1
|
El-Wakil MH, Ghazala RA, El-Dershaby HA, Drozdowska D, Wróbel-Tałałaj A, Parzych C, Ratkiewicz A, Kolesińska B, Abd El-Razik HA, Soliman FSG. Rational design, synthesis, and molecular modelling insights of dual DNA binders/DHFR inhibitors bearing arylidene-hydrazinyl-1,3-thiazole scaffold with apoptotic and anti-migratory potential in breast MCF-7 cancer cells. J Enzyme Inhib Med Chem 2025; 40:2468353. [PMID: 40035286 PMCID: PMC11881662 DOI: 10.1080/14756366.2025.2468353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
In light of searching for new breast cancer therapies, DNA-targeted small molecules were rationally designed to simultaneously bind DNA and inhibit human dihydrofolate reductase (hDHFR). Fourteen new arylidene-hydrazinyl-1,3-thiazoles (5-18) were synthesised and their dual DNA groove binding potential and in vitro hDHFR inhibition were performed. Two compounds, 5 and 11, proved their dual efficacy. Molecular docking and molecular dynamics simulations were performed for those active derivatives to explore their mode of binding and stability of interactions inside DHFR active site. Anti-breast cancer activity was assessed for 5 and 11 on MCF-7 cells using MTX as reference. IC50 measurements revealed that both compounds were more potent and selective than MTX. Cytotoxicity was examined against normal skin fibroblasts to examine safety and selectivity Moreover, mechanistic studies including apoptosis induction and wound healing were performed. Further in silico ADMET assessment was conducted to determine their eligibility as drug leads suitable for future optimisation and development.
Collapse
Affiliation(s)
- Marwa H. El-Wakil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Rasha A. Ghazala
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hadeel A. El-Dershaby
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Danuta Drozdowska
- Department of Organic Chemistry, Medical University of Bialystok, Bialystok, Poland
| | | | - Cezary Parzych
- Department of Physical Chemistry, University of Bialystok, Institute of Chemistry, Bialystok, Poland
| | - Artur Ratkiewicz
- Department of Physical Chemistry, University of Bialystok, Institute of Chemistry, Bialystok, Poland
| | - Beata Kolesińska
- Institute of Organic Chemistry, Lodz University of Technology, Lodz, Poland
| | - Heba A. Abd El-Razik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Farid S. G. Soliman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
Xu J, Wang Y, Zhang J, Tang J, Zhou Z. The role of branched-chain amino acids in cardio-oncology: A review. Life Sci 2025; 372:123614. [PMID: 40189196 DOI: 10.1016/j.lfs.2025.123614] [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: 02/04/2025] [Revised: 03/18/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
Cancer and cardiovascular diseases (CVDs) are global health challenges. In cancer patients, CVD is the second leading cause of death following disease progression. There are few specialized services for cardio-oncology patients worldwide currently. Branched-chain amino acids (BCAAs) are essential amino acids that promote protein synthesis and energy homeostasis. The disruption of BCAAs metabolism facilitates the development of cancer and CVDs while the benefit of BCAA supplement is full of controversy. In this review, we summarized BCAA-related studies in cardiometabolism, cancer and chemotherapy-induced cardiotoxicity, and provided our perspectives on the roles of BCAAs in cardio-oncology. We find that supplementation of BCAAs presents protective effects in cardiometabolic diseases, while the influence on cancer is intricate and varies across different types of cancers. Large-scale clinical studies are needed to understand the long-term effects of BCAA intake and its impact on different stages of the disease. BCAAs have potential to mitigate chemotherapy-induced cardiotoxicity. Continued research is still essential to understand the precise mechanisms, determine optimal dosage and timing, and assess the effectiveness of BCAA supplement in cardio-oncology, in particular clinical research.
Collapse
Affiliation(s)
- Jiaqi Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Cardiology, The First Hospital of Hebei Medical University, Hebei, China
| | - Jing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Jingyi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhongyan Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|
3
|
Ur Rahman M, Hussain HR, Akram H, Sarfraz M, Nouman M, Khan JA, Ishtiaq M. Niosomes as a targeted drug delivery system in the treatment of breast cancer: preparation, classification and mechanisms of cellular uptake. J Drug Target 2025; 33:916-932. [PMID: 39964023 DOI: 10.1080/1061186x.2025.2468750] [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: 09/27/2024] [Revised: 01/24/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
Breast cancer (BC) remains one of the significant health issues across the globe, being diagnosed in millions of women worldwide annually. Conventional therapeutic options have substantial adverse effects due to their non-specificity and limited drug bioavailability. Niosomes, being novel drug delivery systems formed from non-ionic surfactants, with or without cholesterol and charge-inducing agents, are used as therapeutic options in treating BC. Their formulation by various methods enhances the therapeutic efficacy and bioavailability and minimises side effects. Niosomal formulation of tamoxifen exhibits target drug delivery with enhanced stability, whereas docetaxel and methotrexate show sustained and controlled drug release, respectively. 5-Fluorouracil, doxorubicin, paclitaxel, cyclophosphamide and epirubicin show improved cytotoxic effects against BC when combined with other agents. Furthermore, repurposed niosomal formulations of anti-cancer drugs show improved penetration, reduced tumour volume and significantly enhanced anti-tumour effect. This review article focuses on the composition of niosomes and their application in BC treatment and then examines how niosomes could contribute to BC research.
Collapse
Affiliation(s)
| | | | - Habiba Akram
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Sarfraz
- College of Pharmacy, Al-Ain University, Al-Ain, United Arab Emirates
| | - Muhammad Nouman
- College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Jawad Akbar Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore, Pakistan
| | - Memona Ishtiaq
- Department of Pharmacy, Lahore Institute of Professional Studies, Lahore, Pakistan
| |
Collapse
|
4
|
Li Z, Yang X, Wang S, Ma H, Yang K, Shi J, Wang X. Design, synthesis, and biological evaluation of hypoxic-activation prodrug TH-302 derivatives. Bioorg Med Chem Lett 2025; 122:130189. [PMID: 40107631 DOI: 10.1016/j.bmcl.2025.130189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/26/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
This study aims to design and develop novel and efficient anti-hypoxic cell tumor drugs. Using the TH-302 as lead compound, structural modifications are conducted to synthesize a series of novel derivatives to investigate the structural activity relationship (SAR) against ovarian cancer cell line (SKOV3) and glioblastoma cell line (U87MG) in vitro. The structural modifications mainly include four aspects: changes in substituents on N; changes in isomers; changes in nitro group position; changes in substituting halogens in phosphoramide mustard. The results of CCK-8 assay indicate that the steric hindrance and electronic effects of substituents on N have significant impacts on the activity, while changes in nitro group positions have minimal effects on the activity, and Bromo-phosphoramide mustard exhibits better activity than Chloro-phosphoramide mustard. Compounds 15c and 16d exhibit significantly superior antitumor activity compared to TH-302, with IC50 values of 42 μM and 32 μM for SKOV3 cells, and IC50 values of 47 μM and 41 μM for U87MG cells, respectively.
Collapse
Affiliation(s)
- Zhengyi Li
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Petrochemical Engineering, Changzhou University, Changzhou, Jiangsu 213164, China.
| | - Xingchen Yang
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Petrochemical Engineering, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Shun Wang
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Petrochemical Engineering, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Hongzhao Ma
- Jiangsu Huajing Molecular Imaging and Pharmaceutical Research Institute Co. Ltd., Changshu, Jiangsu 215500, China
| | - Ke Yang
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Petrochemical Engineering, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Jing Shi
- Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology, School of Petrochemical Engineering, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Xin Wang
- Jiangnan University Medical Center, JUMC, Wuxi, Jiangsu 214002, China
| |
Collapse
|
5
|
Tao Q, Cai T, Xiao Y, Han T, Shen L, Cheng C, Xu S, Li A, Zhang P, Chen J, Zhang Y, Tong Q, Cai X. Genome-guided discovery of coublibactins from Nocardia coubleae and their gallium complexes with potent antileukemic activity. Bioorg Chem 2025; 160:108508. [PMID: 40280014 DOI: 10.1016/j.bioorg.2025.108508] [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: 03/08/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
The pursuit of highly effective and selective anticancer drugs remains a critical challenge. Metal-based complexes, particularly gallium-containing compounds, offer promising therapeutic avenues due to their unique mechanisms of action. To identify novel scaffolds for such complexes, we performed a comprehensive genomic analysis of Nocardia species, revealing the prevalence of siderophore biosynthetic gene clusters, including the highly conserved nocobactin NA-like clusters. From N. coubleae DSM 44960, we isolated three new siderophores, coublibactins A-C (1-3), along with eight congeners (4-11) with known planar structures, all characterized by exceptional iron-binding affinity. Subsequent gallium substitution yielded gallium complexes (Ga-1-11). Among these, Ga-6 exhibited significant anticancer activity against human acute promyelocytic leukemia NB4 cells with IC50 value of 1.35 μM. Pharmacological studies showed that Ga-6 induces cell cycle arrest and apoptosis in NB4 cells. Our findings revealed microbial siderophores as promising scaffolds for the design of next-generation metal-based anticancer therapeutics, particularly gallium-based agents.
Collapse
Affiliation(s)
- Qiaoqiao Tao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Teng Cai
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yang Xiao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Tao Han
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Ling Shen
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Chang Cheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Shouying Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| | - Aiying Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| | - Peng Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Jiachun Chen
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Qingyi Tong
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| | - Xiaofeng Cai
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Key Laboratory of Neurological Diseases of Hubei Province, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
6
|
Chen S, Tian S, Wang Y, Li M, Tang D. Harnessing bifunctional nanozyme with potent catalytic and signal amplification for innovating electrochemical immunoassay. Biosens Bioelectron 2025; 278:117340. [PMID: 40064571 DOI: 10.1016/j.bios.2025.117340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025]
Abstract
Nanozyme-based electrochemical biosensors have emerged as an alternative to enzyme-based biosensors for next-generation bioanalysis. However, potential antibody modifications limit the catalytic sites of the nanozyme, thereby reducing sensor sensitivity. Here, a sensitive method for determining carcinoembryonic antigen (CEA) was developed. It involved coupling a cascade enzyme - enzyme - like catalytic reaction using Fe - Co Prussian blue analog nanozymes with high peroxidase - like activity (79.42 U mg-1). Briefly, the transduction of biological signals to chemical signals was achieved through the strategy centered on catalytic electroactive probes. Thereafter, with the assistance of the microelectrochemical workstation, the output of signals was realized. The platform exhibited an ultra-wide range of 0.020-100 ng mL-1 and a detection limit of 0.013 ng mL-1 CEA, which was mainly attributed to the excellent peroxidase activity, good conductivity, and synergistic amplification of current signals of synthesized nanozymes. In addition, the modification-free features greatly reduced the complexity of the bioassay and significantly improves its portability and cost-effectiveness. Overall, this study advances the development of nanozymes and their electrochemical biosensing applications and is expected to extend to the development of miniaturized devices in direct detection environments.
Collapse
Affiliation(s)
- Shuyun Chen
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China
| | - Shuo Tian
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China
| | - Yunsen Wang
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China
| | - Meijin Li
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China.
| | - Dianping Tang
- Key Laboratory for Analytical Science of Food Safety and Biology (MOE & Fujian Province), Department of Chemistry, Fuzhou University, Fuzhou, 350108, PR China.
| |
Collapse
|
7
|
Javaid D, Ganie SY, Qadri SS, Reyaz A, Reshi MS. Eco-friendly nanotherapeutics: Metallic nanoparticles for targeting breast cancer. Eur J Pharmacol 2025; 996:177603. [PMID: 40189083 DOI: 10.1016/j.ejphar.2025.177603] [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: 01/08/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/13/2025]
Abstract
Breast cancer continues to be a major cause of death among women globally, with triple-negative breast cancer (TNBC) presenting a particularly difficult challenge due to its aggressive behaviour and the lack of effective treatment options. Nanotechnology, particularly the use of silver nanoparticles (AgNPs), has emerged as a promising avenue in oncological research. This review explores into the escalating field of green synthesis of nanoparticles, emphasizing sustainable approaches utilizing plant-based resources. Critical factors influencing nanoparticle synthesis, including reaction conditions, precursor types, and plant phytochemicals, are explored alongside advanced characterization techniques essential for evaluating nanoparticle properties. Special focus is given to the phytofabrication of silver nanoparticles and their multifaceted roles in breast cancer treatment, with detailed insights into their mechanisms, such as inducing apoptosis, generating reactive oxygen species (ROS), and disrupting mitochondrial function, particularly in TNBC cells. The review further highlights the advantages of plant-derived AgNPs, such as biocompatibility and reduced toxicity, while addressing challenges like scalability, reproducibility, and regulatory hurdles. Concluding with future prospects, this paper reflects the potential of green-synthesized AgNPs as a keystone in next-generation cancer therapeutics, paving the way for innovative and eco-friendly approaches in oncology.
