, University of the Witwatersrand, 1 Jan Smuts Avenue, Johannesburg, 2050, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, 1 Jan Smuts Avenue, Johannesburg, 2050, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smuts Avenue, Johannesburg, 2050, South Africa
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK
| |
Collapse
|
38
|
Zhao QY, Liu WJ, Wang JG, Li H, Lv JL, Wang Y, Wang C. Increasing cisplatin exposure promotes small-cell lung cancer transformation after a shift from glucose metabolism to fatty acid metabolism. J Cancer Res Clin Oncol 2025; 151:126. [PMID: 40155472 PMCID: PMC11953189 DOI: 10.1007/s00432-025-06164-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVES Lung cancer is a leading cause of global cancer mortality. Clinical observations reveal that histological transformation from non-small cell lung cancer (NSCLC) to small cell lung cancer (SCLC) is accompanied by mutations in TP53 and RB1. By applying gradually increasing cisplatin concentrations to mimic the escalating drug pressure within the tumor microenvironment, this study investigated the link between phenotypic transformation to SCLC in cisplatin-resistant human lung adenocarcinoma cells and alterations in cellular energy production pathways. MATERIALS AND METHODS We established two cisplatin-resistant NSCLC cell lines with varying resistance levels. RNAseq analyses identified TP53 and RB1 gene mutations. Comprehensive functional assays were performed to characterize A549/DDP1 μg/mL and A549/DDP3 μg/mL cells, focusing on proliferation and migratory capabilities. Cellular bioenergetics were assessed through glycolysis and oxidative phosphorylation analyses. Western blotting was employed to examine epithelial-mesenchymal transition (EMT), glucose metabolism, and lipid metabolism markers. Cell cycle distribution was analyzed by flow cytometry. Additionally, a xenograft mouse model was developed for in vivo validation. RESULTS TP53 and RB1 mutations were associated with cisplatin concentration-dependent phenotypic transformation, with A549/DDP cells acquiring a more aggressive SCLC-like phenotype (In the article we call the A549/DDPSCLC cells). Analysis of cell bioenergetics profiling and Western blot analyses revealed enhanced glucose metabolism in A549/DDP1 μg/mL cells, while A549/DDPSCLC cells exhibited predominant lipid metabolism. Compound3K and Etomoxir specifically inhibit the activity of PKM2 and CPT1A, respectively, with Etomoxir demonstrating substantially inhibited A549/DDPSCLC cells growth and more cell cycle arrest in the G0/G1 phase. Combinatorial of Compound3K and Etomoxir effectively induced cell death in A549/DDPSCLC phenotype cells in vitro. Etomoxir alone or combined with Compound3K significantly inhibited tumor growth in vivo, with enhanced efficacy in the combination group. CONCLUSIONS This study provides the first evidence of cisplatin concentration-dependent metabolic reprogramming during NSCLC-to-SCLC transformation. We identified a phenotypic transition from NSCLC to SCLC accompanied by a metabolic shift from glucose to fatty acid metabolism, offering new insights into therapeutic strategies for treatmentresistant lung cancer.
Collapse
MESH Headings
- Cisplatin/pharmacology
- Humans
- Lung Neoplasms/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Animals
- Small Cell Lung Carcinoma/pathology
- Small Cell Lung Carcinoma/metabolism
- Small Cell Lung Carcinoma/drug therapy
- Small Cell Lung Carcinoma/genetics
- Mice
- Glucose/metabolism
- Fatty Acids/metabolism
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Xenograft Model Antitumor Assays
- Antineoplastic Agents/pharmacology
- Drug Resistance, Neoplasm
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Protein p53/genetics
- Cell Proliferation/drug effects
- Mutation
- Mice, Nude
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Cell Line, Tumor
- Epithelial-Mesenchymal Transition/drug effects
- A549 Cells
Collapse
Affiliation(s)
- Qiu-Yu Zhao
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Wen-Jun Liu
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Jian-Guang Wang
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - He Li
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Jia-Lu Lv
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Yumeng Wang
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, 201210, China
| | - Chun Wang
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China.
