1
|
Zhang M, Zhang R, Feng C, Jiang X, Xu X, Wang J. Ginsenoside compound K-based multifunctional liposomes for the treatment of rheumatoid arthritis. Drug Deliv 2025; 32:2464190. [PMID: 39957241 PMCID: PMC11834820 DOI: 10.1080/10717544.2025.2464190] [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: 06/18/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/18/2025] Open
Abstract
The clinical treatment of rheumatoid arthritis (RA) with first-line therapeutic drugs is hindered by the poor solubility, low bioavailability, off-target toxicity, and insufficient accumulation in inflamed joints. Liposomes have been shown to mitigate some of these limitations in drug delivery systems. However, the use of cholesterol to stabilize liposomal structures remains controversial due to its potential association with cardiovascular diseases. Here, we developed a novel liposome based on ginsenoside compound K (CK), which not only serves as an effective therapeutic agent for RA but also replaces cholesterol as a membrane stabilizer to address these challenges. Compared with conventional liposomes, ginsenoside CK Liposomes (CK@Lipo) are excellent nanoparticles, with CK stabilizing the liposomal structure and providing targeting functionality toward inflamed joints. When encapsulated with dexamethasone (Dex), CK@Lipo exhibits a synergistic anti-inflammatory effect, slowing the progression of RA. This study provides a theoretical basis for the future development of multifunctional novel ginsenoside CK@Lipo.
Collapse
Affiliation(s)
- Meng Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ru Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
| | - Chunbo Feng
- R&D Center, Shanghai Jahwa United Co., Ltd., Shanghai, China
| | - Xinnan Jiang
- R&D Center, Shanghai Jahwa United Co., Ltd., Shanghai, China
| | - Xinchun Xu
- Shanghai Xuhui Central Hospital, Xuhui Hospital Attached to Fudan University, Shanghai, China
| | - Jianxin Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
| |
Collapse
|
2
|
Wu Y, He L, Zhao S, Jiang Y, Yang Z, Deng X. Tumor microenvironment pH-responsive size-transformable peptide self-assembling nanocarriers for tumor-specific treatment. BIOMATERIALS ADVANCES 2025; 173:214293. [PMID: 40168894 DOI: 10.1016/j.bioadv.2025.214293] [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: 01/24/2025] [Revised: 03/11/2025] [Accepted: 03/22/2025] [Indexed: 04/03/2025]
Abstract
Peptide-based drug carriers with exceptional biodegradability offer promising avenues for tumor-targeted therapy. Nonetheless, almost all existing drug carriers harness receptor recognition to target tumors, which ultimately fall short in addressing tumor heterogeneity. Such a strategy requires intricate chemical modifications for carriers to selectively bind to specific receptors. While these modifications may induce long-term toxicity, tumor receptors are not absolutely specific but also exist in normal cells. Thus, precision therapeutic agents may inadvertently harm healthy cells as well. Tumors possess a distinctive weak acidic (pH 6.0-6.8) tumor microenvironment (TME) that contrasts with normal tissues (pH ~7.4). Hence, we developed a TME pH-triggered multilevel self-assembling peptide with simple modifications. The drug-encapsulating self-assembled peptide is size transformable from aggregates (~1.56 μm) at pH 7.4 to positively charged nanomicelles (~100 nm) at an acidic TME by protonation, which avoids being taken up by normal cells but could readily enter tumor cells, allowing TME pH-triggered tumor-specific therapy. This study establishes a breaking strategy of using peptide for TME-based tumor-specific treatment and advances the medical applications of peptide nanomaterials.