Collapse
Affiliation(s)
- Darakhshan Javaid
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Shahid Yousuf Ganie
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Syed Sanober Qadri
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Adfar Reyaz
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Mohd Salim Reshi
- Toxicology and Pharmacology Laboratory, Department of Zoology, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India.
| |
Collapse
|
8
|
Abdel-Maksoud MS, Alatawi RA, Albalawi SSA, Alrashidi MN, Abo-Dya NE, Elsherbiny N, Ragab YM, Awaji AA, El-Sherbiny M, Elfadil H, Abd-Alhaseeb MM. Diacerein's antiproliferative effects alone and with 5-fluorouracil in an Ehrlich solid tumour model: Molecular docking, molecular dynamics Simulation studies, and experimental Verification. Eur J Pharmacol 2025; 996:177564. [PMID: 40157706 DOI: 10.1016/j.ejphar.2025.177564] [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: 12/15/2024] [Revised: 03/16/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
The current study used an experimental model of mammary gland carcinoma to assess the chemo-sensitizing effectiveness of the combined administration of diacerein and 5-Fluorouracil (5-FU). With docking scores of -8.1, -7.6, and -9.2 kcal/mol, respectively, the molecular docking experiments showed that diacerein exhibits significant binding affinities to Caspase-3, NF-κB, and AKT1. Molecular dynamics Simulations revealed that diacerein has favourable binding free energy (ΔGbind) of -26.7 kcal/mol for Caspase-3, -24.2 kcal/mol for NF-κB, and -39.9 kcal/mol for AKT1, combined with low root mean square deviation (RMSD) values of 3.1 Å, 1.6 Å, and 2.1 Å for the three targets respectively. To validate these findings in vivo, Ehrlich solid tumor (EST) was induced in female Swiss mice. Four groups of animals were randomly assigned: EST + vehicle, EST + 5-FU, EST + diacerein, and EST + combination. Diacerein and 5-FU combination treatment increased EST mice's life span and reduced the solid tumor's weight and volume. Furthermore, diacerein and 5-FU combination significantly suppressed oxidative stress, inhibited AKT phosphorylation, decreased downstream inflammation (NF-κB, TNF-α, IL-1β), and increased apoptosis by modulating Bax, Bcl2, P53, and caspase-3 levels in tumor tissues. In conclusion, by inhibiting the AKT/NF-κB axis, diacerein and 5-FU combination showed possible antiproliferative effectiveness in the EST model.
Collapse
Affiliation(s)
- Mohamed S Abdel-Maksoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | | | | | | | - Nader E Abo-Dya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia.
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia.
| | - Yasser M Ragab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, North Sinai, Egypt; Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Aeshah A Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, University of Tabuk, 71491, Saudi Arabia.
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia.
| | - Hassabelrasoul Elfadil
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohammad M Abd-Alhaseeb
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt; Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
9
|
Sailo BL, Garhwal A, Mishra A, Hegde M, Vishwa R, Girisa S, Abbas M, Alqahtani MS, Abdulhammed A, Sethi G, Kempson I, Kunnumakkara AB. Potential of capsaicin as a combinatorial agent to overcome chemoresistance and to improve outcomes of cancer therapy. Biochem Pharmacol 2025; 236:116828. [PMID: 40023449 DOI: 10.1016/j.bcp.2025.116828] [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: 11/22/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Capsaicin (CAPS), a bioactive alkaloid derived from chili peppers, has garnered significant interest for its potential role as a combinatorial and chemosensitizing agent in cancer therapy. Numerous preclinical studies have demonstrated that CAPS enhanced the efficacy of various anticancer agents by promoting apoptosis, modulating autophagy and inhibiting angiogenesis, tumor growth, and metastasis. Additionally, CAPS modulated critical regulators of chemoresistance, such as P-glycoprotein (P-gp), extracellular signal-regulated kinase (ERK), nuclear factor-kappa B (NF-κB) pathway, and signal transducer and activator of transcription 3 (STAT3) pathway, thereby contributing to the reversal of multidrug resistance (MDR). Moreover, when administered in combination with chemotherapeutic agents, CAPS has been shown to improve treatment efficacy at lower drug concentrations. Given its multitargeted mechanism of action, CAPS represents a promising adjunct to conventional cancer therapies. However, due to its lipophilic nature, the development of optimized formulation strategies is essential to enhance its bioavailability and ensure consistent therapeutic outcomes. In conclusion, CAPS holds significant potential as a combinatorial and chemosensitizing agent, helping to overcome chemoresistance and enhance treatment outcomes across various malignancies. These promising findings warrant further preclinical and clinical investigations.
Collapse
Affiliation(s)
- Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Anushka Garhwal
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Anamika Mishra
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421 Abha, Saudi Arabia
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421 Abha, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Ayman Abdulhammed
- Department of Biochemistry and Hormone, King Fahad Central Hospital, Gizan 82666, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117699, Singapore.
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam 781039, India.
| |
Collapse
|
10
|
Essa AF, Elghonemy MM, Taher RF, Allam RM, Elshamy AI. Undescribed diterpenes from Euphorbia mauritanica L. as modulators of the breast cancer resistance: Mechanistic and in silico studies. PHYTOCHEMISTRY 2025; 234:114418. [PMID: 39889865 DOI: 10.1016/j.phytochem.2025.114418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
As part of efforts to identify natural modulators of multi-drug-resistant breast cancer, Euphorbia mauritanica L. chloroform extract yielded four undescribed oxygenated diterpenes, including three nor-ent-abietanes, euphomauritanol C-E (1-3), and one polyacylated jatrophane, euphomauritanolide A (4), along with two knowns, helioscopinolide A (5) and enukokurin (6). The chemical structures and configurations of compounds were established by combination of HRMS, FTIR, and NMR spectroscopic tools along with experimental and calculated TDDFT-ECD. The cytotoxicity evaluation of isolated compounds against the MCF-7ADR revealed 4 and 2 are the most potent with IC50 values of 3.2 ± 0.58 and 4.67 ± 0.29 μM, respectively. Co-administration of compounds 4 and 2 with DOX improved its cytotoxic effect, with a combination index value of 0.41 for 4, indicating a synergistic effect. Mechanistically, 4 modulated DOX anticancer properties via potentiating DOX-induced Go/G1 cell cycle arrest rather than G2M arrest of DOX alone and shifting the cell death of DOX to be mainly apoptotic cell death. Furthermore, 4 alone and combined with DOX showed promising anti-migratory effects against MCF-7ADR. In conclusion, 4 showed promising co-chemotherapeutic effects to the DOX against MCF-7ADR, indicating that this compound possesses potential as an auspicious lead chemical to target breast cancer cells resistant to doxorubicin.
Collapse
Affiliation(s)
- Ahmed F Essa
- Chemistry of Natural Compounds Department, National Research Centre, 33 El Bohouth St, Dokki, Giza, 12622, Egypt
| | - Mai M Elghonemy
- Chemistry of Natural Compounds Department, National Research Centre, 33 El Bohouth St, Dokki, Giza, 12622, Egypt
| | - Rehab F Taher
- Chemistry of Natural Compounds Department, National Research Centre, 33 El Bohouth St, Dokki, Giza, 12622, Egypt
| | - Rasha M Allam
- Pharmacology Department, Medical and Clinical Research Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Abdelsamed I Elshamy
- Chemistry of Natural Compounds Department, National Research Centre, 33 El Bohouth St, Dokki, Giza, 12622, Egypt; Academy of Scientific Research and Technology (ASRT), 101 Kasr El Ainy St. Kasr El Ainy, 11516, Cairo, Egypt.
| |
Collapse
|
11
|
Xie K, Wang J, Jiang J, Deng Z, Hu Q, Wang D. Efficacy and safety outcomes of emerging EGFR‑TKIs for patients with non‑small cell lung cancer with EGFR exon 20 insertion mutations: A systematic review and meta‑analysis. Oncol Lett 2025; 29:316. [PMID: 40337604 PMCID: PMC12056541 DOI: 10.3892/ol.2025.15062] [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: 05/08/2024] [Accepted: 03/28/2025] [Indexed: 05/09/2025] Open
Abstract
Lung cancer remains a leading cause of mortality worldwide, with non-small cell lung cancer (NSCLC) accounting for ~85% of all lung cancer cases. Epidermal growth factor receptor (EGFR) exon 20 insertion mutant NSCLC is rare and associated with poor outcomes. Several novel generations (third-generation) of EGFR-tyrosine kinase inhibitors (TKIs) have been developed for the treatment of NSCLC and have shown antitumour potential. Therefore, the present study reviewed their efficacy and safety outcomes for this condition. A thorough literature searching was performed using the Cochrane Library, Web of Science, PubMed and Embase databases. Clinical trials published in English and reporting overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS) and treatment relevant adverse events (TRAEs) of grade ≥3 were included for further analysis. A total of 13 studies were included. All included studies reported ORRs with a pooled ORR of 0.486 [95% confidence interval (CI), 0.369-0.602]. Subgroup analysis revealed the following ORRs: 0.731 (95% CI, 0.560-0.901; I2=0%) for YK-029A; 0.608 (95% CI, 0.511-0.705; I2=0%) for sunvozertinib; 0.602 (95% CI, 0.440-0.764; I2=80.2%) for furmonertinib; 0.602 (95% CI, 0.486-0.718; I2=84.5%) for befotertinib; 0.566 (95% CI, 0.236-0.896; I2=96.3%) for amivantamab; 0.444 (95% CI, 0.215-0.674; I2=0%) for BEBT-109; and 0.256 (95% CI, 0.178-0.334; I2=75.0%) for poziotinib. The pooled DCR, median PFS and median OS were 0.843 (95% CI, 0.740-0.946), 10.11 months (95% CI, 9.58-10.64 months; I2=78.8%; P<0.001) and 23.00 months (95% CI, 20.30-25.69 months; I2=44.8; P=0.178), respectively. The pooled incidence of TRAEs of grade ≥3 was 0.458 (95% CI, 0.336-0.580; I2=96.9%; P<0.001), with the incidence of the three most reported TRAEs (diarrhoea, thrombocytopenia and anaemia) demonstrated to be 0.112 (95% CI, 0.060-0.164), 0.065 (95% CI, -0.012-0.141) and 0.040 (95% CI, 0.005-0.076), respectively. In conclusion, the emerging EGFR-TKIs for NSCLC with EGFR exon 20 insertion have a promising treatment outcome with a manageable safety profile. However, further analysis is needed when more clinical data are released.
Collapse
Affiliation(s)
- Kang Xie
- Precision Medicine Centre, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jing Wang
- Precision Medicine Centre, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Juan Jiang
- Precision Medicine Centre, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhujun Deng
- Precision Medicine Centre, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qiongxia Hu
- Precision Medicine Centre, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Denian Wang
- Precision Medicine Centre, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
12
|
Nerella SG, Shaik MG, Singh P, Arifuddin M, Ullah Q, Supuran CT. Antibody-drug conjugates and radioconjugates targeting carbonic anhydrase IX and XII in hypoxic tumors: Bench to clinical applications. Bioorg Chem 2025; 159:108408. [PMID: 40154235 DOI: 10.1016/j.bioorg.2025.108408] [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: 01/23/2025] [Revised: 03/10/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
Treating hypoxic tumors is challenging due to their aggressive nature, resistance to standard treatments, often leading to poor outcomes. Hypoxic tumors create a unique environment that reduces the effectiveness of traditional treatments such as chemotherapy and radiotherapy. Human carbonic anhydrases (hCA IX and hCA XII) are involved in tumors survival and metabolism by regulating pH homeostasis, ferroptosis, metastatization, and other processes. Developing drugs that specifically target these enzymes has been demonstrated to disrupt the tumor survival mechanisms, leading to significant antitumor effects. This review discusses recent developments on antibody-drug conjugates (ADCs) and radioconjugates targeting hCA IX and hCA XII in hypoxic tumors. New approaches based on small molecule inhibitors and monoclonal antibodies such as girentuximab provided encouraging results in preclinical research and clinical trials. These advances highlight the potential of hCA-targeted therapies to improve cancer treatment for hypoxic tumors.
Collapse
Affiliation(s)
- Sridhar Goud Nerella
- Department of Neuroimaging and Interventional Radiology (NI & IR), National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru 560 029, India; Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health (NIH), Bethesda, MD-20892, USA.
| | - Mahammad Ghouse Shaik
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045, USA
| | - Priti Singh
- Department of Chemistry and the Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, 30324, USA
| | - Mohammed Arifuddin
- Department of Chemistry, Centre for Distance and Online Education, Maulana Azad National Urdu University, Hyderabad, 500032, India
| | - Qasim Ullah
- Physical Science Section, School of Sciences, Maulana Azad National Urdu University, Hyderabad, 500032, India
| | - Claudiu T Supuran
- Università degli Studi di Firenze, Neurofarba Dept., Sezione di Scienze Farmaceutiche e Nutraceutiche, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
13
|
Teng Y, Wang D, Yang Z, Wang R, Ning S, Zhang R, Yang H, Feng X, Liu J, Yang L, Tian Y. Bioorthogonal strategy-triggered In situ co-activation of aggregation-induced emission photosensitizers and chemotherapeutic prodrugs for boosting synergistic chemo-photodynamic-immunotherapy. Biomaterials 2025; 317:123092. [PMID: 39793168 DOI: 10.1016/j.biomaterials.2025.123092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/04/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
In situ activation of prodrugs or photosensitizers is a promising strategy for specifically killing tumor cells while avoiding toxic side effects. Herein, we originally develop a bioorthogonally activatable prodrug and pro-photosensitizer system to synchronously yield an aggregation-induced emission (AIE) photosensitizer and a chemotherapeutic drug for synergistic chemo-photodynamic-immunotherapy of tumors. By employing molecular engineering strategy, we rationally design a family of tetrazine-functionalized tetraphenylene-based photosensitizers, one of which (named TzPS5) exhibits a high turn-on ratio, a NIR emission, a typical AIE character, and an excellent ROS generation efficiency upon bioorthogonal-activation. With the aid of integrin- or mitochondria-pretargeting, TzPS5 is successfully applied for highly effective PDT ablation of cancer cells both in vitro and in vivo. On this basis, tumor-targeting TzPS5 (TzPS5-cRGD) is constructed and used jointly with a bioorthogonal prodrug, DOX-TCO, and the two are mutually activated to induce cooperative and tumor-specific PDT and chemotherapy, resulting in amplified therapeutic outcomes and improved biosafeties. Moreover, this combination modality elicits robust immunogenic cell death, stimulates systemic antitumor immunity, thereby suppressing both primary and distant tumors, and blocking the pulmonary tumor metastasis. This work is expected to provide a useful guidance for the rational design of activatable phototheranostic agents, and offer a new strategy for co-activation of prodrugs/pro-photosensitizers to boost synergistic antitumor chemo-photodynamic-immunotherapy.