| |
Collapse
|
39
|
Li S, Zhang D, Li Y, Zhou J, Chen J, Zhang Y. All-in-one multifunctional tri-block glycopolymers for targeted delivery of cisplatin and cancer chemotherapy. Colloids Surf B Biointerfaces 2025; 252:114639. [PMID: 40132337 DOI: 10.1016/j.colsurfb.2025.114639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/11/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
Cisplatin (CDDP) as a first-line chemotherapy drug has long suffered drawbacks of severe side effects and poor pharmacokinetics. Thus, various nano-carriers of complex architectures and multi-step modifications have been developed to overcome these challenges, but a simple delivery system with integrated multi-functions is still in demand. Herein, we synthesized a new type of glyco-based triple hydrophilic block copolymers for the delivery of CDDP and thereof cancer therapy. The incorporated glucuronic acid part is complexed with CDDP with high entrapment efficiency, while providing pH-responsive release in the acidic tumor microenvironment. The galactose segment is implanted for liver-targeting effect, which can also significantly lower the side effects of CDDP. As a result, the CDDP-loaded nanoparticle PGG2/Pt showed selectively faster endocytosis rates into HepG2 cells in vitro, with the calculated IC50 value even comparable to that of free CDDP. The in vivo experiments on a HepG2-bearing mouse model, PGG2/Pt showed excellent anti-tumor activity and enhanced drug accumulation on tumor, together with much lowered nephrotoxicity, hepatotoxicity, and splenic toxicity. This work provides a new strategy for CDDP delivery with higher loading efficiency as well as biosafety.
Collapse
Affiliation(s)
- Sichen Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Difei Zhang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Yi Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Jingjing Zhou
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China.
| | - Yan Zhang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|
40
|
Yu W, Yin S, Tang H, Li H, Zhang Z, Yang K. PER2 interaction with HSP70 promotes cuproptosis in oral squamous carcinoma cells by decreasing AKT stability. Cell Death Dis 2025; 16:192. [PMID: 40113747 PMCID: PMC11926231 DOI: 10.1038/s41419-025-07523-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Oral squamous cell carcinoma (OSCC) has a poor prognosis, with unclear mechanisms posing a challenge for the development of effective treatment strategies. Cuproptosis is a novel cell death mode that disrupts mitochondrial metabolism. Clarifying the mechanisms that regulate cuproptosis may provide important new insights to guide OSCC treatment. Here, we found that the biological clock gene Period2 (PER2) was under-expressed in OSCC, with consequent inhibition of cellular cuproptosis, whereas it was overexpression of PER2 in vitro and in vivo and promoted OSCC cellular cuproptosis. Mechanistically, PER2 bound to heat shock protein 70 (HSP70) through its C-terminal domain, subsequently reducing the interaction between HSP70 and AKT and leading to enhanced degradation of AKT ubiquitination, and promoting cuproptosis in OSCC cells by inhibiting the AKT pathway and upregulating DLAT, PDHB, and SLC31A1 expression. Activating transcription factor 3 (ATF3) is an upstream regulator of PER2, that binds to the -807 to -796 bp site of the PER2 promoter. Overexpression of ATF3 in vitro and in vivo is dependent on transcriptional activation of PER2 and promotes cuproptosis in OSCC cells. The anti-tumor effect of ATF3 inducer 1-targeted upregulation of PER2 combined with copper ionophore elesclomol (ES) was found to be significantly enhanced compared with that of monotherapy in an OSCC xenograft model. These findings reveal a critical role of ATF3-dependent regulation of cuproptosis by PER2 in OSCC development, suggesting targeted upregulation of PER2 or ATF3 in combination to induce cuproptosis as a novel strategy to potentially improve the prognosis of OSCC patients.