Collapse
Affiliation(s)
- Yuhan Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Li He
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Shoubo Zhao
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Yuqiu Jiang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Zuojun Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics and Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
3
|
Ghosh R, Kumar M, Kumar S, Komal K, Sharma R, Kurmi BD. Small molecule therapeutics for receptor-mediated targeting through liposomes in breast cancer treatment: A comprehensive review. Bioorg Chem 2025; 160:108442. [PMID: 40199009 DOI: 10.1016/j.bioorg.2025.108442] [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: 01/24/2025] [Revised: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Breast cancer (BC) remains a significant global health challenge, with conventional treatment approaches such as surgery, chemotherapy, and radiation therapy. These approaches face limitations in targeting, toxicity, and efficacy. Liposomal drug delivery systems have emerged as promising tools for targeted breast cancer therapies. Liposomes can encapsulate both hydrophilic and hydrophobic drugs, improve drug distribution, and reduce the side effects. Passive targeting exploits the enhanced permeability and retention effect in tumor tissues, whereas active targeting employs small molecule ligands such as aptamers, folic acid (FA), transferrin, and monoclonal antibodies to specifically bind to overexpressed receptors on cancer cells. Aptamer-functionalized liposomes exhibit high specificity and affinity, folate and transferrin receptor targeting enhances cellular uptake and cytotoxicity, and antibody-conjugated liposomes improve drug delivery and efficacy by targeting specific antigens. Dual-responsive liposomes are sensitive to multiple stimuli and further enhance targeting precision. However, challenges remain, including tumor heterogeneity, limited penetration, and potential immunogenicity. Current research has focused on developing stable and effective formulations and exploring combination-targeting strategies to overcome these limitations. With further advancements, targeted liposomal drug delivery systems hold great promise in improving breast cancer treatment outcomes and reducing adverse effects.
Collapse
Affiliation(s)
- Rashmi Ghosh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India.
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Rohit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| |
Collapse
|
4
|
Rawat E, Sharma S, Vyas S, Alsaidan OA, Kapoor DU, Prajapati BG. Advances in alginate-based nanoformulations: Innovative and effective strategies for targeting and treating brain disorders. Int J Pharm 2025; 681:125851. [PMID: 40516772 DOI: 10.1016/j.ijpharm.2025.125851] [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: 04/01/2025] [Revised: 05/27/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Brain disorders, encompassing neurodegenerative conditions and intracranial neoplasms, present formidable obstacles in the realm of pharmacological delivery due to the existence of athe blood-brain barrier (BBB) and the restricted bioavailability of therapeutic agents. Alginate-derived nanoformulations have emerged as highly promising systems for drug delivery, offering attributes such as biocompatibility, regulated release, and improved targeting efficacies. This review investigates contemporary advancements in alginate-based nanoformulations, with a particular emphasis on their efficacy in surmounting obstacles to successful pharmacological delivery to the brain. Initially, we furnish a comprehensive overview of alginate, underscoring its pertinent properties, biomedical applications, and inherent limitations. Subsequently, the discourse progresses to strategies for nanoformulation, which encompass lipid-based, polymeric, and inorganic methodologies, with a focus on their benefits in relation to cerebral targeting. Moreover, this review entails the therapeutic potential of alginate-based nanoformulations in addressing significant neurological disorders, including Alzheimer's disease, Parkinson's disease, brain tumours, traumatic brain injury, epilepsy, and amyotrophic lateral sclerosis. By amalgamating cutting-edge nanotechnology with the distinctive properties of alginate, these formulations signify a promising pathway for the advancement of efficacious therapies aimed at brain targeting. Additionally, prospective research trajectories and challenges associated with the optimization of alginate-based nanocarriers for clinical applications are also elucidated.
Collapse
Affiliation(s)
- Ekta Rawat
- Manav Rachna University, Faridabad 121004 Haryana, India
| | - Suman Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022 Rajasthan, India
| | - Shweta Vyas
- Department of Pure & Applied Chemistry, University of Kota, Kota 324005 Rajasthan, India
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia 72341
| | - Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Bardoli 394601 Gujarat, India.