Collapse
Affiliation(s)
- Yu Teng
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Dianyu Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Ziyu Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Ruxuan Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100007, PR China
| | - Shuyi Ning
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Rongrong Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Hong Yang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Xinchi Feng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China.
| | - Lijun Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China.
| | - Yulin Tian
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
14
|
Celesti C, Mele A, Espro C, Raffaini G, Laganà A, Visalli G, Giofrè SV, Gaetano FD, Neri G, Caronna T, Iannazzo D. A smart β-Cyclodextrin-Aza[5]Helicene system for enhanced gemcitabine delivery and tracking in cancer cells. Int J Pharm 2025; 676:125611. [PMID: 40252865 DOI: 10.1016/j.ijpharm.2025.125611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
A novel β-cyclodextrin-aza[5]helicene conjugate as theranostic platform for anticancer agents delivery in cancer cells is here reported. The carrier was synthesized via monotosylation of hydroxyethyl-β-cyclodextrin (HE-β-CD), followed by reaction with the synthesized aza[5]helicene, yielding the corresponding ammonium tosylate salt. The system was characterized by NMR, FTIR, UV-vis, and PL measurements, demonstrating favorable optical properties. The suitability of the fluorescent system to act as smart drug delivery system for cancer therapy was investigated by choosing gemcitabine (GMC) as a model drug. The GMC inclusion inside the system was evaluated by experimental and computational studies which confirmed the formation of a 1:1 complex between β-CD and GMC. The inclusion of GMC within the β-CD cavity led to a marked enhancement in its water solubility. Biological tests conducted on A549 cells revealed high cell internalization (∼80 %) and low cytotoxicity (IC50 = 262.7 µg mL-1) of the β-CD-aza[5]helicene conjugate. The results obtained by exploiting the host-guest chemistry of β-cyclodextrin combined with the unique photophysical properties of aza[5]helicene could pave the way for new anticancer therapies, by increasing the therapeutic index of anticancer agents endowed with poor solubility in water and characterized by systemic toxicity and, thanks to the fluorescent properties of the inserted probe, following their release into biological pathways.
Collapse
Affiliation(s)
- Consuelo Celesti
- Department of Engineering, University of Messina, C.da Di Dio, I-98166 Messina, Italy.
| | - Andrea Mele
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milano, Italy
| | - Claudia Espro
- Department of Engineering, University of Messina, C.da Di Dio, I-98166 Messina, Italy
| | - Giuseppina Raffaini
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milano, Italy
| | - Antonio Laganà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina 98125 Messina, Italy
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina 98125 Messina, Italy
| | - Salvatore Vincenzo Giofrè
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Federica De Gaetano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Giovanni Neri
- Department of Engineering, University of Messina, C.da Di Dio, I-98166 Messina, Italy
| | - Tullio Caronna
- Department of Engineering and Applied Sciences, University of Bergamo, Bergamo 24044, Italy
| | - Daniela Iannazzo
- Department of Engineering, University of Messina, C.da Di Dio, I-98166 Messina, Italy
| |
Collapse
|
15
|
Ahsan MJ, Kumar V, Ali A, Ali A, Yusuf M, Ahmad I, Patel H, Salahuddin, Ahsan MF. Design and synthesis of newer 5-aryl- N-(naphthalen-2-yl)-1,3,4-oxadiazol-2-amine analogues as anticancer agents. Future Med Chem 2025:1-12. [PMID: 40371593 DOI: 10.1080/17568919.2025.2504335] [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] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
AIM Cancer is the second leading cause of death and chemotherapy is widely used and well-known for treating cancer, yet it has lots of adverse side effects, making the search for novel compounds imperative. We reported here design, synthesis, DFT analysis, anticancer evaluation and in-silico studies of new 1,3,4-oxadiazoles (4a-e). MATERIAL AND METHODS IMC-038525 and IMC-094332 tubulin inhibitors' oxadiazole-linked aryl cores inspired the innovative compounds, and synthesis was accomplished in two steps followed by their characterization by spectral data. The HOMO and LUMO energy gap (ΔE) was determined to investigate compounds' (4a-e) stability followed by their anticancer activity at 10 μM and in-silico studies. RESULTS AND CONCLUSION 5-(4-Nitrophenyl)-N-(naphthalene-2-yl)-1,3,4-oxadiazol-2-amine (4b) demonstrated substantial anticancer activity against a few cell lines like SR, MDA-MB-435, MOLT-4, K-562, and HL-60(TB). 5-(3,4,5-Trimethoxyphenyl)-N-(naphthalene-2-yl)-1,3,4-oxadiazol-2-amine (4e) demonstrated promising anticancer activity against cell lines, UO-31, NCI-H226, CAKI-1, PC-3, and MCF7. The molecular docking against tubulin's colchicine binding site (PDB ID: 1AS0), displayed a docking score of -7.295 Kcal/mol and a H-bond interaction with Ala317 residue for the ligand 4e. The ligand 4e was found to interacted 24 amino acids of the tubulin protein in MD simulation investigation with moderate local conformational changes with ligand 4e (< 1 Å).
Collapse
Affiliation(s)
- Mohamed Jawed Ahsan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jahangirabad Institute of Technology, Barabanki, Uttar Pradesh, India
| | - Vivek Kumar
- Department of Pharmaceutical Chemistry, Maharishi Arvind College of Pharmacy, Ambabari Circle, Jaipur, Rajasthan, India
| | - Amena Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Abuzer Ali
- Department of Pharmacognosy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Mohammad Yusuf
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Iqrar Ahmad
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Salahuddin
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering & Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Md Faiyaz Ahsan
- Department of Chemistry, Bihar National College, Patna, Bihar, India
| |
Collapse
|
16
|
Luo J, Huang C, Liao Z, Ma X, Si T, Chen H, Li Z, Fan J. Unidirectional Drug Delivery and Responsive Release Guided by Nanofunnel-Shaped Heterojunction. NANO LETTERS 2025; 25:7853-7859. [PMID: 40323291 DOI: 10.1021/acs.nanolett.5c00617] [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: 05/15/2025]
Abstract
Drug resistance often involves preventing drug entry or expelling drugs from cells, severely affecting the therapeutic effect. We propose nanofunnel-shaped devices by pairing truncated graphene/boron nitride nanocones and nanotubes using curvature gradients and material properties to modulate energy barriers for unidirectional drug delivery. Molecular dynamics simulations demonstrate spontaneous delivery across models (ΔG = -14.14 to -27.87 kcal·mol-1 for C∨-C||, BN∨-BN||, C∨-BN||). The potential of mean force calculations reveal energy barriers scale as ΔG ∝ 1/R2, with BN nanotubes showing 20-30% higher barriers (e.g., -19.63 vs -14.14 kcal·mol-1 for graphene) due to stronger van der Waals interactions. In the BN∨-C|| model, increasing the tube radius (9.59 to 20.34 Å) or decreasing the cone angle (180-60°) can flip ΔG, enabling bidirectional control. This curvature-material synergy bypasses efflux mechanisms, offering a tunable platform to combat drug resistance and enhance therapeutic precision.
Collapse
Affiliation(s)
- Jun Luo
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, 999077, China
| | - Changxiong Huang
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, 999077, China
| | - Zhenyu Liao
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, 999077, China
| | - Xinyao Ma
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, 999077, China
| | - Ting Si
- Department of Physics, City University of Hong Kong, Hong Kong, 999077, China
| | - Huan Chen
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, 999077, China
| | - Zhen Li
- School of Materials Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Jun Fan
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong, 999077, China
- Center for Advanced Nuclear Safety and Sustainable Development, City University of Hong Kong, Hong Kong, 999077, China
- Department of Mechanical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
17
|
Bhattacharya E, Shaw S, Nayak R, Bose S. Advances in targeted therapy for inflammatory breast cancer: nanomaterials, conventional treatments, and clinical applications. NANOTECHNOLOGY 2025; 36:222002. [PMID: 40294602 DOI: 10.1088/1361-6528/add165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/28/2025] [Indexed: 04/30/2025]
Abstract
Inflammatory breast cancer (IBC) presents a formidable challenge due to its rapid progression and unique clinical characteristics within the various manifestations of breast cancer. Despite being rare, its aggressive nature demands innovative approaches beyond conventional treatments. Nanomedicine offers exciting possibilities for improving all types of breast cancer therapeutics including IBC. In this review, we critically assess the current treatment landscape for IBC, highlighting the limitations of traditional methods and addressing the pressing need for new therapeutic strategies. Although many nanomaterials have been explored for breast cancer therapeutics, either alone or in combination with other therapies, only a limited number of nanotherapeutics have been extensively studied for IBC treatment. This review further explores how advancements in nanotechnology, such as nanoparticle- mediated photothermal therapy, Photodynamic therapy, and nanomedicinal targeted therapies can offer novel avenues for addressing the unique biological, technological, and regulatory challenges posed by IBC. IBC-related various nanomedicines based combinatorial therapies are highlighted in this review. It also provides a forward-looking perspective on key research directions and clinical applications.
Collapse
Affiliation(s)
- Eshana Bhattacharya
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Siuli Shaw
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Sudeep Bose
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
18
|
Payamifar S, Khalili Y, Foroozandeh A, Abdouss M, Hasanzadeh M. Magnetic mesoporous silica nanoparticles as advanced polymeric scaffolds for efficient cancer chemotherapy: recent progress and challenges. RSC Adv 2025; 15:16050-16074. [PMID: 40370857 PMCID: PMC12076205 DOI: 10.1039/d5ra00948k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
Magnetic mesoporous silica nanoparticles (MMS NPs) stand out as excellent options for targeted chemotherapy owing to their remarkable features, such as extensive surface area, substantial pore volume, adjustable and uniform pore size, facile scalability, and versatile surface chemistry. This review comprehensively explores the latest developments in MMS NPs, emphasizing their design, functionalization, and application in cancer therapy. Initially, we discuss the critical need for targeted and controlled drug delivery (DD) in oncology, highlighting the role of magnetic and MMs in addressing some challenges. Subsequently, the key features of MMS NPs, such as their high surface area, pore structure, and functionalization strategies, are examined for their impact on their DD performance for efficient cancer chemotherapy. The integration of chemotherapy methods such as photothermal therapy and photodynamic therapy with MMS NPs is also explored, showcasing multifunctional platforms that combine imaging and therapeutic capabilities. Finally, we identify the current challenges and provide future perspectives for the development and clinical translation of MMS NPs, underscoring their potential to reshape CT paradigms.
Collapse
Affiliation(s)
- Sara Payamifar
- Department of Chemistry, Amirkabir University of Technology Tehran Iran
| | - Yasaman Khalili
- School of Chemistry, Faculty of Science, University of Tehran Iran
| | - Amin Foroozandeh
- Department of Chemistry, Amirkabir University of Technology Tehran Iran
| | - Majid Abdouss
- Department of Chemistry, Amirkabir University of Technology Tehran Iran
| | - Mohammad Hasanzadeh
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
19
|
Truong TT, Doan VHM, Nguyen DQ, Nguyen QD, Choi J, Subramaniyan B, Ahn J, Lee B, Oh J, Mondal S. Synergistic Therapeutic Effects of Prussian Blue Erbium-Doped Hydroxyapatite Nanoparticles in Photothermal Photodynamic Cancer Therapy. ACS Biomater Sci Eng 2025; 11:2639-2652. [PMID: 40168056 DOI: 10.1021/acsbiomaterials.5c00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
This study explores the synergistic therapeutic potential of Prussian Blue Erbium-Doped Hydroxyapatite (PB-Er-HAp) bioceramics in the context of photothermal therapy (PTT) and photodynamic therapy (PDT) for cancer treatment, highlighting their role in multimodal therapeutic approaches and imaging. PB-Er-HAp nanoparticles (NPs) were synthesized using a facile coprecipitation method to incorporate erbium (Er) into nanostructured hydroxyapatite (HAp) at various concentrations. Prussian Blue (PB) was functionalized onto the surfaces of these NPs, resulting in a final particle size of less than 50 nm. The therapeutic efficacy of the synthesized 1.0 mol % PB-Er-HAp NPs was evaluated in vitro, using MDA-MB-231 breast cancer cells. In vitro studies demonstrated that the PB-Er-HAp NPs exhibited significant PTT and PDT effects under 808 nm laser irradiation, effectively inducing cancer cell death through heat generation and reactive oxygen species production, respectively. In vitro experiments validated the ability of NPs to inhibit tumor growth in the MDA-MB-231 breast cancer cell line. This study emphasizes the potential of PB-Er-HAp NPs as a versatile platform for synergistic cancer therapy, combining PTT and PDT effects, while offering capabilities for biomedical imaging. Future research aims to further optimize these NPs and explore their clinical application, aiming toward enhanced therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
- Thi Thuy Truong
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Vu Hoang Minh Doan
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Dinh Quan Nguyen
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Quoc Dung Nguyen
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Jaeyeop Choi
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | | | - Jaesung Ahn
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Byeongil Lee
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
- Digital Healthcare Research Center, Pukyong National University, Busan 48513, Republic of Korea
| | - Junghwan Oh
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
- Digital Healthcare Research Center, Pukyong National University, Busan 48513, Republic of Korea
- Ohlabs Corp., Busan 48513, Republic of Korea
| | - Sudip Mondal
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
- Digital Healthcare Research Center, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
20
|
Tran NA, Moonshi SS, Lam AK, Lu CT, Vu CQ, Arai S, Ta HT. Nanomaterials in cancer starvation therapy: pioneering advances, therapeutic potential, and clinical challenges. Cancer Metastasis Rev 2025; 44:51. [PMID: 40347350 PMCID: PMC12065774 DOI: 10.1007/s10555-025-10267-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025]
Abstract
Gaining significant attention in recent years, starvation therapy based on the blocking nutrients supply to cancer cells via blood occlusion and metabolic interventions is a promisingly novel approach in cancer treatment. However, there are many crucial obstacles to overcome to achieve effective treatment, for example, poor-targeting delivery, cellular hypoxia, adverse effects, and ineffective monotherapy. The starvation-based multitherapy based on multifunctional nanomaterials can narrow these gaps and pave a promising way for future clinical translation. This review focuses on the progression in nanomaterials-mediated muti-therapeutic modalities based on starvation therapy in recent years and therapeutic limitations that prevent their clinical applications. Moreover, unlike previous reviews that focused on a single aspect of the field, this comprehensive review presents a broader perspective on starvation therapy by summarising advancements across its various therapeutic strategies.