Collapse
Affiliation(s)
- Wenguang Yu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shilin Yin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Tang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hengyan Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhiwei Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Kai Yang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
41
|
Warren GM, Shuman S. In vivo nucleotide excision repair by mycobacterial UvrD1 requires ATP hydrolysis but does not depend on cysteine disulfide-mediated dimerization and DNA unwinding. Nucleic Acids Res 2025; 53:gkaf269. [PMID: 40193706 PMCID: PMC11975281 DOI: 10.1093/nar/gkaf269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Mycobacterial UvrD1 is an SF1-type ATPase that participates in nucleotide excision repair (NER). UvrD1 consists of N-terminal ATPase and C-terminal Tudor domains. The monomeric UvrD1 characterized originally displays vigorous DNA-dependent ATPase activity but only feeble helicase activity. A recent study demonstrated that: (i) cysteine disulfide-mediated homodimerization of UvrD1 generates a highly active helicase; and (ii) an obligate monomeric UvrD1 (by virtue of mutating the domain 2B cysteine) is active as an ATP-dependent 3'-to-5' single-stranded DNA translocase but not as a double-stranded DNA-unwinding helicase. Here we test genetically which physical and functional states of UvrD1 are relevant for its functions in DNA repair, by complementation of an NER-defective Mycobacterium smegmatis ΔuvrD1 strain with a series of biochemically-defined UvrD1 mutants. By assaying complemented strains for sensitivity to UVC, MMC, cisplatin, and psoralen-UVA, we conclude that monomeric UvrD1 ATPase activity suffices for the NER functions of UvrD1 in vivo. Decoupling ATP hydrolysis from duplex unwinding does not affect the repair activity of UvrD1, nor does interdiction of domain 2B cysteine disulfide-mediated dimerization or deletion of the Tudor domain. Our results militate against a proposed model in which UvrD1's repair function is governed by the redox state of the bacterium via its impact on UvrD1 dimerization and helicase activity.
Collapse
Affiliation(s)
- Garrett M Warren
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, NY, NY 10065, United States
| | - Stewart Shuman
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, NY, NY 10065, United States
| |
Collapse
|
42
|
Kiran S, Xue Y, Sarker DB, Sang QXA. Effects of Induced Pluripotent Stem Cell-Derived Astrocytes on Cisplatin Sensitivity in Pediatric Brain Cancer Cells. Cancers (Basel) 2025; 17:997. [PMID: 40149331 PMCID: PMC11940393 DOI: 10.3390/cancers17060997] [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: 01/27/2025] [Revised: 03/06/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background: ATRTs and DIPGs are deadly pediatric brain tumors with poor prognosis. These tumors can develop resistance to chemotherapies, which may be significantly influenced by their microenvironment. Since astrocytes are the most abundant glial cell type in the brain microenvironment and may support tumor growth and chemoresistance, this study investigated the effects of induced pluripotent stem cell-derived astrocytes (iPSC-astrocytes) on cisplatin sensitivity in CHLA-05-ATRT and SF8628 (DIPG) cells. iPSCs provide an unlimited and standardized source of nascent astrocytes, which enables modeling the interaction between childhood brain tumor cells and iPSC-astrocytes within a controlled coculture system. Methods: To study the effects on tumor growth, the iPSC-astrocytes were cocultured with tumor cells. Additionally, the tumor cells were exposed to various concentrations of cisplatin to evaluate their chemosensitivity in the presence of astrocytes. Results: The paracrine interaction of iPSC-astrocytes with tumor cells upregulated astrocyte activation markers GFAP and STAT3 and promoted tumor cell proliferation. Moreover, the cisplatin treatment significantly decreased the viability of CHLA-05-ATRT and SF8628 cells. However, tumor cells exhibited reduced sensitivity to cisplatin in the coculture with iPSC-astrocytes. During cisplatin treatment, DIPG cells in particular showed upregulation of resistance markers, ERK1, STAT3, and MTDH, which are associated with enhanced proliferation and invasion. They also had increased expression of APEX1, which is involved in the base excision repair pathway following cisplatin-induced DNA damage. Conclusion: These findings underscore the significance of the tumor microenvironment in modulating tumor cell survival and chemosensitivity.