| | - Bhupendra G Prajapati
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401 Punjab, India; Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
5
|
Li MY, Wu LM, Xu MJ, Shen ML, Chen JH, Wu QJ, Wang R, Zeng Y, Wu YG, Si HB, Chen XT, Li H, Gong CY, Xie HQ, Shen B. Biomimetic Liposomes for G-Protein-Coupled Receptor 120 Exo/Endogenous Coactivation to Reprogram Lipid Metabolism in Obesity-Associated Osteoarthritis. ACS NANO 2025; 19:20711-20728. [PMID: 40445329 PMCID: PMC12164828 DOI: 10.1021/acsnano.5c02515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 06/11/2025]
Abstract
Osteoarthritis (OA) associated with obesity is increasingly recognized as a distinct phenotype, driven by lipid metabolic imbalance and related inflammation. A particularly troublesome issue is that even after successfully correcting obesity, OA progression and lipid metabolic imbalance persist within the joint microenvironment, suggesting local lipid metabolism regulation as a potential treatment option. G-protein-coupled receptor 120 (GPR120), a primary receptor for long-chain fatty acids (including docosahexaenoic acid, DHA), has recently been found to play a pivotal role in regulating lipid homeostasis and suppressing inflammation. Here, we present ChD-FL/sgGPR, enabling dual endogenous-exogenous GPR120 activation. ChD-FL/sgGPR is a chondrocyte-biomimetic, fluorinated phenylboronic acid (FPBA)-modified ionizable liposome that codelivers DHA and a CRISPRa system comprising GPR120-specific sgRNA (single guide RNA) and dCas9-VPR mRNA (dead Cas9 fused to VP64-p65-Rta activator domain). Specifically, FPBA modification of liposomes enhances lysosomal escape and nuclear entry of RNA, while coextrusion with chondrocyte membranes facilitates cartilage-targeted delivery. In the coculture system of adipocytes and OA chondrocytes, ChD-FL/sgGPR significantly boosts chondrocytes GPR120 expression, facilitates lipid clearance via PPARγ signaling, and diminishes inflammatory mediators. In obese rat OA models, intra-articular injection of ChD-FL/sgGPR prolongs local retention, inhibits cartilage catabolism, and mitigates subchondral bone deterioration, collectively decelerating OA progression. By integrating CRISPR-mediated gene upregulation with DHA-induced receptor stimulation, this platform rebalances lipid metabolism in OA cartilage, offering a promising, mechanism-driven therapy for obesity-associated OA.
Collapse
Affiliation(s)
- Ming-Yang Li
- Department
of Orthopedic Surgery and Orthopedic Research Institute, West China
Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Li-Min Wu
- Department
of Orthopedic Surgery and Orthopedic Research Institute, West China
Hospital, Sichuan University, Chengdu, Sichuan610041, China
- Department
of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy,
West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Ming-Jie Xu
- Core
Facilities, West China Hospital, Sichuan
University, Chengdu, Sichuan610041, China
| | - Mei-Ling Shen
- Department
of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy,
West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Jie-Hao Chen
- Animal
Laboratory Center, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Qin-Jie Wu
- Department
of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy,
West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Rui Wang
- Animal
Laboratory Center, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Yi Zeng
- Department
of Orthopedic Surgery and Orthopedic Research Institute, West China
Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Yuan-Gang Wu
- Department
of Orthopedic Surgery and Orthopedic Research Institute, West China
Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Hai-Bo Si
- Department
of Orthopedic Surgery and Orthopedic Research Institute, West China
Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Xiao-Ting Chen
- Animal
Laboratory Center, West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Hang Li
- Department
of Orthopedic Surgery and Orthopedic Research Institute, West China
Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Chang-Yang Gong
- Department
of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy,
West China Hospital, Sichuan University, Chengdu, Sichuan610041, China
| | - Hui-Qi Xie
- Department
of Orthopedic Surgery and Orthopedic Research Institute, West China
Hospital, Sichuan University, Chengdu, Sichuan610041, China
- Stem
Cell and Tissue Engineering Research Center, State Key Laboratory
of Biotherapy, West China Hospital, Sichuan
University, Chengdu, Sichuan610041, China
| | - Bin Shen
- Department
of Orthopedic Surgery and Orthopedic Research Institute, West China
Hospital, Sichuan University, Chengdu, Sichuan610041, China
| |
Collapse
|
6
|
Keskin H, Reçber T, Filazi N, Gelen-Gungor D, Ozturk S, Eroğlu H, Nemutlu E, Özkul A, Ulubayram K, Eroğlu İ. Development of molnupiravir and peramivir loaded liposome formulations for combined antiviral therapy. Pharm Dev Technol 2025:1-21. [PMID: 40470763 DOI: 10.1080/10837450.2025.2516239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 05/20/2025] [Accepted: 06/02/2025] [Indexed: 06/11/2025]
Abstract
The pandemic caused by the SARS-CoV-2 virus has led scientists to intensify research on antiviral drugs and vaccines. As a result of these studies, it was observed that molnupiravir (MLP) and peramivir (PRV) could be used against pandemic. MLP affects SARS-CoV-2 replication, but it necessitates high doses, which can cause adverse effects in patients. PRV is a neuraminidase inhibitor, but the bioavailability of the drug after oral administration is very low. In this study, MLP-, PRV-loaded and combined liposome (COMB-Lipo) formulations were prepared via the thin film hydration method. Phospholipon 90 G-based formulations exhibited the most favorable characteristics, with a particle size of 111-145 nm, a polydispersity index (PDI) of less than 0.4, and a zeta potential (ZP) of 6-12 mV). Cell culture studies demonstrated that developed stable formulations are nontoxic to L929 and Vero E6 cells. Antiviral activity assessments against SARS-CoV-2 suggested the effectiveness of liposomes in inhibiting viral activity. These findings demonstrate that a possible synergistic effect of the newly developed sustained-release COMB-Lipo formulation is suggested with the complementary antiviral mechanisms of the combined agents. As a result, the therapeutic potential of co-delivery of anti-SARS-CoV-2 drugs for pulmonary application is considered a promising approach for long-acting treatment of COVID-19.