Collapse
Affiliation(s)
- Nam Anh Tran
- School of Environment and Science, Griffith University, Nathan, QLD, 4111, Australia
| | - Shehzahdi S Moonshi
- School of Environment and Science, Griffith University, Nathan, QLD, 4111, Australia
| | - Alfred K Lam
- School of Medicine and Dentistry, Griffith University, Southport, QLD, 4215, Australia
- Gold Coast University Hospital, Southport, QLD, 4215, Australia
| | - Cu Tai Lu
- School of Medicine and Dentistry, Griffith University, Southport, QLD, 4215, Australia
- Gold Coast University Hospital, Southport, QLD, 4215, Australia
| | - Cong Quang Vu
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Japan
| | - Satoshi Arai
- WPI Nano Life Science Institute, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Japan
| | - Hang Thu Ta
- School of Environment and Science, Griffith University, Nathan, QLD, 4111, Australia.
| |
Collapse
|
21
|
Liu L, Tu B, Sun Y, Liao L, Lu X, Liu E, Huang Y. Nanobody-based drug delivery systems for cancer therapy. J Control Release 2025; 381:113562. [PMID: 39993634 DOI: 10.1016/j.jconrel.2025.02.058] [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: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Targeted delivery can elevate the local drug concentration within tumor tissues, while minimizing drug distribution to normal tissues, thus enhancing the effectiveness of anti-tumor medications and reducing adverse effects and systemic toxicities. Nanobodies, the novel molecular pattern of antibodies characterized by their small size, high stability, strong specificity, and low immunogenicity, have been extensively applied in targeted drug delivery for tumor therapy. This review discusses structural disparities and functional advantages of nanobodies compared to other antibody fragments and full-length antibody. It also highlights nanobody applications in targeted tumor therapy, focusing on their use in modifying delivery systems, e.g., liposomes, EVs, micelles, albumin nanoparticles, gold nanoparticles, polymeric nanoparticles, and as nanobody-drug conjugates. This review delves into the methods applied for integrating nanobodies into different drug delivery carriers, in order to provide useful information for researchers developing nanobody-based targeted drug delivery systems.
Collapse
Affiliation(s)
- Lin Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Bin Tu
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yao Sun
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Lingling Liao
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Xiaoling Lu
- College of Stomatology, Guangxi Medical University, Nanning 530021, China
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Yongzhuo Huang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| |
Collapse
|
22
|
Chavengkijsakol D, Kongjan J, Suwanwattanakul T, Trakarnsanga B, Areepium N, Siritientong T. Cancer Patients' Behavior and Perception on the Use of Medical Foods and Dietary Supplements During Chemotherapy. Patient Prefer Adherence 2025; 19:1385-1395. [PMID: 40370472 PMCID: PMC12075470 DOI: 10.2147/ppa.s514170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
Purpose The emerging clinical implications of medical foods and dietary supplements in cancer patients have been recognized. This study aimed to evaluate the perception and usage of these products in cancer patients undergoing chemotherapy. Patients and Methods Cross-sectional descriptive research was conducted by face-to-face interviews between October 2017 and February 2018. The participants provided written informed consent before data collection. Results This study included 201 patients (mean age 55.9 years) with gastrointestinal, breast, gynecological, and respiratory tract cancers, primarily receiving antimetabolite or platinum-based regimens. Awareness of medical foods and dietary supplements was high, at 97% and 98%, respectively. Most patients (91.5% for medical foods, 80.1% for dietary supplements) believed these products could be used safely without side effects, and over 70% thought they could be used concurrently with chemotherapy. More than half of the patients reported receiving supplement information from friends or relatives, while 65.2% stated that healthcare providers did not ask about their uses. Notably, 69.7% and 51.2% of patients reported current use of medical foods and dietary supplements, respectively, but 61.7% did not disclose this to their healthcare providers primarily since they were not asked. Conclusion These findings highlight the need for healthcare professionals to actively address the use of medical foods and dietary supplements with cancer patients. Enhanced communication and guidance could ensure safe and effective integration of these products into supportive cancer care.
Collapse
Affiliation(s)
- Danupol Chavengkijsakol
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Jirayut Kongjan
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Thanapoom Suwanwattanakul
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | | | - Nutthada Areepium
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tippawan Siritientong
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
23
|
Naz Z, Fareed M, Chaudhary ARH, Snigdha NT, Zafar A, Alsaidan OA, Mangu K, Ahmad S, Aslam M, Rizwanullah M. Exploring the therapeutic potential of ligand-decorated nanostructured lipid carriers for targeted solid tumor therapy. Int J Pharm 2025; 678:125687. [PMID: 40348302 DOI: 10.1016/j.ijpharm.2025.125687] [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: 03/11/2025] [Revised: 04/19/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
Solid tumors present significant therapeutic challenges due to their complex pathophysiology, including poor vascularization, dense extracellular matrix, multidrug resistance, and immune evasion. Conventional treatment strategies, such as chemotherapy, radiotherapy, and surgical interventions, are often associated with systemic toxicity, suboptimal drug accumulation at the tumor site, and chemoresistance. Nanostructured lipid carriers (NLCs) have emerged as a promising approach to enhance anticancer therapy. NLCs offer several advantages, including high drug loading capacity, improved bioavailability, controlled release, and enhanced stability. Recent advancements in active targeting strategies have led to the development of ligand-decorated NLCs, which exhibit selective tumor targeting, improved cellular uptake, and reduced systemic toxicity. By functionalizing NLCs with different targeting ligands, site-specific drug delivery can be achieved for better therapeutic efficacy. This review comprehensively explores the potential of ligand-decorated NLCs in solid tumor therapy, highlights their design principles, and mechanisms of tumor targeting. Furthermore, it discusses various receptor-targeted NLCs for the effective treatment of solid tumors. The potential of ligand-decorated NLCs in combination therapy, gene therapy, photothermal therapy, and photodynamic therapy is also explored. Overall, ligand-decorated NLCs represent a versatile and effective strategy to achieve better therapeutic outcomes in solid tumor therapy.
Collapse
Affiliation(s)
- Zrien Naz
- Department of Pharmaceutics, College of Pharmacy, Al Asmarya University, Zliten 218521, Libya
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | | | - Niher Tabassum Snigdha
- Department of Dental Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 602105 Tamil Nadu, India
| | - Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka-72341, Al-Jouf, Saudi Arabia
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka-72341, Al-Jouf, Saudi Arabia
| | - Karthik Mangu
- Kogniverse Education and Research, Bionest, Avishkaran (NIPER), Hyderabad-500037, Telangana, India
| | - Shahnawaz Ahmad
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001 Kurdistan Region, Iraq
| | - Md Rizwanullah
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401 Punjab, India.
| |
Collapse
|
24
|
Hatim MS, Al-Saffar AZ, Al-Aadhami MAWS. 5-Bromouracil-gracillin (5BrU-G) complex: an APOBEC3-activated therapeutic strategy exploiting cancer-specific enzymatic activity for selective cytotoxicity. Med Oncol 2025; 42:203. [PMID: 40335833 DOI: 10.1007/s12032-025-02745-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
Recent cancer treatment development has focused on smart drugs, primarily using nanomaterials as carriers. However, concerns about nanomaterial fate and body clearance have led to exploring alternative approaches. This study presents a novel targeted smart drug that uses normal lymphocytic cells as carriers and exploits cancer microenvironment characteristics for drug release, avoiding systemic damage. The research investigated a complex combining gracillin (natural carrier) and the chemotherapeutic agent 5-bromouracil (5-BrU). Molecular docking showed the 5BrU-G complex had superior binding affinity (- 7.96 kcal mol-1) to glycosylated adhesion domain of human T lymphocyte glycoprotein CD2 (1CDB) cell surface receptors in silico. The complex was successfully synthesized through double replacement, precipitation, and neutralization reactions, confirmed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Cytotoxic and genotoxic studies revealed the selectivity of 5BrU-G against cancer cells (MDA-MB-231 and Caco-2) while saving normal cells (MCF-10A and CCD 841 CoN). Unlike 5-BrU alone, which showed significant genotoxicity in normal cells, the 5BrU-G complex demonstrated minimal toxic effects. The selective targeting mechanism of 5BrU-G relies on APOBEC3 enzyme activity, which is elevated in cancer cells but is absent in normal cells. This was confirmed when APOBEC3 inhibition prevented the complex's cancer-killing activity. This novel approach offers promising alternatives for improving cancer therapy efficacy while reducing side effects.
Collapse
Affiliation(s)
- Mays S Hatim
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Ali Z Al-Saffar
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq.
| | | |
Collapse
|
25
|
Ragheb MA, Ragab MS, Mahdy FY, Elsebaie MS, Saber AM, AbdElmalak YO, Elsafoury RH, Elatreby AA, Rochdi AM, El-Basyouni AW, Shoukry MM, Eldeeb MA, El-Sherif RM, Abdelhamid IA, Salah-Eldin DS. Folic acid-modified chitosan nanoparticles for targeted delivery of a binuclear Co(II) complex in cancer therapy. Int J Biol Macromol 2025; 311:144034. [PMID: 40345288 DOI: 10.1016/j.ijbiomac.2025.144034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/21/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
A salient challenge in cancer chemotherapy is the successful delivery of drugs to cancer cells. Therapeutic agents can be delivered to cancer cells in a targeted and efficient manner using nanoparticles (NPs). Herein, we present the molecular characterization of a novel binuclear Co(II) complex with octahedral geometry based on Schiff base from dehydroacetic acid and piperazine derivatives. DNA and BSA binding interactions were investigated using UV-Vis spectroscopy and gel electrophoresis. In vitro cytotoxicity of Co(II) complex was assessed against microbes and human cells (Cancer: MDA-MB-231, MCF7, A375, HepG2; Non-cancerous: HSF, WI-38) using well diffusion and MTT assays. Chitosan decorated with folic acid (CS-FA) was fabricated to encapsulate Co(II) complex, which may serve as a nano-targeted drug delivery system, to dampen its adverse effects on non-cancerous cells. TEM and DLS analysis confirmed nano-sized and stable monodisperse nanosuspension of both (CS-FA) and (CS-FA-Co(II) complex) systems. CS-FA-Co(II) complex NPs exhibited an 8.3-fold increase in cytotoxicity against folate-receptor-positive MDA-MB-231 cells, while remaining safe for folate-receptor-negative HSF cells. They also induced cell cycle arrest, inhibited migration, and triggered apoptosis by modulating Bax, Bcl-2, caspase-3, and CDH1. These findings highlight CS-FA NPs as a promising targeted delivery system for Co(II) complex-based cancer therapeutic agents, offering improved efficacy.
Collapse
Affiliation(s)
- Mohamed A Ragheb
- Chemistry Department (Biochemistry Division), Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt; Biotechnology Department, Faculty of Nanotechnology for Postgraduate Studies, Cairo University, Sheikh Zayed Branch Campus, Giza 12588, Egypt.
| | - Mona S Ragab
- Chemistry Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Fatma Y Mahdy
- Biotechnology Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Mohamed S Elsebaie
- Biotechnology Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Amal M Saber
- Biotechnology Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Youstina O AbdElmalak
- Biotechnology Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Reem H Elsafoury
- Biotechnology Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Amal A Elatreby
- Biotechnology Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Ahmed M Rochdi
- Biotechnology Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Ahmed W El-Basyouni
- Biotechnology Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Mohamed M Shoukry
- Chemistry Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| | - Mohamed A Eldeeb
- Department of Chemistry, Illinois State University, IL, United States.
| | - Rabab M El-Sherif
- Chemistry Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt; Faculty of Postgraduate Studies for Nanotechnology, Cairo University, P.O. 12588, Giza, Egypt
| | - Ismail A Abdelhamid
- Chemistry Department, Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt.
| | - Doaa S Salah-Eldin
- Chemistry Department (Biochemistry Division), Faculty of Science, Cairo University, Giza, P.O. 12613, Egypt
| |
Collapse
|
26
|
Saremi Poor A, Davaeil B, Ramezanpour M, Shafiee Ardestani M, Moosavi-Movahedi AA, Asghari SM. Nanoparticle Albumin-Bound Bortezomib: Enhanced Antitumor Efficacy and Tumor Accumulation in Breast Cancer Therapy. Mol Pharm 2025; 22:2482-2493. [PMID: 40223780 DOI: 10.1021/acs.molpharmaceut.4c01283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Nanoparticle albumin-bound (NAB) formulations are emerging as a viable strategy for the intravenous delivery of poorly water-soluble drugs. This study aims to improve the therapeutic profile of Bortezomib (BTZ), addressing its low solubility and significant systemic toxicity through the development of NAB-BTZ nanoparticles. The synthesized nanoparticles exhibited an average size of 296.47 ± 10 nm and a high drug encapsulation efficiency of 75%, and a drug loading of 10%. NAB-BTZ displayed a controlled, pH-sensitive release profile, with 59% release at pH 5.4 (mimicking tumor environments) and 46% at pH 7.4 after 12 h. In vitro assays demonstrated that NAB-BTZ significantly reduced the viability of 4T1 mammary carcinoma cells in a dose- and time-dependent manner, increasing late apoptosis from 6% to 54% after 48 h, compared to 24% for free BTZ. At molecular level, NAB-BTZ induced apoptosis by upregulating p53 and Bax, downregulating Bcl-2, and activating caspases 3 and 7. In vivo tests in a murine 4T1 breast cancer model showed that NAB-BTZ substantially inhibited tumor growth, achieving an average tumor volume of 916 mm3 by day 31 versus 1400 mm3 for free BTZ, leading to an improved survival rate of 100% compared to 83% in the BTZ group. Technetium-99m (99mTc) labeling and SPECT imaging confirmed enhanced targeting capability, showing preferential accumulation of NAB-BTZ in tumor sites compared to free BTZ. These findings suggest that NAB-BTZ not only improves antitumor efficacy but also enhances its safety profile, underscoring its clinical potential in breast cancer therapy.