Collapse
Affiliation(s)
- Sonia Kiran
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Yu Xue
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Drishty B. Sarker
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA; (S.K.); (Y.X.); (D.B.S.)
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
43
|
Stocchetti S, Vančo J, Bresciani G, Biancalana L, Belza J, Zacchini S, Dvořák Z, Benetti S, Biver T, Bortoluzzi M, Trávníček Z, Marchetti F. Anticancer diiron aminocarbyne complexes with labile N-donor ligands. Eur J Med Chem 2025; 286:117304. [PMID: 39862748 DOI: 10.1016/j.ejmech.2025.117304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The novel diiron amine complexes [Fe2Cp2(CO)(NH2R')(μ-CO){μ-CN(Me)(Cy)}]CF3SO3 [R' = H, 3; Cy, 4; CH2CH2NH2, 5; CH2CH2NMe2, 6; CH2CH2(4-C6H4OMe), 7; CH2CH2(4-C6H4OH), 8; Cp = η5-C5H5, Cy = C6H11 = cyclohexyl] were synthesized in 49-92 % yields from [Fe2Cp2(CO)2(μ-CO){μ-CN(Me)(Cy)}]CF3SO3, 1a, using a straightforward two-step procedure. They were characterized by IR and multinuclear NMR spectroscopy, and the structure of 7 was confirmed through X-ray diffraction analysis. Complexes 3-8 and the acetonitrile adducts [Fe2Cp2(CO)(NCMe)(μ-CO){μ-CN(Me)(R)}]CF3SO3 (R = Cy, 2a; Me, 2b; Xyl = 2,6-C6H3Me2, 2c) were assessed for their water solubility, octanol-water partition coefficient and stability in physiological-like solutions. The in vitro antiproliferative activity of 2a-c and 3-8 was tested on seven human cancer cell lines (A2780, A2780R, PC3, A549, MCF7, HOS and HT-29), while the selectivity was evaluated using normal MRC-5 cells. Overall, the complexes exhibited variable cytotoxicity, with IC50 values reaching the low micromolar range for 3, 7 and 8 in A2780 and A2780R cells, along with significant selectivity. Targeted experiments covered cell cycle modification, induction of cell death, mitochondrial membrane potential, ROS production and interaction with DNA and bovine serum albumin (BSA) as a model protein. The interaction of 3 with BSA was further investigated through computational studies. Results showed a negligible increase in intracellular ROS levels (except for 2b) and insignificant changes in mitochondrial membrane potential.
Collapse
Affiliation(s)
- Sara Stocchetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Ján Vančo
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Giulio Bresciani
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Lorenzo Biancalana
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Jan Belza
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Stefano Zacchini
- University of Bologna, Department of Industrial Chemistry "Toso Montanari", I-40136, Bologna, Italy
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Sara Benetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Tarita Biver
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Marco Bortoluzzi
- University of Venezia "Ca' Foscari", Department of Molecular Science and Nanosystems, Via Torino 155, I-30170, Mestre (VE), Italy
| | - Zdeněk Trávníček
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic.
| | - Fabio Marchetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy.