Collapse
Affiliation(s)
- Hadiye Keskin
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Nazlıcan Filazi
- Department of Virology, Ankara University, Ankara, Turkey
- Department of Virology, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Dilek Gelen-Gungor
- Department of Forensic Sciences, Turkish National Police Academy, Ankara, Turkey
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sukru Ozturk
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Hakan Eroğlu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
- IPQ Pharma Medicines and Medical Devices Ltd, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
- Department of Vaccine Technology, Vaccine Institute, Hacettepe University, Ankara, Turkey
| | - Aykut Özkul
- Department of Virology, Ankara University, Ankara, Turkey
| | - Kezban Ulubayram
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - İpek Eroğlu
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
7
|
Zhang D, Zhang H, Fu M, Liu J, Zheng N, Shao X, Ji X. Isotropic Liposomal Microreactor for Catalytic Oxidative Desulfurization. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025. [PMID: 40490930 DOI: 10.1021/acs.langmuir.5c01543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The efficient removal of refractory sulfides from fuels to achieve clean oil is a primary research focus in the petrochemical industry. This study introduces extraction and catalytic oxidation desulfurization (ECODS) as a technique for effective desulfurization. A cross-linking strategy was employed to construct structurally stable isotropic microreactors based on ionic liquid (IL)-modified liposomes (poly[MimA11, A11][heteropolyanions]), where exposed imidazolium cations anchor catalytic heteropolyanions (e.g., [PW12O40]3-) to ensure active site accessibility. In this interfacial catalytic reaction, the microreactor resembles an emulsified droplet with a spherical surface, significantly enhancing the catalytic interface. Additionally, the isotropic imidazolium cations of spherical vesicles provide the equivalent driving force for the attachment of heteropolyanions (such as [PW12O40]3- and [PMo12O40]3-), ensuring full exposure of active sites and reducing mass transfer resistance. The optimized poly[MimA11, A11][PW12O40] catalyst achieved complete dibenzothiophene (DBT) removal within 1.5 h and retained 92.4% efficiency after six cycles, demonstrating exceptional activity and recyclability. The morphology, structure, and properties of the microreactors were characterized, and optimal reaction conditions were established. Building on this foundation, the removal performances across various systems and sulfur-containing targets were assessed, thereby confirming the structure-activity relationship of this type of microreactor. Furthermore, the desulfurization mechanism was inferred from the identified oxidation products. Overall, this isotropic poly[MimA11, A11][heteropolyanion] demonstrates excellent desulfurization performance and holds significant potential for broad applications.