Collapse
Affiliation(s)
- Anita Saremi Poor
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, 1417614411 Tehran, Iran
| | - Bagher Davaeil
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, 1417614411 Tehran, Iran
| | - Marziyeh Ramezanpour
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, 1417614411 Tehran, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, 1461884513 Tehran, Iran
- Research Center for Molecular Medicine, Shariati Hospital, North Kargar Avenue, 1411713135 Tehran, Iran
| | | | - S Mohsen Asghari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, 1417614411 Tehran, Iran
| |
Collapse
|
27
|
Tomić K, Kostevšek N, Romeo S, Bassanini I. Neuropeptide Y receptors 1 and 2 as molecular targets in prostate and breast cancer therapy. Biomed Pharmacother 2025; 187:118117. [PMID: 40319656 DOI: 10.1016/j.biopha.2025.118117] [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: 02/24/2025] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025] Open
Abstract
Recent advances have revealed the overexpression of Neuropeptide Y (NPY) receptors in multiple cancers, positioning them as attractive molecular targets for cancer diagnostics and therapeutics. Despite this, a comprehensive roadmap for the rational development of anticancer agents targeting NPY receptors remains lacking. Therefore, we present the characteristics of NPY receptor subtypes, their abundance, and the correlation of their expression in different cancer types. It was found that NPY receptor subtypes 1 and 2 were extensively studied, especially in connection with breast and prostate cancer. Many tumors express NPYR, but only breast cancer tissue shows a significant difference in NPYR subtype expression levels between tumor and normal tissues, and, therefore, can represent a promising target. In the context of anticancer therapy, this review provides key findings from the use of wild-type and synthetic NPY analogs. We highlight the critical residues in the NPY sequence that play a critical role in interactions with receptors and provide the recent literature findings on NPY analogues as efficient and specific cancer-targeting agents. Potential solutions to improve NPY analogs' stability are provided, such as sequence modifications of linear peptides, peptide stapling, and conjugation for drug delivery systems. In general, NPY treatment can not be used efficiently as a single therapy but as a combinatorial therapy with anticancer drugs to improve the specificity of the treatment via high-affinity binding to the cancer cells and sensitizing them to chemotherapy.
Collapse
Affiliation(s)
- Katarina Tomić
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova 39, Ljubljana 1000, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana 1000, Slovenia
| | - Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova 39, Ljubljana 1000, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana 1000, Slovenia.
| | - Sergio Romeo
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, Milan, Italy; Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, Via Mario Bianco 9, Milan, Italy
| | - Ivan Bassanini
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, Via Mario Bianco 9, Milan, Italy
| |
Collapse
|
28
|
Wang Y, Zhang G, Zhang Z, Zhang M, Chen J, Wang K, Liu L, Bao J, Chen M, Qi X, Gao M. Plasma DNMT1 Activity for Assessing Tumor Burden and Predicting Neoadjuvant Therapy Response in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501064. [PMID: 40317882 DOI: 10.1002/advs.202501064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/03/2025] [Indexed: 05/07/2025]
Abstract
DNA methylation is mediated by DNA methyltransferases (DNMTs), and the stability of their activity is essential for cellular fate. DNMT1 is considered one of the most promising targets for research. However, current detection techniques are limited in accurately quantifying its activity in peripheral blood. Here, a reaction system is developed known as DNMT1 Identification by Variable Activity (DIVA) for the highly sensitive detection of DNMT1 activity in the peripheral blood of breast cancer patients. DIVA can detect DNMT1 at levels as low as 10-7 U mL-1, with minimal time and cost. This method is applied to analyze 271 clinical samples, successfully evaluating tumor burden in patients staged I-IV. Finally, this method is utilized to assess the prognosis of 22 patients undergoing neoadjuvant therapy, demonstrating good consistency with ultrasound imaging results. It is believed that DIVA could serve as an effective auxiliary technique for both the early detection of breast cancer and evaluation of neoadjuvant therapy.
Collapse
Affiliation(s)
- Yingran Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Guozhi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Zhizhao Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Mengsi Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Jiao Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Ke Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Lu Liu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Jing Bao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Mingxuan Gao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| |
Collapse
|
29
|
Wang C, Gao X, Jiang Y, Zhang M, Liu L, Zhang S, Ye D, Jiang R. Molecular dynamics study of functionalized carbon nanotube loaded with multiple doxorubicin targeted to folate receptor α. J Mol Graph Model 2025; 136:108964. [PMID: 39908587 DOI: 10.1016/j.jmgm.2025.108964] [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: 11/03/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Two novel targeted drug delivery systems (DDSs) were designed: folate (FOL) conjugated (9, 9) carbon nanotube (CNT) loaded with 20 doxorubicin (DOX) molecules (FOL-CNT/20DOX) and folate (FOL) conjugated carboxylated (9, 9) CNT (COOH-CNT) loaded with 24 doxorubicin (DOX) molecules (FOL-COOH-CNT/24DOX). The targeted property to folate receptor α (FRα) was calculated using molecular dynamics (MD) calculations. The structures of the FRα/FOL-CNT/20DOX and FRα/FOL-COOH-CNT/24DOX complexes were analyzed in detail. Radial distribution functions were calculated to analyze the distribution of DOX molecules around the CNTs in the complexes. The variation of representative distances and angles between novel DDSs and FRα, number of hydrogen bonds, and secondary structures of FRα during the MD simulations were studied to analyze the dynamic properties of the novel DDSs targeted to FRα. We further analyzed the root mean square displacement and root mean square fluctuation in detail. The results indicate that the two novel DDSs were very stable and well targeted with FRα, and FOL-COOH-CNT/24DOX had better targeting and stability than FOL-CNT/20DOX. This study is expected to provide insights for the design of efficient nano drug delivery systems with good FRα targeting and controllable drug loading dosage.
Collapse
Affiliation(s)
- Cuihong Wang
- School of Science, Tianjin Chengjian University, Tianjin, China.
| | - Xin Gao
- School of Science, Tianjin Chengjian University, Tianjin, China
| | - Yue Jiang
- School of Science, Tianjin Chengjian University, Tianjin, China
| | - Meiling Zhang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Lijuan Liu
- School of Science, Tianjin Chengjian University, Tianjin, China
| | - Shouchao Zhang
- School of Science, Tianjin Chengjian University, Tianjin, China.
| | - Dan Ye
- School of Science, Tianjin Chengjian University, Tianjin, China
| | - Rongyun Jiang
- School of Science, Tianjin Chengjian University, Tianjin, China
| |
Collapse
|
30
|
Sharif M, Irfan M, Kousar K, Mamurova A, Duarte-Peña L, Hernández-Parra H, Cortés H, Peña-Corona SI, Khan K, Habtemariam S, Leyva-Gómez G, Sharifi-Rad J. Unlocking the biological potential of methyl antcinate A: a new frontier in cancer and inflammation application. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4727-4745. [PMID: 39630281 DOI: 10.1007/s00210-024-03544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/14/2024] [Indexed: 04/11/2025]
Abstract
Antrodia camphorata is a valued and scarce parasitic mushroom that exclusively proliferates on the inner cavity wall of the endangered tree Cinnamomum kanehirai Hay (Lauraceae), endemic to Taiwan. Historically, this fungus has been utilized in traditional medicine to treat liver cancer, diarrhea, abdominal pain, hypertension, and food and drug intoxication, among other ailments. Literature searches were performed in scientific databases. The results were compiled from peer-reviewed studies; the search was refreshed through January 2024 to incorporate the most recent research. In vitro studies have revealed that Antrodia camphorata possesses various pharmacological properties that prevent cancer, reduce inflammation, and improve liver function. This medicinal mushroom contains unique ergostane-type triterpenoids known as antcins, which exhibit numerous pharmacological properties. Seven naturally occurring methyl analogs of antcins have been identified so far. In this article, we reviewed and analyzed the properties of methyl antcinate A (MAA), a constituent of Antrodia camphorata and methyl derivative of antcin A. MAA has demonstrated important anti-apoptotic, anti-inflammatory, and gastro-protective properties, as well as significant anti-tumor, anti-cancer, and cytotoxic activities. The anti-cancer effect of MAA in various cancers is attributed to its ability to modulate signaling cascades in apoptotic pathways. A significant challenge is to initiate preclinical and clinical trials to assess its anti-tumor action in vivo, as this data is currently missing. Additionally, future research on the structure-activity relationship of antcins and their derivatives is expected to support their development as therapeutic agents for clinical use.
Collapse
Affiliation(s)
- Maria Sharif
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Muhammad Irfan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
- Cancer Clinical Research Unit, Trials360, Lahore, 54000, Pakistan
| | - Kafila Kousar
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Assem Mamurova
- Department of Biodiversity of Bioresources, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Lorena Duarte-Peña
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Hector Hernández-Parra
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City, México
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Khushbukhat Khan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan.
- Cancer Clinical Research Unit, Trials360, Lahore, 54000, Pakistan.
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, Central Avenue, Chatham-Maritime, Kent, ME4 4TB, UK
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico.
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Ecuador.
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
31
|
Kandasamy T, Sarkar S, Zochedh A, Kathiresan T, Ghosh SS. Synergistic Effects of Epirubicin-Vorinostat-Pimozide Drug Cocktail on Proliferation, Stemness, Invasiveness, and Fatty Acid Metabolism in Breast Cancer Cells. IUBMB Life 2025; 77:e70020. [PMID: 40305333 DOI: 10.1002/iub.70020] [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: 01/15/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Chemotherapeutic treatments for breast cancer are often associated with severe toxicity due to the requirement of high concentrations of the drugs for efficacy. The combination of chemotherapy drugs along with repurposed drugs offers a promising strategy to enhance efficacy while reducing toxicity. However, the effectiveness of such combinations is likely to be hindered by improper metabolism of the drugs due to the sharing of the same metabolizing enzymes. In this study, we explored a novel approach to enhance the efficacy of Pimozide (repurposed drug) by combining it with chemotherapeutic drugs that utilize different metabolizing enzymes than Pimozide, thereby reducing metabolic load and toxicity. The Epirubicin-SAHA(Vorinostat)-Pimozide (ESP) combination emerged as highly synergistic, reducing the IC50 of Pimozide from 16.54 to 0.57 μM in MCF-7 cells and from 17.5 to 3.35 μM in MDA-MB-231 cells, representing a significant enhancement in efficacy. Mechanistic studies revealed increased intracellular reactive oxygen species (ROS) generation and activation of the intrinsic apoptosis pathway, as indicated by a 10-fold increase in the cleaved PARP levels. In MDA-MB-231 cells, there was also a 2-fold increase in p53 and a 10-fold increase in p21 expression, with a concomitant reduction in AKT signaling. Furthermore, the ESP combination reduced cancer stemness, invasiveness, fatty acid uptake, and lipid droplet accumulation, pointing to its broad impact on cancer cell survival and metabolism. These findings suggest that the ESP combination holds promise as an effective therapeutic strategy for breast cancer, with reduced toxicity and enhanced efficacy.
Collapse
Affiliation(s)
- Thirukumaran Kandasamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Shilpi Sarkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Azar Zochedh
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, India
| | - Thandavarayan Kathiresan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
32
|
Li X, Xia C, Jin Z, He Q. Ascorbate/methionine-based CH 4 delivery nanomedicine for tumor-targeted therapy. Biomaterials 2025; 316:123002. [PMID: 39675143 DOI: 10.1016/j.biomaterials.2024.123002] [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: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
Methane (CH4) is identified to be an emerging anti-inflammation and anti-cancer molecule with high bio-safety, but the targeted delivery of CH4 is a thorny challenge. Herein, we propose a CH4 delivery strategy based on an intratumoral H2O2-triggered cascade reaction of ascorbic acid (AA)/methionine (Met), and have constructed a new nanomedicine (AMN) for tumor-targeted CH4 therapy. Encouragingly, AMN realizes the effective tumor-targeted delivery and intratumoral H2O2-responsive release of CH4, and exhibits significant anti-cancer effects and high bio-safety. Mechanistically, we have discovered that intratumoral released CH4 can not only induce the apoptosis of 4T1 tumor cells by inhibiting their mitochondrial metabolism, but also activate tumor immunotherapy by reprogramming tumor-associated macrophages (TAMs) phenotype (M2 to M1). The combination of the above anti-cancer pathways by virtue of tumor-targeted CH4 delivery makes contribution to outstanding anti-cancer efficacy of AMN. The proposed CH4 delivery strategy opens a new window for safe and effective tumor therapy.