| |
Collapse
|
44
|
Liu XM, Li Z, Wang XY, Ding BW, Wang JQ, Qiao X, Feng YK, Hao JH, Xu JY. Self-assembled HO-1i-Pt(IV) nanomedicine targeting p38/MAPK and MDR pathways for cancer chemo-immunotherapy. J Control Release 2025; 379:797-813. [PMID: 39848589 DOI: 10.1016/j.jconrel.2025.01.050] [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: 09/05/2024] [Revised: 01/08/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
Platinum(II)-based antitumor drugs are widely used in clinics but limited by severe side effects and resistance. Multi-target Platinum(IV) complexes are emerging as ideal alternatives. Heme oxygenase-1 (HO-1) works as a rate-limiting step in heme degradation and is overexpressed in malignant tumors. Herein, HO-1i-based Platinum(IV) prodrugs are prepared and candidate complex 15 is further developed into self-assembled nanoparticles (15-NPs). 15 and 15-NPs significantly increase cytotoxicity, particularly in HepG2 (74.77- and 96.14-fold increases) and A549cisR (38.6- and 47.24-fold increases), while reducing toxicity towards normal cells compared to cisplatin. In vitro experiments show 15 and 15-NPs activated multiple pathways, including p38/MAPK- and MDR-related proteins, achieving multi-target synergistic chemosensitization and anti-resistance, further verified by RNA-sequencing analysis. In vivo tests demonstrate that 15 and 15-NPs efficiently inhibit tumor growth and systemic toxicity, especially 15-NPs with optimal tumor-inhibition rate and survival (80% and 100%), superior to cisplatin (40% and 50%), attributing to its extra endocytosis, EPR effect, and precisely tumor-targeted release besides the advantage of a free HO-1i-Pt(IV) prodrug. Additionally, 15 and 15-NPs distinctly regulate T-cell and macrophage functions, thereby exhibiting a chemoimmuno-combined action. This study highlights that multi-functional Platinum(IV) prodrug target-delivered to tumors via carrier-free nanoparticles may represent an effective modality for improving cancer therapy.
Collapse
Affiliation(s)
- Xiao-Meng Liu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiao-Ya Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Bo-Wen Ding
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jia-Qian Wang
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yu-Kuan Feng
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Ji-Hui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
45
|
Balewski Ł, Inkielewicz-Stępniak I, Gdaniec M, Turecka K, Hering A, Ordyszewska A, Kornicka A. Synthesis, Structure, and Stability of Copper(II) Complexes Containing Imidazoline-Phthalazine Ligands with Potential Anticancer Activity. Pharmaceuticals (Basel) 2025; 18:375. [PMID: 40143151 PMCID: PMC11946467 DOI: 10.3390/ph18030375] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Recently, there has been great interest in metallopharmaceuticals as potential anticancer agents. In this context, presented studies aim to synthesize and evaluate of two copper(II) complexes derived from phthalazine- and imidazoline-based ligands against on three human cancer cell lines: cervix epithelial cell line (HeLa), breast epithelial-like adenocarcinoma (MCF-7), and triple-negative breast epithelial cancer cell line (MDA-MB-231), as well as non-tumorigenic cell line (HDFa). Moreover their antimicrobial, and antioxidant properties were assessed. Methods: The synthetized compounds-both free ligands L1, L2, L3 and copper(II) complexes C1 and C2-were characterized by elemental analysis, infrared spectroscopy. Additionally, a single-crystal X-ray diffraction studies we performed for free ligand L3 and its copper(II) complex C2. The stability of Cu(II)-complexes C1 and C2 was evaluated by UV-Vis spectroscopy. The cytotoxic potency of free ligands and their copper(II) complexes was estimated on HeLa, MCF-7, MDA-MB-231, as well as non-cancerous HDFa by use of an MTT assay after 48 h of incubation. Moreover, the antimicrobial activity of ligands L1 and L3 and their copper(II) complexes C1 and C2 was evaluated using reference strains of the following bacteria and yeasts: Staphylococcus aureus, Escherichia coli, and Candida albicans. The free radical scavenging properties of free ligands L1, L3 and the corresponding copper(II) complexes C1, C2 was tested with two colorimetric methods-ABTS, DPPH, and reduction ability assay (FRAP). Additionally, the ADME webtool was used to assess the drug-likeness of the synthesized compounds, as well as their physicochemical and pharmacokinetic properties. Results: Copper(II) complex C2 exhibited antitumor properties towards MDA-MB-231 compared with Cisplatin (cancer cell viability rate of 23.6% vs. 22.5%). At a concentration of 200 μg/mL, complexes C1 and C2 were less cytotoxic than the reference Cisplatin against a normal, non-cancerous skin fibroblast cell line (HDFa). According to in vitro tests, C2 reduced the viability of HeLa, MCF-7, and MDA-MB-231 cells by about 57.5-81.2%. It was evident that all compounds were devoid of antibacterial or antifungal activity. In vitro assays revealed that a moderate antiradical effect was observed for free ligand L1 containing phthalazin-1(2H)-imine in the ABTS radical scavenging assay (IC50 = 23.63 µg/mL). Conclusions: The anticancer studies revealed that the most potent compound was copper(II) complex C2 bearing a phthalazin-1(2H)-one scaffold. None of the tested compounds showed antimicrobial or antifungal activity. This feature seems to be beneficial in terms of their potential uses as anticancer agents in the future. In vitro antiradical assays revealed that a moderate antioxidant effect was observed only for free ligand L1 containing phthalazin-1(2H)-imine.