Collapse
Affiliation(s)
- Duo Zhang
- School of Chemical and Environment Science, Shaanxi Key Laboratory of Catalysis, Shaanxi University of Technology, Hanzhong 723001, China
| | - Hairan Zhang
- School of Chemical and Environment Science, Shaanxi Key Laboratory of Catalysis, Shaanxi University of Technology, Hanzhong 723001, China
| | - Mengmei Fu
- School of Chemical and Environment Science, Shaanxi Key Laboratory of Catalysis, Shaanxi University of Technology, Hanzhong 723001, China
| | - Jie Liu
- School of Chemical and Environment Science, Shaanxi Key Laboratory of Catalysis, Shaanxi University of Technology, Hanzhong 723001, China
| | - Nan Zheng
- School of Chemical and Environment Science, Shaanxi Key Laboratory of Catalysis, Shaanxi University of Technology, Hanzhong 723001, China
| | - Xianzhao Shao
- School of Chemical and Environment Science, Shaanxi Key Laboratory of Catalysis, Shaanxi University of Technology, Hanzhong 723001, China
| | - Xiaohui Ji
- School of Chemical and Environment Science, Shaanxi Key Laboratory of Catalysis, Shaanxi University of Technology, Hanzhong 723001, China
| |
Collapse
|
8
|
Abadi SK, Peyvandi K, Shariat S. Investigation of the preparation of iron sulfate-loaded niosomes by an experimental novel method. Sci Rep 2025; 15:19965. [PMID: 40481054 PMCID: PMC12144141 DOI: 10.1038/s41598-025-04305-9] [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: 01/27/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Iron is a crucial nutrient for our bodies, and its absence can lead to serious health issues like anemia. Unfortunately, oral iron supplements are frequently poorly absorbed and can lead to gastrointestinal discomfort due to iron's oxidative properties. However, using nanocarrier systems to encapsulate iron can mitigate these issues, enhancing its absorption and protecting it from oxidation, ultimately improving its effectiveness. In this research, niosomes loaded with iron sulfate were prepared using surfactant span 60 and Tween 80 via thin film hydration method. Cholesterol and 1-Dodecanol were used as stabilizers in different ratios. Physiochemical properties of niosomes loaded with iron sulfate, such as particle size, polydispersity index (PDI), zeta potential, and encapsulation efficiency (EE%), were investigated. The vesicle sizes varied between 453 and 3,276 nm, with encapsulation efficiencies ranging from 75 to 93%. The zeta potential measured between - 9.91 and 6.3 mV, while the particle size distribution (PDI) was observed to be between 0.004 and 0.59 in a phosphate buffer solution at pH 6.8. Ultimately, F1 Formula was established as the most effective formula, achieving an efficiency of 90.8%, a particle size of 546.8 nm, a zeta potential of 5.5, and a PDI of 0.57.
Collapse
Affiliation(s)
- Samira Khalil Abadi
- Faculty of Chemical, Petroleum and Gas Engineering, Semnan University, Semnan, Iran
| | - Kiana Peyvandi
- Faculty of Chemical, Petroleum and Gas Engineering, Semnan University, Semnan, Iran.
| | - Sheida Shariat
- Department of Pharmacy, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
9
|
Cheng D, Lian W, Jia X, Wang T, Sun W, Jia Z, Liu Y, Ni C. Senescent endothelial cell-derived Galectin 3 promotes silicosis through endothelial-fibroblast and endothelial-macrophage crosstalk. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137605. [PMID: 39955992 DOI: 10.1016/j.jhazmat.2025.137605] [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: 08/30/2024] [Revised: 01/03/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Silicosis is an occupational and irreversible interstitial lung disease, which is caused by the inhalation of respirable crystalline silica. Recent studies suggested that the senescence of endothelial cells is implicated in the pathogenesis of lung diseases. However, the role of senescent endothelial cells in silicosis remains poorly understood. By establishing multiple endothelial cell senescence models, and a silica-induced pulmonary fibrosis mouse model, we found that silica-induced endothelial cell senescence was accompanied by the increased expression of Galectin 3 (Gal3, gene name LGALS3). Mechanistically, silica-induced senescent cells synthesized a substantial amount of Gal3, which was subsequently released into the cellular microenvironment. Then, Gal3 directly binds to TGFBR1 on the cell membrane of lung fibroblasts and TLR4 on the macrophages, respectively. This cell communication facilitates the progression of silicosis by promoting fibroblast-myofibroblast transition (FMT) and NLRP3 inflammasome activation. Furthermore, Gal3 is regulated by the transcriptional regulatory factor CEBPB (CCAAT/ enhancer-binding protein beta) in senescent endothelial cells. In vivo, the administration of Lgals3 siRNA-loaded liposomes significantly ameliorated silica-induced pulmonary fibrosis. Collectively, our study demonstrated the critical role of endothelial cell senescence through the secretion of Gal3, which contributes to pulmonary fibrosis by promoting endothelial-fibroblast and endothelial-macrophage crosstalk.