Collapse
Affiliation(s)
- Xiaoyu Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China; Shenzhen Research Institute, Shanghai Jiao Tong University, Shenzhen, 518057, China; Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chao Xia
- Shenzhen Research Institute, Shanghai Jiao Tong University, Shenzhen, 518057, China; Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhaokui Jin
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Qianjun He
- Shenzhen Research Institute, Shanghai Jiao Tong University, Shenzhen, 518057, China; Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
33
|
Esmaeilpour D, Ghomi M, Zare EN, Sillanpää M. Recent advances in DNA nanotechnology for cancer detection and therapy: A review. Int J Biol Macromol 2025; 307:142136. [PMID: 40107552 DOI: 10.1016/j.ijbiomac.2025.142136] [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: 12/28/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Deoxyribonucleic acid (DNA) nanotechnology has rapidly emerged as a transformative field in biomedical research, offering innovative solutions for the detection and treatment of cancer. This review provides a comprehensive analysis of the role of DNA-based nanosystems in oncology, emphasizing their potential to address the limitations of conventional diagnostic and therapeutic approaches. Key advancements in DNA nanotechnology include the development of highly specific and sensitive nanostructures for early cancer detection, as well as precision-targeted delivery systems that enhance the efficacy of cancer therapies while minimizing side effects. The objectives of this review are threefold: first, to summarize the latest advancements in DNA nanotechnology, highlighting innovations in cancer biomarker detection and therapeutic applications; second, to explore the molecular mechanisms that enable these DNA-based nanosystems to interact with cancer cells with remarkable precision, including their design principles, self-assembly processes, and biological interactions; and third, to discuss the future implications of these technologies, considering the challenges, potential breakthroughs, and the steps needed to integrate DNA nanotechnology into clinical practice. By achieving these objectives, the review aims to offer insights into how DNA nanotechnology could revolutionize cancer care, providing new strategies for more personalized and effective treatments, and ultimately improving patient outcomes in the battle against cancer.
Collapse
Affiliation(s)
- Donya Esmaeilpour
- Center for Nanotechnology in Drug Delivery, School of Pharmacy, Shiraz University of Medical Science, Shiraz 71345-1583, Iran.
| | - Matineh Ghomi
- Department of Chemistry, Jundi-Shapur University of Technology, Dezful, Iran
| | - Ehsan Nazarzadeh Zare
- School of Chemistry, Damghan University, Damghan 36716-45667, Iran; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Mika Sillanpää
- Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam; School of Engineering & Technology, Duy Tan University, Da Nang, Viet Nam.
| |
Collapse
|
34
|
Nathani A, Sun L, Li Y, Lazarte J, Aare M, Singh M. Targeting EGFR-TKI resistance in lung cancer: Role of miR-5193/miR-149-5p loaded NK-EVs and Carboplatin combination. Int J Pharm 2025; 675:125573. [PMID: 40204039 DOI: 10.1016/j.ijpharm.2025.125573] [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: 02/02/2025] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Lung cancer remains the leading cause of cancer-related deaths, and there is an urgent need for innovative therapies. MicroRNA (miRNA)-based gene therapy has shown promise, but efficient delivery systems are required for its success. This study investigates the use of extracellular vehicles (EVs) secreted by natural killer (NK) cells as delivery systems for miRNAs targeting PD-L1/PD-1 immune checkpoint and FOXM1, in combination with Carboplatin, to enhance anticancer efficacy in lung cancer models. NK-EVs were isolated from NK92-MI cells and characterized using nanoparticle tracking analysis (NTA), proteomics and Western blotting, confirming their exosomal characteristics. Gene ontology profiling and RNA-seq identified highly expressed miRNAs such as miR-5193 and miR-149-5p, which were loaded into NK-EVs via electroporation. Agarose gel electrophoresis confirmed their entrapment and Quickdrop spectrophotometer was used to estimate the quantity. In vitro, miRNA-loaded NK-EVs demonstrated significant cytotoxicity against Osimertinib-resistant PDX (TM0019, Jackson Labs) and H1975R (with L858R mutations) lung cancer cells, with approximately 1.2 to 1.6-fold (p < 0.01) decrease in cell viability compared to NK-EVs alone. In vivo, the combination of miRNA-loaded NK-EVs and Carboplatin significantly reduced tumor volumes (3.5 to 4-fold, p < 0.001) in PDX and H1975R xenograft models, with the most pronounced effect observed in combination therapies. Western blot analysis showed downregulation of tumor-associated markers: PD-1/PD-L1, FOXM1, Survivin, NF-κB and others vs untreated group, p < 0.001) suggesting immune checkpoint inhibition, apoptosis and anti-inflammatory activity. These findings highlight the potential of NK-EVs as effective carriers for miRNAs in combination with chemotherapy, offering a promising therapeutic strategy for NSCLC with EGFR mutations.
Collapse
Affiliation(s)
- Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA; Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Jassy Lazarte
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA.
| |
Collapse
|
35
|
Yang C, Ge C, Zhang W, Xu J. LSM2 drives glioma progression through alternative splicing dysregulation: a multi-omics approach to identify a potential therapeutic target. Front Oncol 2025; 15:1521608. [PMID: 40365337 PMCID: PMC12069061 DOI: 10.3389/fonc.2025.1521608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Background Glioma, particularly glioblastoma (GBM), remains a highly aggressive and challenging tumour, characterised by poor prognosis and limited therapeutic options. LSM2, an RNA-binding protein, has been implicated in tumour progression, yet its role in glioma remains underexplored. This study aims to investigate the expression, prognostic significance, and molecular mechanisms of LSM2 in glioma, focusing on its impact on RNA splicing regulation. Methods Clinical and transcriptomic data from 163 GBM and 518 lower-grade glioma (LGG) cases from The Cancer Genome Atlas (TCGA) were analysed to assess LSM2 expression and its prognostic value. RNA sequencing was performed on LSM2 knockdown in T98G glioblastoma cells to identify differentially expressed genes (DEGs) and alternative splicing events (ASEs). Bioinformatics tools were employed to perform functional enrichment analyses and construct protein-protein interaction (PPI) networks. Results LSM2 expression was significantly elevated in gliomas, particularly in GBM and in tumours with 1p/19q non-deletion or IDH1 mutation (p < 0.001). High LSM2 expression was correlated with shorter overall survival (HR = 1.7, p = 0.01). Knockdown of LSM2 in T98G cells identified 728 upregulated and 1,720 downregulated genes, alongside 1,949 splicing alterations, which primarily affected pathways related to RNA metabolism, DNA damage response, and cell cycle regulation. Key hub genes such as TLN1, FN1, and IRF7 were associated with glioma progression and poor prognosis. Conclusion Our findings demonstrate that LSM2 plays a critical role in glioma progression through the regulation of RNA splicing dynamics. Elevated LSM2 expression serves as a prognostic biomarker and offers promising potential as a therapeutic target in glioma.
Collapse
Affiliation(s)
| | | | | | - Jingxuan Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Xinjiang Medical
University, Urumqi, China
| |
Collapse
|
36
|
Zhang X, Zhao B, Fu S, Seruya RS, Fanos HE, Petrisor AA, Liu Y, Yang Z, Zhang F. Controlling stimulus sensitivity by tailoring nanoparticle core hydrophobicity. Biomater Sci 2025; 13:2332-2339. [PMID: 40138203 PMCID: PMC11939950 DOI: 10.1039/d5bm00163c] [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: 02/02/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a significant global health challenge, necessitating the development of more effective therapeutic strategies. This work presents a novel glutathione (GSH)-responsive platform designed to enhance the delivery and efficacy of the anticancer drug mertansine (DM1) through the modulation of pendant groups in polycarbonate-drug conjugates. By systematically varying the hydrophobicity of the pendant groups, we investigated their effects on nanostructures, GSH sensitivity, colloidal stability, drug release profiles, and the in vitro anticancer efficacy of these polymeric nanoparticles, revealing that more hydrophobic pendant groups effectively reduce GSH accessibility for the nanoparticle cores, improve colloidal stability, and slow drug release rates. The results underscore the critical importance of polymer structures in optimizing drug delivery systems and offer valuable insights for future research on advanced nanomaterials with enhanced drug delivery for cancer therapies.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Bowen Zhao
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Shiwei Fu
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Ronald S Seruya
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Hannah E Fanos
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Ashley A Petrisor
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Yilin Liu
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Zixin Yang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Fuwu Zhang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- The Dr John T. Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, 1951 NW 7th Ave, Miami, Florida, 33136, USA
| |
Collapse
|
37
|
Maksimović T, Minda D, Șoica C, Mioc A, Mioc M, Colibășanu D, Lukinich-Gruia AT, Pricop MA, Jianu C, Gogulescu A. Anticancer Potential of Cymbopogon citratus L. Essential Oil: In Vitro and In Silico Insights into Mitochondrial Dysfunction and Cytotoxicity in Cancer Cells. PLANTS (BASEL, SWITZERLAND) 2025; 14:1341. [PMID: 40364370 PMCID: PMC12073404 DOI: 10.3390/plants14091341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
This study aims to assess the potential anticancer activity of lemongrass essential oil (LEO) using in vitro and in silico methods. The steam hydrodistillation of the aerial parts yielded 3.2% (wt) LEO. The GC-MS analysis of the LEO revealed the presence of α-citral (37.44%), β-citral (36.06%), linalool acetate (9.82%), and d-limonene (7.05%) as major components, accompanied by several other minor compounds. The antioxidant activity, assessed using the DPPH assay, revealed that LEO exhibits an IC50 value of 92.30 μg/mL. The cytotoxic effect of LEO, as well as LEO solubilized with Tween-20 (LEO-Tw) and PEG-400 (LEO-PEG), against a series of cancer cell lines (A375, RPMI-7951, MCF-7, and HT-29) was assessed using the Alamar Blue assay; the results revealed a high cytotoxic effect against all cell lines used in this study. Moreover, neither one of the tested concentrations of LEO, LEO-PG, or LEO-TW significantly affected the viability of healthy HaCaT cells, thus showing promising selectivity characteristics. Furthermore, LEO, LEO-PG, and LEO-TW increased ROS production and decreased the mitochondrial membrane potential (MMP) in all cancer cell lines. Moreover, LEO treatment decreased all mitochondrial respiratory rates, thus suggesting its ability to induce impairment of mitochondrial function. Molecular docking studies revealed that LEO anticancer activity, among other mechanisms, could be attributed to PDK1 and PI3Kα, where the major contributors are among the minor components of the essential oil. The highest active theoretical inhibitor against both proteins was β-caryophyllene oxide.
Collapse
Affiliation(s)
- Tamara Maksimović
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (T.M.); (C.Ș.)
- Research Center for Experimental Pharmacology and Drug Design (X-Pharm Design), “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (M.M.); (D.C.)
| | - Daliana Minda
- Department Pharmacognosy-Phytotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania;
| | - Codruța Șoica
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (T.M.); (C.Ș.)
- Research Center for Experimental Pharmacology and Drug Design (X-Pharm Design), “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (M.M.); (D.C.)
| | - Alexandra Mioc
- Department of Pharmacology-Pharmacotherapy, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (T.M.); (C.Ș.)
- Research Center for Experimental Pharmacology and Drug Design (X-Pharm Design), “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (M.M.); (D.C.)
| | - Marius Mioc
- Research Center for Experimental Pharmacology and Drug Design (X-Pharm Design), “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (M.M.); (D.C.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Daiana Colibășanu
- Research Center for Experimental Pharmacology and Drug Design (X-Pharm Design), “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (M.M.); (D.C.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Alexandra Teodora Lukinich-Gruia
- OncoGen Centre, Clinical County Hospital “Pius Branzeu”, Blvd. Liviu Rebreanu 156, 300723 Timisoara, Romania; (A.T.L.-G.); (M.-A.P.)
| | - Maria-Alexandra Pricop
- OncoGen Centre, Clinical County Hospital “Pius Branzeu”, Blvd. Liviu Rebreanu 156, 300723 Timisoara, Romania; (A.T.L.-G.); (M.-A.P.)
- Department of Applied Chemistry and Environmental Engineering and Inorganic Compounds, Faculty of industrial Chemistry, Biotechnology and Environmental Engineering, Polytechnic University of Timisoara, Vasile Pârvan 6, 300223 Timisoara, Romania
| | - Calin Jianu
- Faculty of Food Engineering, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Calea Aradului 119, 300629 Timișoara, Romania;
| | - Armand Gogulescu
- Department XVI: Balneology, Medical Rehabilitation and Rheumatology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu, 300041 Timisoara, Romania;
| |
Collapse
|
38
|
Álvarez-Gómez C, Fonseca-Benítez AV, Guevara-Pulido J. Design, synthesis, and in vitro evaluation of a carbamazepine derivative with antitumor potential in a model of Acute Lymphoblastic Leukemia. PLoS One 2025; 20:e0319415. [PMID: 40293986 PMCID: PMC12036894 DOI: 10.1371/journal.pone.0319415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 02/01/2025] [Indexed: 04/30/2025] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a significant concern in both pediatric and adult demographics. Despite 156 approved cancer therapies based on small molecules, a mere five apply to all types of leukemia. Unfortunately, adherence to these treatments is low due to adverse side effects. Consequently, there is an urgent need to identify more effective treatment options for ALL. This study presents a potential solution. We have designed over fifty analogs of carbamazepine, utilizing a combination of ligand-based and structure-based drug design methodologies. Among these analogs, we identified the CR80 analog, which demonstrated predicted binding values of -8.66 kcal/mol against beta-tubulin, a favorable LogP, and IC50 values suitable for in vitro evaluation. The CR80 compound was synthesized with a yield of 50% and subsequently assessed in vitro against the U-937 cell line. It obtained an IC50 value of 0.8 micromolar to 1 micromolar and a selectivity index of two, thus marking it as a promising candidate for in vivo studies.