Collapse
Affiliation(s)
- Łukasz Balewski
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Iwona Inkielewicz-Stępniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gen. Dębinki 7, 80-211 Gdańsk, Poland
| | - Maria Gdaniec
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Katarzyna Turecka
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Anna Hering
- Department of Biology and Pharmaceutical Botany, Faculty of Pharmacy, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Anna Ordyszewska
- Department of Inorganic Chemistry, Faculty of Chemistry and Advanced Materials Centers, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Anita Kornicka
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| |
Collapse
|
46
|
Srikanth Y, Reddy DH, Anusha VL, Dumala N, Viswanadh MK, Chakravarthi G, Nalluri BN, Yadagiri G, Ramakrishna K. Unveiling the Multifaceted Pharmacological Actions of Indole-3-Carbinol and Diindolylmethane: A Comprehensive Review. PLANTS (BASEL, SWITZERLAND) 2025; 14:827. [PMID: 40094833 PMCID: PMC11902694 DOI: 10.3390/plants14050827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Cruciferae family vegetables are remarkably high in phytochemicals such as Indole-3-carbinol (I3C) and Diindolylmethane (DIM), which are widely known as nutritional supplements. I3C and DIM have been studied extensively in different types of cancers like breast, prostate, endometrial, colorectal, gallbladder, hepatic, and cervical, as well as cancers in other tissues. In this review, we summarized the protective effects of I3C and DIM against cardiovascular, neurological, reproductive, metabolic, bone, respiratory, liver, and immune diseases, infections, and drug- and radiation-induced toxicities. Experimental evidence suggests that I3C and DIM offer protection due to their antioxidant, anti-inflammatory, antiapoptotic, immunomodulatory, and xenobiotic properties. Apart from the beneficial effects, the present review also discusses the possible toxicities of I3C and DIM that are reported in various preclinical investigations. So far, most of the reports about I3C and DIM protective effects against various diseases are only from preclinical studies; this emphasizes the dire need for large-scale clinical trials on these phytochemicals against human diseases. Further, in-depth research is required to improve the bioavailability of these two phytochemicals to achieve the desirable protective effects. Overall, our review emphasizes that I3C and DIM may become potential drug candidates for combating dreadful human diseases.
Collapse
Affiliation(s)
- Yadava Srikanth
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Dontiboina Harikrishna Reddy
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Vinjavarapu Lakshmi Anusha
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Naresh Dumala
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Matte Kasi Viswanadh
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Guntupalli Chakravarthi
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Buchi N. Nalluri
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| | - Ganesh Yadagiri
- Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kakarla Ramakrishna
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522302, India; (Y.S.); (D.H.R.); (V.L.A.); (N.D.); (M.K.V.); (G.C.); (B.N.N.)