Collapse
Affiliation(s)
- Demin Cheng
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wenxiu Lian
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinying Jia
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ting Wang
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210000, China
| | - Wenqing Sun
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhenhua Jia
- Hebei Yiling Hospital, High-level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang, Hebei 050091, China; National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, Hebei 050035, China.
| | - Yi Liu
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Chunhui Ni
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 320700, China.
| |
Collapse
|
10
|
Pacheco MI, Carvalho-Pereira J, Costa-Barbosa A, Marques F, Durães PM, Gonçalves MST, Sousa MJ, Sampaio P. Benzo[a]phenoxazine derivative C34 efficacy against fluconazole-resistant Candida spp. Microb Pathog 2025; 203:107498. [PMID: 40122407 DOI: 10.1016/j.micpath.2025.107498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Candida infections pose a significant health risk, prompting the search for new antifungal solutions due to the diminishing effectiveness of traditional drugs. Benzo[a]phenoxazine derivatives, described to have antimicrobial activity, are promising candidates. This study assessed the antifungal efficacy of five benzo[a]phenoxazine derivatives against Candida species for an effective antifungal strategy. The antifungal activity of various compounds against C. albicans, C. auris, C. glabrata C. parapsilosis, C. krusei and C. bracarensis was assessed using the yeast EUCAST protocol. C34, the most effective compound, was encapsulated in DODAB:MO liposomes. Antifungal efficacy, adhesion, and filamentation effects were compared for free and encapsulated C34. Cytotoxicity was determined via the MTT assay in the J774A.1 macrophage-like cell line, which was also employed to assess macrophage yeast killing in the presence of C34. The MIC values ranged from 3.75 to 60 μM, with C34 emerging as the most effective compound against all tested species/strains, specifically against fluconazole-resistant strains. Encapsulation in DODAB:MO liposomes improved C34's antifungal activity for most species, reducing MIC values. Both free and encapsulated C34 effectively reduced Candida adhesion and filamentation. Cytotoxicity assessment allowed the identification of a non-cytotoxic concentration of C34 that significantly enhanced macrophage activity against C. albicans. C34 displayed potent antifungal activity against various strains, including fluconazole-resistant ones as C. auris. It reduced key virulence factors, such as adhesion and filamentation, and enhanced macrophage-mediated clearance, making it a compound of interest for further development as a potential therapeutic option.
Collapse
Affiliation(s)
- Maria Inês Pacheco
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; Centre of Chemistry (CQUM), Department of Chemistry, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal.
| | - Joana Carvalho-Pereira
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; Institute of Science and Innovation for Bio Sustainability (IB S), University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| | - Augusto Costa-Barbosa
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; Institute of Science and Innovation for Bio Sustainability (IB S), University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| | - Fabiana Marques
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; Institute of Science and Innovation for Bio Sustainability (IB S), University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| | - Pedro M Durães
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; Institute of Science and Innovation for Bio Sustainability (IB S), University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| | - M Sameiro T Gonçalves
- Centre of Chemistry (CQUM), Department of Chemistry, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| | - Maria João Sousa
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; Institute of Science and Innovation for Bio Sustainability (IB S), University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| | - Paula Sampaio
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal; Institute of Science and Innovation for Bio Sustainability (IB S), University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| |
Collapse
|
11
|
Arya DK, Pandey P, Kumar A, Chidambaram K, Fatease AA, Pandey G, Srivastava S, Rajinikanth PS. Dual-ligand functionalized liposomes with iRGD/trastuzumab co-loaded with gefitinib and lycorine for enhanced metastatic breast cancer therapy. J Liposome Res 2025; 35:173-187. [PMID: 39895032 DOI: 10.1080/08982104.2025.2457453] [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: 11/21/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
Personalized treatment strategies have greatly improved the efficacy of anticancer drugs. Nanocarriers, especially liposomes, function as excellent platform for the delivery of both hydrophilic and hydrophobic agents. iRGD is a peptide composed of 9-amino acid denoted as (iRGDP), enhances selective and intratumoral delivery of anticancer drugs. Trastuzumab (TMAB), mainly targets HER2-positive advanced stage breast cancer is an FDA-approved monoclonal antibody. Gefitinib (GEB) is an anticancer drug, effective against metastatic breast cancer (MBC), while Lycorine hydrochloride (LCOH), a naturally derived compound, possess both anti-inflammatory and anticancer properties. This research is mainly emphasizing on the preparation of GEB and LCOH-entrapped TPGS-COOH coated-liposomes, camouflaged with an antibody (TMAB) and cyclic peptide (iRGDP) for targeted delivery in MBC therapy. The developed multifunctional liposomes were studied for extensive in vitro cell line studies on MCF-7 cells. The half-maximum inhibitory concentration (IC-50) values of GEB and LCOH co-loaded single functionalized liposome (SFL) (iRGDP-LiP, and TMAB-LiP) and dual-functionalized liposome (DFL) (iRGDP-TMAB-LiP) on MCF-7 cells were 1.04 ± 0.023 μg/mL, 0.71 ± 0.018 μg/mL, and 0.56 ± 0.028 μg/mL, respectively. Inverted confocal laser scanning microscopy (ICLSM) revealed enhanced cellular internalization in SFL and DFL-treated groups tagged with coumarin-6 and rhodamine-B dye as compared to conventional liposome. The scratch assay revealed a marked reduction in cell migration, while DAPI staining confirmed enhanced nuclear condensation (NC) and nuclear fragmentation (NF) in SFL and DFL-treated groups. Moreover, flow cytometry demonstrated enhanced early and late apoptosis in SFL and DFL groups. These findings indicate that GEB and LCOH co-loaded multifunctional liposome holds promise as a multifaceted therapeutic approach for MBC therapy.