Collapse
|
39
|
Misawa S, Kogawa T, Naito Y, Suzuki T, Takada M, Denda T, Hino A, Suichi T, Kodama S, Miyoshi A, Shiosakai K, Kuwabara S. A subgroup analysis of the MiroCIP study to evaluate chemotherapy-induced peripheral neuropathy: symptom profile, severity, and analgesia efficacy depending on type of chemotherapy. Expert Opin Pharmacother 2025. [PMID: 40288415 DOI: 10.1080/14656566.2025.2499665] [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: 02/26/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The multicenter, prospective MiroCIP observational study investigated the incidence, risk factors, and outcomes of chemotherapy-induced peripheral neuropathy (CIPN) by oxaliplatin- and taxane-based chemotherapies but did not examine their differences in detail. This post hoc subanalysis explored the differences between oxaliplatin- and taxane-based chemotherapy, in terms of CIPN symptom profile, severity, and response to analgesics. RESEARCH DESIGN AND METHODS Patients with colorectal, gastric, non-small cell lung, or breast cancer, scheduled to receive oxaliplatin- or taxane-based chemotherapy, were followed for 12 months to assess the severity of sensory CIPN, by the Common Terminology Criteria for Adverse Events, and associated subjective and objective symptoms. RESULTS Overall, 91 patients received oxaliplatin and 131 received a taxane. At 12 months, CIPN prevalence was 74.6% with oxaliplatin and 55.2% with a taxane. Grade ≥ 2 CIPN peaked at 9 months with oxaliplatin and at 3 months with a taxane, with most symptom scores following a similar trajectory. Analgesic efficacy differed between subgroups, providing marked reductions in pain/tingling scores in the taxane group but minimal effect in the oxaliplatin group. CONCLUSIONS CIPN course and symptoms vary with oxaliplatin- or taxane-based chemotherapy. Effective management should be tailored to the type of chemotherapy: oxaliplatin-treated patients may benefit from continuous monitoring of CIPN symptoms, whereas it may be beneficial for taxane-treated patients to receive appropriate analgesics at CIPN onset. TRIAL REGISTRATION Japan Registry of Clinical Trials jRCTs031210101.
Collapse
Affiliation(s)
- Sonoko Misawa
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
- Department of Neurology and Neurological Science, Institute of Science Tokyo, Tokyo, Japan
| | - Takahiro Kogawa
- Division of Early Clinical Development for Cancer, Department of Advanced Medical Development, Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Yoichi Naito
- Department of General Internal Medicine/Experimental Therapeutics/Medical Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Mamoru Takada
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chuo-ku, Chiba, Japan
| | - Aoi Hino
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Internal Medicine, Chiba Medical Center, Chuo-ku, Chiba, Japan
| | - Tomoki Suichi
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Sho Kodama
- ASCA Primary Product Department, ASCA Business Division, Daiichi Sankyo Co., Ltd, Chuo-ku, Tokyo, Japan
| | - Arisa Miyoshi
- Primary Medical Science Department, Medical Affairs Division, Daiichi Sankyo Co., Ltd, Chuo-ku, Tokyo, Japan
| | - Kazuhito Shiosakai
- Data Intelligence Department, Global DX, Daiichi Sankyo Co., Ltd, Shinagawa-ku, Tokyo, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Chiba, Japan
| |
Collapse
|
40
|
Liu L, Zhang S, Ren Y, Wang R, Zhang Y, Weng S, Zhou Z, Luo P, Cheng Q, Xu H, Ba Y, Zuo A, Liu S, Liu Z, Han X. Macrophage-derived exosomes in cancer: a double-edged sword with therapeutic potential. J Nanobiotechnology 2025; 23:319. [PMID: 40287762 PMCID: PMC12034189 DOI: 10.1186/s12951-025-03321-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
Solid cancer contains a complicated communication network between cancer cells and components in the tumor microenvironment (TME), significantly influencing the progression of cancer. Exosomes function as key carriers of signaling molecules in these communications, including the intricate signalings of tumor-associated macrophages (TAMs) on cancer cells and the TME. With their natural lipid bilayer structures and biological activity that relates to their original cell, exosomes have emerged as efficient carriers in studies on cancer therapy. Intrigued by the heterogeneity and plasticity of both macrophages and exosomes, we regard macrophage-derived exosomes in cancer as a double-edged sword. For instance, TAM-derived exosomes, educated by the TME, can promote resistance to cancer therapies, while macrophage-derived exosomes generated in vitro have shown favorable potential in cancer therapy. Here, we depict the reasons for the heterogeneity of TAM-derived exosomes, as well as the manifold roles of TAM-derived exosomes in cancer progression, metastasis, and resistance to cancer therapy. In particular, we emphasize the recent advancements of modified macrophage-derived exosomes in diverse cancer therapies, arguing that these modified exosomes are endowed with unique advantages by their macrophage origin. We outline the challenges in translating these scientific discoveries into clinical cancer therapy, aiming to provide patients with safe and effective treatments.
Collapse
Affiliation(s)
- Long Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Siying Zhang
- Medical School of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ruizhi Wang
- Medical School of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, 450052, Henan, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
41
|
Wang L, Wang Q, Dong C, Teng C, Wang L, Zhou Y, Yang B, Kuang H, Sun Y. Exploring Tetrastigma hemsleyanum polysaccharides: A recent advance in preparation, structural features, bioactivities, and potential application prospects. Int J Biol Macromol 2025; 310:143477. [PMID: 40288710 DOI: 10.1016/j.ijbiomac.2025.143477] [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: 12/31/2024] [Revised: 04/13/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Tetrastigma hemsleyanum Diels et Gilg (T. hemsleyanum) is a traditional Chinese herb recognized as a 'plant antibiotic' due to its multiple beneficial effects on the human body. As a valuable plant, its wild resources are on the verge of extinction. Fortunately, advancements in artificial cultivation over the past two decades have led to an increase in high-quality plant resources. Consequently, research on this herb has been gaining popularity. Polysaccharides are an important component of T. hemsleyanum and have received extensive attention from scholars due to their various biological activities. Currently, various extraction and purification methods have been developed to isolate T. hemsleyanum polysaccharides (THPs). These polysaccharides have demonstrated significant effects in experiments, including antioxidant, anti-tumor, anti-inflammatory, immune regulation, metabolic-regulatory, and thermoregulatory effects. Furthermore, they possess broad application potential in fields such as food, medicine, and cosmetic industries. Unfortunately, a comprehensive review of the literature on THPs is currently lacking, which poses challenges for future research endeavors. This work aims to summarize the latest progress in the extraction, purification, structural characterization, biological activities, and applications of THPs across fields comprehensively from the past to the present, analyze the shortcomings of recent research, and discuss potential applications and future research directions.
Collapse
Affiliation(s)
- Lihao Wang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Qian Wang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Chenqing Dong
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Chi Teng
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Li Wang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Yuanyuan Zhou
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, Harbin 150040, China.
| |
Collapse
|
42
|
Xu M, Bai L, Sun M, Yan X, Xiong Y, Wang Y, Guo Y, Liu X, Yu L, Zhong X, Ran M, Wang B, Tang Y, Lee RJ, Xie J. ROS-Responsive Biomimetic Nanocomplexes of Liposomes and Macrophage-Derived Exosomes for Combination Breast Cancer Therapy. Int J Nanomedicine 2025; 20:5161-5180. [PMID: 40297405 PMCID: PMC12036690 DOI: 10.2147/ijn.s514375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Purpose Breast cancer is the most diagnosed cancer in women globally and it poses a major threat to women's lives and health. As an essential therapeutic approach for breast cancer, chemotherapy encounters various clinical challenges like multidrug resistance and systemic toxicity. Nanotechnology has shown progress in addressing chemotherapy drug limitations. However, externally introduced nanoparticles are typically captured by the mononuclear phagocyte system (MPS) post-administration. To mitigate chemotherapy drug toxicity and enhance drug delivery efficiency, we combined ROS-responsive cationic liposomes (cLip) with macrophage-derived exosomes to create biomimetic nanocomplex (E-cLip-DTX/si) for co-delivery docetaxel (DTX) and Bcl-2 siRNA. Methods We encapsulated docetaxel (DTX) and Bcl-2 siRNA as model drugs into biomimetic nanocomplexes and validated their antitumor efficacy in vitro and in vivo. Results In vitro and vivo tests show that E-cLip-DTX/si can react to ROS, promote apoptosis of tumor cells effectively, and prolong circulation time. In breast cancer mouse model, E-cLip-DTX/si displays notable tumor accumulation efficiency, remarkable anti-tumor effects, and a favorable safety profile. Conclusion We have developed a ROS-responsive biomimetic nanocomplexes that efficiently delivers DTX and Bcl-2 siRNA into the tumor site, overcoming the MPS barrier and extending the blood circulation time of the drug. Hence, biomimetic nanocomplex is a promising drug delivery platform with controlled drug release and biocompatibility for effective anti-tumor treatment.
Collapse
Affiliation(s)
- Minhao Xu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Lu Bai
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Meng Sun
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Xinlei Yan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Ying Xiong
- Enrollment and Employment Department, Alumni-Office, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Yu Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Yue Guo
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Xingyou Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Leijie Yu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Xing Zhong
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Mengqiong Ran
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Ben Wang
- Chengcheng County Hospital, Weinan, 715200, People’s Republic of China
| | - Yaqin Tang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| | - Robert J Lee
- College of Pharmacy, The Ohio State University, Columbus, OH, 43210-1291, USA
| | - Jing Xie
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People’s Republic of China
| |
Collapse
|
43
|
Lu Y, Jiang X, Yang B, Ding M, Shen Y, Jin J, Yu J, Lu W, Chen Y, Zhu S. Comparative Study on Covalent and Noncovalent Endogenous Albumin-Binding β-Glucuronidase-Activated SN38 Prodrugs for Antitumor Efficacy. J Med Chem 2025; 68:8361-8376. [PMID: 40189819 DOI: 10.1021/acs.jmedchem.4c03096] [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: 04/25/2025]
Abstract
Albumin-binding prodrugs have been explored to overcome the limitations of small-molecule anticancer chemotherapy agents, such as inadequate physiological and pharmaceutical compatibility, as well as rapid renal clearance. Herein, we investigated two endogenous albumin-binding prodrugs, M-g-SN38 and S-g-SN38, forming macromolecular conjugates. Both prodrugs exhibited robust stability in murine and human plasma, crucial for their therapeutic potential. Selective activation by β-glucuronidase ensures minimal toxicity in their inactive state. Notably, M-g-SN38 exhibited higher cellular uptake, a longer circulation half-life, and enhanced tumor accumulation compared to S-g-SN38, suggesting its greater potential for improved antitumor efficacy. In vivo, M-g-SN38 exhibited significant antitumor activity, leading to profound tumor reduction and, in many cases, marked depletion and complete eradication in all treated mice in the HCT116 xenograft model. Furthermore, M-g-SN38 also demonstrated pronounced antitumor efficacy in the BxPC-3 xenograft model. Together, these findings provide new insights for the development of albumin-binding prodrugs.
Collapse
Affiliation(s)
- Yingxin Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Xing Jiang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Biyu Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Mengyuan Ding
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Yanyan Shen
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Jiyu Jin
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Jiahui Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Yi Chen
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, PR China
| | - Shulei Zhu
- Innovation Center for AI and Drug Discovery, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
- ATLATL Innovation Center, 1077 Zhangheng Road, Shanghai 201203, PR China
| |
Collapse
|
44
|
Varela-Quitián YF, Mendez-Rivera FE, Bernal-Estévez DA. Cationic antimicrobial peptides: potential templates for anticancer agents. Front Med (Lausanne) 2025; 12:1548603. [PMID: 40342581 PMCID: PMC12058764 DOI: 10.3389/fmed.2025.1548603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/07/2025] [Indexed: 05/11/2025] Open
Abstract
Cancer is a major global health concern and one of the leading causes of death worldwide. According to the World Health Organization (WHO), there is an urgent need for novel therapeutic agents to treat this disease. Some antimicrobial peptides (AMPs) have demonstrated activity against both microbial pathogens and cancer cells. Among these, cationic AMPs (CAMPs) have garnered significant attention because of their ability to selectively interact with the negatively charged surfaces of cancer cell membranes. CAMPs present several advantages such as high specificity for targeting cancer cells, minimal toxicity to normal cells, reduced probability of inducing resistance, stability under physiological conditions, ease of chemical modification, and low production costs. This review focuses on CAMPs with anticancer properties such as KLA, bovine lactoferricin derivatives, and LTX-315, and briefly explores common bioinformatics tools for Anticancer Peptides (ACPs) selection pipeline from AMPs.