| |
Collapse
|
47
|
Manickam V, Zallocchi M. Paving the way for better ototoxicity assessments in cisplatin therapy using more reliable animal models. Front Cell Neurosci 2025; 19:1552051. [PMID: 40109278 PMCID: PMC11919870 DOI: 10.3389/fncel.2025.1552051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025] Open
Abstract
Cisplatin-induced hearing loss is a common and irreversible side effect affecting a significant proportion of cancer patients. While various strategies to mitigate this toxicity have been explored, there remains a critical need for effective treatments. A major challenge in developing new therapies is the lack of reliable animal models that accurately replicate the clinical use of cisplatin in humans, which typically involves multiple cycles of low-dose administration. Traditional models using high doses of cisplatin have resulted in high mortality and variable hearing loss, complicating the assessment of potential treatments. To address this, a multi-cycle model using lower cisplatin doses in mice was developed, providing hearing loss without mortality. However, variability in outcomes across different research groups persisted. In the present study, we optimize the multi-cycle model of cisplatin-induced ototoxicity by using clinical-grade cisplatin rather than laboratory-grade formulations. The use of clinical cisplatin ensures greater consistency, reliability, and relevance to human treatment protocols, as it adheres to the rigorous quality standards required for patient use. This new administration protocol will minimize variability across research laboratories and more accurately mimic the dosing regimens typically administered to cancer patients. Additionally, we have enhanced a zebrafish model for high-throughput screening of potential therapeutics, further improving the consistency of results. These improvements to the animal models are critical for accelerating the discovery and testing of therapies to prevent cisplatin-induced hearing loss, supporting the development of effective treatments for cancer patients undergoing cisplatin chemotherapy.
Collapse
Affiliation(s)
- Vijayprakash Manickam
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Marisa Zallocchi
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| |
Collapse
|
48
|
Wang C, Chen Z, Ni W, Wang J, Zhou W. Research and progress of microRNA-136 in metastatic tumors. Front Oncol 2025; 15:1555270. [PMID: 40104500 PMCID: PMC11913677 DOI: 10.3389/fonc.2025.1555270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Background MiR-136 is abnormally expressed in many types of metastatic tumors and is closely associated with tumor cell proliferation, apoptosis, invasion, and metastasis, indicating its important role in tumor development and progression. This review summarizes current knowledge regarding miR-136's molecular mechanisms, functional roles, and impact on chemotherapy in different human cancers. Methods A literature search was conducted in PubMed and Web of Science using "miR-136" and "metastatic tumors" as English keywords, and in CNKI and Wanfang databases using the same terms in Chinese. Studies related to miR-136 research in metastatic tumors and high-quality evidence from similar studies were included. Meta-analyses, dissertations, conference papers, low-quality articles, unavailable full-text articles, and republished articles were excluded. Results This review synthesizes the current understanding of miR-136's role in various cancers, including osteosarcoma, gastric cancer, gallbladder cancer, esophageal cancer, prostate cancer, colorectal cancer, breast cancer, glioma, and thyroid cancer. miR-136 acts as a tumor suppressor by targeting various genes, including MTDH, PTEN, MAP2K4, MUC1, LRH-1, MIEN1, RASAL2, CYR61, and KLF7. It influences multiple signaling pathways, including the ERK/mitogen-activated protein kinase, Wnt/β-catenin, Ha-Ras, PI3K/Akt, Aurora-A kinase, nuclear factor-κB, and JNK pathways. Furthermore, miR-136 is involved in chemoresistance by modulating ROCK1, PPP2R2A, and the miR-136-Notch3 signaling axis. Conclusions MiR-136 demonstrates promising potential as a novel biomarker and therapeutic target in various human cancers. Further research is needed to fully elucidate its complex roles in cancer development, progression, and drug resistance, particularly regarding its potential in immunotherapy.
Collapse
Affiliation(s)
- Chenwen Wang
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zixiong Chen
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Ni
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jiang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Zhou
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
49
|
Wei C, Zhang X, Li H, Gu J, Xue F, Xie W, Ji G. STMN1 regulates the stemness of gastric cancer cells by binding to HN1L to activate the STAT3 signaling pathway. Discov Oncol 2025; 16:263. [PMID: 40035993 PMCID: PMC11880488 DOI: 10.1007/s12672-025-01879-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND STMN1 is highly expressed in gastric cancer (GC) tissues and the aim of this study was to investigate the role of STMN1 in GC cell stemness. METHODS Analysis of the expression and correlation of STMN1 and its target genes in GC through bioinformatics. Construction of interference plasmids for STMN1 and transfection into GC cells. Sphere formation assay was conducted to detect stem cell sphere-forming ability. WB analysis was performed to detect the expression of stemness genes CD133, ALDH1, CD44, SOX2, Nanog, and STAT3-related proteins. Additionally, CCK-8 assay and TUNEL staining were used to assess GC cell sensitivity to cisplatin (DDP). Construction of a xenograft mouse model to detect the in vivo tumorigenic ability of GC cells. Immunoprecipitation (IP) experiment was conducted to validate the binding of STMN1 and HN1L in GC cells. Overexpression plasmids of HN1L were used for mechanism validation. RESULTS STMN1 and its target HN1L were highly expressed in GC tissues and cells, and were associated with a poor prognosis in GC. Interfering with STMN1 significantly reduced the self-renewal ability of GC cells, downregulated the expression of CD133, ALDH1, CD44, SOX2, Nanog, p-STAT3 and PD-L1. Interfering with STMN1 increased the sensitivity of GC cells to DDP and promoted apoptosis. IP experiments demonstrate that STMN1 and HN1L combine in GC cells. Overexpression of HN1L significantly reversed the effects of Si-STMN1 on GC cells. In vivo experiments demonstrate that the addition of DDP or interference with STMN1 reduced tumor size and weight, and downregulated the expression of CD133, KI67, HN1L, p-STAT3, and PD-L1 in tumor tissues. The combined use of DPP and Si-STMN1 had a more significant effect. CONCLUSION STMN1 regulates GC cell stemness by binding HN1L to activate the HN1L/STAT3/ PD-L1 signaling pathway.
Collapse
Affiliation(s)
- Chunyang Wei
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Xing Zhang
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Hao Li
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Jianzhong Gu
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Fei Xue
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Wenna Xie
- Department of Gastrointestinal Surgery, The First People's Hospital of Yinchuan, Ningxia, 750001, China
| | - Gang Ji
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Me dical University, Xi'an, 710032, China.
| |
Collapse
|
50
|
Zhong X, Zhang Y, Wei J. Recent advances in ruthenium (III) complex-loaded nanomaterial for enhanced cancer therapy efficacy. Drug Dev Ind Pharm 2025; 51:169-179. [PMID: 39836522 DOI: 10.1080/03639045.2025.2455428] [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: 09/20/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Amid the escalating global cancer incidence, the development of effective and safe anticancer drugs is a critical priority in medical research. Addressing the clinical shortcomings of ruthenium-based anticancer drugs are currently a prominent focus of research. SIGNIFICANCE AND METHODS Since the pioneering work with platinum derivatives, significant progress has been made in the fundamental studies of metal complexes for the treatment of a wide range of cancers, and there has been a growing interest in their properties and biomedical applications. Although chemotherapy is crucial in clinical tumor management, platinum(II) anticancer drugs like cisplatin and carboplatin suffer from severe toxicity and drug resistance issues. Among various metal-based drugs, ruthenium(III) complexes are notable for their selectivity, cytotoxic activity in vitro, and effective anticancer properties in vivo. Despite some drug candidates reaching late-stage clinical trials, their clinical application remains constrained by problems such as low solubility, poor stability, and inadequate cellular uptake. RESULTS The development of efficient and stable nanocarrier-based drug delivery systems for ruthenium(III) complexes, enhancing pharmacokinetic properties, and enabling slow, controlled release and targeted drug delivery, offers potential solutions to these limitations. CONCLUSIONS This review delves into the recent strides in nanomaterial-based drug delivery for ruthenium complexes, encompassing research on platinum (II) and ruthenium (III) metal complexes, nano-delivery system designs, and addresses pivotal challenges and future trajectories in this domain.
Collapse
Affiliation(s)
- Xuemei Zhong
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, China
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, China
| | - Jianhua Wei
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, China
| |
Collapse
|