Collapse
|
45
|
Mlilo S, Sibanda S, Sithole S, Mukanganyama S, Naik YS. Evaluation of the antiproliferative, cytotoxic and phytochemical properties of Zimbabwean medicinal plants used in cancer treatment. BMC Complement Med Ther 2025; 25:156. [PMID: 40275320 PMCID: PMC12023620 DOI: 10.1186/s12906-025-04883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 04/03/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Cancer cases have been on the rise globally and several treatment strategies have been developed but mortality rates remain high. Zimbabwe, like many other countries, has also experienced a surge in cancer cases. In Zimbabwe, medicinal plants have been widely used to treat cancer for centuries. However, there has been limited research on the effectiveness, safety, and chemical composition of these plants. The current study assessed antiproliferative, cytotoxic and phytochemical properties of selected Zimbabwean medicinal plants. METHOD Cytotoxic activity of Agelenthus pungu, Carissa edulis, Dombeya rotundifolia, Flacourtia indica, Lannea discolor, Leonotis ocymifolia, Leucas martinicensis, Plicosepalus kalachariensis, Pseudolachnostylis maproneifolia, Solanum incanum, Strychnos cocculoides, Strychnos spinosa and Viscum verrucosum extracts were evaluated on normal murine peritoneal cells and sheep erythrocytes while antiproliferative activity was assessed on Jurkat T and HL60 cell lines. Cell viability was determined using the trypan blue exclusion and sulforhodamine B assay. Additionally, the effect of reduced glutathione on cytotoxic extracts was examined. The phytochemicals of the methanolic extracts were qualitatively determined using standard methods. RESULTS Agelenthus pungu, Carissa edulis, Flacourtia indica, Strychnos cocculoides, Strychnos spinosa and Viscum verrucosum were cytotoxic to normal murine peritoneal cells. Flacourtia indica and Viscum verruscosum caused haemolysis of sheep erythrocytes at a concentration of 250 µg/mL for both plant extracts and 125 µg/mL for Viscum verrucosum. Cell viability increased on addition of 25 µg/mL of reduced glutathione to the extracts considered the most cytotoxic extracts, Agelenthus pungu and Viscum verrucosum. Agelenthus pungu, Carissa edulis, Leonotis ocymifolia, Leucas martinicensis and Viscum verrucosum significantly inhibited Jurkat T and HL60 cell proliferation. Viscum verrucosum was the most potent with the lowest half-maximum inhibitory concentration (IC50) values of 33 and 34 µg/mL on Jurkat T and HL60 cell lines respectively. The most dominant phytochemical classes were alkaloids, flavonoids and saponins. CONCLUSION This study demonstrates that Agelenthus pungu, Carissa edulis, Leonotis ocymifolia, Leucas martinicensis and Viscum verrucosum have antiproliferative activity against Jurkat T and HL60 cell lines. Viscum verrucosum was the most potent. These findings emphasise the importance of medicinal plants as well as their potential use as sources of novel compounds in anticancer drug discovery.
Collapse
Affiliation(s)
- Sigcono Mlilo
- Department of Applied Chemistry, National University of Science and Technology (NUST), P.O.Box AC 939, Ascot, Bulawayo, Zimbabwe.
| | - Samson Sibanda
- Department of Applied Chemistry, National University of Science and Technology (NUST), P.O.Box AC 939, Ascot, Bulawayo, Zimbabwe
| | - Simbarashe Sithole
- Department of Soil Science and Productivity, Sciences and Technology (MUAST), Marondera University of Agricultural, P.O Box 35, Marondera, Zimbabwe
| | - Stanley Mukanganyama
- African Institute of Biomedical Science and Technology 911 Boronia Farm, Beatrice, Zimbabwe
| | - Yogehkumar S Naik
- Environmental Science, National University of Science and Technology (NUST), Bulawayo, Zimbabwe
| |
Collapse
|
46
|
Armah J, Alzahid SK, Pei Q, Stacciarini JMR, Heldermon C, Starkweather A. Exercise to Manage Fatigue During and After Chemotherapy in Adolescents and Young Adults With Cancer: A Systematic Review and Meta-Analysis. Oncol Nurs Forum 2025; 52:E77-E92. [PMID: 40293930 PMCID: PMC12056843 DOI: 10.1188/25.onf.e77-e92] [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: 07/01/2024] [Accepted: 10/02/2024] [Indexed: 04/30/2025]
Abstract
PROBLEM IDENTIFICATION There is a gap in knowledge about the efficacy of exercise in managing cancer-related fatigue (CRF) in adolescents and young adults (AYAs) during and after chemotherapy. LITERATURE SEARCH A systematic search was conducted in Scopus®, PubMed®, and CINAHL®, as well as citation searching, for studies about the impact of exercise on CRF in the AYA population. Abstract and full-text screening of 2,234 studies produced 15 studies for systematic review and 13 for meta-analysis. DATA EVALUATION Effect size was calculated using standardized mean difference and confidence intervals. Subgroup analysis was conducted to assess the impact of various exercise types on CRF. A meta-regression was performed using exercise frequency, intensity, and duration as predictors to determine relationships with effect size on CRF and adherence to exercise. SYNTHESIS Exercise had a large effect size on CRF in AYAs. No difference in effect size estimates for exercise type subgroups was observed. A linear association was found between exercise duration and CRF (p = 0.005), and exercise intensity and adherence (p = 0.037). IMPLICATIONS FOR PRACTICE Exercise is effective in managing CRF among AYAs, and efforts should be aimed toward including it in routine oncologic care.
Collapse
|
47
|
Pallavi R, Soni BL, Jha GK, Sanyal S, Fatima A, Kaliki S. Tumor heterogeneity in retinoblastoma: a literature review. Cancer Metastasis Rev 2025; 44:46. [PMID: 40259075 PMCID: PMC12011974 DOI: 10.1007/s10555-025-10263-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/06/2025] [Indexed: 04/23/2025]
Abstract
Tumor heterogeneity, characterized by the presence of diverse cell populations within a tumor, is a key feature of the complex nature of cancer. This diversity arises from the emergence of cells with varying genomic, epigenetic, transcriptomic, and phenotypic profiles over the course of the disease. Host factors and the tumor microenvironment play crucial roles in driving both inter-patient and intra-patient heterogeneity. These diverse cell populations can exhibit different behaviors, such as varying rates of proliferation, responses to treatment, and potential for metastasis. Both inter-patient heterogeneity and intra-patient heterogeneity pose significant challenges to cancer therapeutics and management. In retinoblastoma, while heterogeneity at the clinical presentation level has been recognized for some time, recent attention has shifted towards understanding the underlying cellular heterogeneity. This review primarily focuses on retinoblastoma heterogeneity and its implications for therapeutic strategies and disease management, emphasizing the need for further research and exploration in this complex and challenging area.
Collapse
Affiliation(s)
- Rani Pallavi
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, Telangana, India.
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India.
| | - Bihari Lal Soni
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, Telangana, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Gaurab Kumar Jha
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, Telangana, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Shalini Sanyal
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, Telangana, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Azima Fatima
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, Telangana, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Swathi Kaliki
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, Telangana, India.
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, Telangana, India.
| |
Collapse
|
48
|
Zhang J, Yang H, Li L, Peng C, Li J. Noninvasive Transdermal Delivery of STING Agonists Reshapes the Immune Microenvironment of Melanoma and Potentiates Checkpoint Blockade Therapy Efficacy. ACS APPLIED BIO MATERIALS 2025; 8:3156-3166. [PMID: 40191891 DOI: 10.1021/acsabm.4c02004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The emergence of immunotherapy as a revolutionary therapeutic modality has fostered confidence and underscored its potent efficacy in tumor therapy. However, enhancing the therapeutic efficacy of immunotherapy by precise and judicious administration poses a significant challenge. In this context, we have developed a disulfide-bearing transdermal nanovaccine by integrating a thiol-reactive agent lipoic acid (LA) into a metal-coordinated cyclic dinucleotide nanoassembly, designated as LA-Mn-cGAMP (LMC) nanovaccines. Upon topical application to the skin with melanoma, the dithiolane moiety of LA enables thiol-disulfide dynamic exchange in the skin, hence facilitating penetration into both the skin and subcutaneous tumor tissues via the thiol-mediated uptake (TMU) mechanism. Our findings demonstrate that transdermal administration of LMC significantly enhances STING activation, mitigates the immunosuppressive tumor microenvironment (TME), and retards melanoma progression. Moreover, the remodeled TME amplifies the efficacy of immune checkpoint inhibitors. This advancement offers an administration strategy for existing STING agonist therapy, potentially improving the biosafety of immunotherapy.
Collapse
Affiliation(s)
- Junjie Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Hui Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Liang Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, China
| | - Changkun Peng
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
| | - Jingying Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 3501116, China
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, China
| |
Collapse
|
49
|
Corsi M, Maurina E, Surdo S, Vandini E, Daini E, Vilella A, Leo G, Farshchian M, Grisendi G, Golinelli G, Dominici M, Bocci G, Giuliani D, Barillaro G. In vivo and in situ monitoring of doxorubicin pharmacokinetics with an implantable bioresorbable optical sensor. SCIENCE ADVANCES 2025; 11:eads0265. [PMID: 40238874 PMCID: PMC12002126 DOI: 10.1126/sciadv.ads0265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/11/2025] [Indexed: 04/18/2025]
Abstract
Cancer treatment, particularly chemotherapy, requires balancing efficacy and toxicity. Although traditional monitoring methods can lead to suboptimal outcomes, emerging implantable chemical sensors can complement them by providing precise, real-time drug monitoring at tumor sites, although the technology remains in its early stages. Here, we introduce an ultrathin, bioresorbable implantable biosensor for real-time doxorubicin monitoring in vivo with high spatiotemporal resolution. The sensor amplifies the drug's fluorescence, enabling successful tracking of doxorubicin through the skin in live mice following intravenous injection. When paired with a reusable electronic patch, the biosensor facilitates seamless data collection and wireless transmission. A 3-month biocompatibility study, including systemic toxicity assessments, histological and blood analyses, confirms complete biodegradation with no observed toxicity. By directly measuring chemotherapeutic drug levels in tissues over time, our sensor enhances traditional monitoring methods, enabling clinicians to optimize dosing during cancer treatment and reduce the risk of locoregional recurrence following tumor removal.
Collapse
Affiliation(s)
- Martina Corsi
- Department of Information Engineering, University of Pisa, via G. Caruso 16, 56122 Pisa, Italy
| | - Elena Maurina
- Department of Information Engineering, University of Pisa, via G. Caruso 16, 56122 Pisa, Italy
| | - Salvatore Surdo
- Department of Information Engineering, University of Pisa, via G. Caruso 16, 56122 Pisa, Italy
| | - Eleonora Vandini
- Department of Biomedical Metabolic and Neural Sciences, University of Modena and Reggio Emilia, via G. Campi 287, 41125 Modena, Italy
| | - Eleonora Daini
- Department of Biomedical Metabolic and Neural Sciences, University of Modena and Reggio Emilia, via G. Campi 287, 41125 Modena, Italy
| | - Antonietta Vilella
- Department of Biomedical Metabolic and Neural Sciences, University of Modena and Reggio Emilia, via G. Campi 287, 41125 Modena, Italy
| | - Giuseppina Leo
- Department of Biomedical Metabolic and Neural Sciences, University of Modena and Reggio Emilia, via G. Campi 287, 41125 Modena, Italy
| | - Moein Farshchian
- Department of Medical and Surgical Science for Children and Adults, University Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41124 Modena, Italy
| | - Giulia Grisendi
- Department of Medical and Surgical Science for Children and Adults, University Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41124 Modena, Italy
| | - Giulia Golinelli
- Department of Medical and Surgical Science for Children and Adults, University Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41124 Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Science for Children and Adults, University Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41124 Modena, Italy
| | - Guido Bocci
- Department of Translational Research and New Technologies in Medicine and Surgery School of Medicine, University of Pisa, Via Risorgimento 36, 56126 Pisa, Italy
| | - Daniela Giuliani
- Department of Biomedical Metabolic and Neural Sciences, University of Modena and Reggio Emilia, via G. Campi 287, 41125 Modena, Italy
| | - Giuseppe Barillaro
- Department of Information Engineering, University of Pisa, via G. Caruso 16, 56122 Pisa, Italy
| |
Collapse
|
50
|
Zhou Y, Camisasca A, Dominguez-Gil S, Bartkowski M, Rochfort KD, Piletti M, White A, Krizsan D, O'Connor R, Quinn SJ, Iacopino D, Eustace AJ, Giordani S. Synthesis of carbon dots from spent coffee grounds: transforming waste into potential biomedical tools. NANOSCALE 2025; 17:9947-9962. [PMID: 40067158 DOI: 10.1039/d4nr05186f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Carbon dots (CDs) are small-sized, spherical nanoparticles presenting amorphous carbon cores with nanocrystalline regions of a graphitic structure. They show unique properties such as high aqueous solubility, robust chemical inertness, and non-toxicity and can be manufactured at a relatively low cost. They are also well known for outstanding fluorescence tunability and resistance to photobleaching. Together, these properties boost their potential to act as drug delivery systems (DDSs). This work presents a low-cost synthesis of CDs by upcycling spent coffee grounds (SCGs) and transforming them into value-added products. This synthetic route eliminates the use of highly toxic heavy metals, high energy-consuming reactions and long reaction times, which can improve biocompatibility while benefiting the environment. A series of physico-chemical characterisation techniques demonstrated that these SCG-derived CDs are small-sized nanoparticles with tunable fluorescence. In vitro studies with 120 h of incubation of SCG-derived CDs demonstrated their specific antiproliferative effect on the breast cancer CAL-51 cell line, accompanied by increased reactive oxygen species (ROS) production. Importantly, no impact was observed on healthy breast, kidney, and liver cells. Confocal laser scanning microscopy confirmed the intracellular accumulation of SCG-derived CDs. Furthermore, the drug efflux pumps P-glycoprotein (P-gp) and the breast cancer resistance protein (BCRP) did not impact CD accumulation in the cancer cells.
Collapse
Affiliation(s)
- Yingru Zhou
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Adalberto Camisasca
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Sofia Dominguez-Gil
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Michał Bartkowski
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
| | - Keith D Rochfort
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Martina Piletti
- Tyndall National Institute, University College Cork, Cork, Ireland
| | - Anita White
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Dorottya Krizsan
- School of Chemistry, University College Dublin, Belfield, Dublin, Ireland
| | - Robert O'Connor
- School of Physical Sciences, Dublin City University, Glasnevin, Dublin, Ireland
| | - Susan J Quinn
- School of Chemistry, University College Dublin, Belfield, Dublin, Ireland
| | - Daniela Iacopino
- Tyndall National Institute, University College Cork, Cork, Ireland
| | - Alex J Eustace
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
- School of Biotechnology, Dublin City University, Glasnevin, Dublin, Ireland
| | - Silvia Giordani
- School of Chemical Science, Dublin City University, Glasnevin, Dublin, Ireland.
- Life Sciences Institute, Dublin City University, Glasnevin, Dublin, Ireland.
| |
Collapse
|