Collapse
Affiliation(s)
- Dilip Kumar Arya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Prashant Pandey
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Anit Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Giriraj Pandey
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
- School of Pharmacy, Taylor's University, Lakeside Campus, Subang Jaya, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Jain A, Mishra AK, Hurkat P, Shilpi S, Mody N, Jain SK. Navigating liver cancer: Precision targeting for enhanced treatment outcomes. Drug Deliv Transl Res 2025; 15:1935-1961. [PMID: 39847205 DOI: 10.1007/s13346-024-01780-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] [Accepted: 12/18/2024] [Indexed: 01/24/2025]
Abstract
Cancer treatments such as surgery and chemotherapy have several limitations, including ineffectiveness against large or persistent tumors, high relapse rates, drug toxicity, and non-specificity of therapy. Researchers are exploring advanced strategies for treating this life-threatening disease to address these challenges. One promising approach is targeted drug delivery using prodrugs or surface modification with receptor-specific moieties for active or passive targeting. While various drug delivery systems have shown potential for reaching hepatic cells, nano-carriers offer significant size, distribution, and targetability advantages. Engineered nanocarriers can be customized to achieve effective and safe targeting of tumors by manipulating physical characteristics such as particle size or attaching receptor-specific ligands. This method is particularly advantageous in treating liver cancer by targeting specific hepatocyte receptors and enzymatic pathways for both passive and active therapeutic strategies. It highlights the epidemiology of liver cancer and provides an in-depth analysis of the various targeting approaches, including prodrugs, liposomes, magneto-liposomes, micelles, glycol-dendrimers, magnetic nanoparticles, chylomicron-based emulsion, and quantum dots surface modification with receptor-specific moieties. The insights from this review can be immensely significant for preclinical and clinical researchers working towards developing effective treatments for liver cancer. By utilizing these novel strategies, we can overcome the limitations of conventional therapies and offer better outcomes for liver cancer patients.
Collapse
Affiliation(s)
- Ankit Jain
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Ashwini Kumar Mishra
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
- Central Ayurveda Research Institute, Jhansi, Uttar Pradesh, 284003, India
| | - Pooja Hurkat
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | - Satish Shilpi
- School of Pharmaceuticals and Population Health Informatics, FOP, DIT University, Dehradun, Uttarakahnad, India
| | - Nishi Mody
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | | |
Collapse
|
13
|
Fayazi M, Rostami M, Amiri Moghaddam M, Nasiri K, Tadayonfard A, Roudsari MB, Ahmad HM, Parhizgar Z, Majbouri Yazdi A. A state-of-the-art review of the recent advances in drug delivery systems for different therapeutic agents in periodontitis. J Drug Target 2025; 33:612-647. [PMID: 39698877 DOI: 10.1080/1061186x.2024.2445051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
Periodontitis (PD) is a chronic gum illness that may be hard to cure for a number of reasons, including the fact that no one knows what causes it, the side effects of anti-microbial treatment, and how various kinds of bacteria interact with one another. As a result, novel therapeutic approaches for PD treatment must be developed. Additionally, supplementary antibacterial regimens, including local and systemic medication administration of chemical agents, are necessary for deep pockets to assist with mechanical debridement of tooth surfaces. As our knowledge of periodontal disease and drug delivery systems (DDSs) grows, new targeted delivery systems like extracellular vesicles, lipid-based nanoparticles (NPs), metallic NPs, and polymer NPs have been developed. These systems aim to improve the targeting and precision of PD treatments while reducing the systemic side effects of antibiotics. Nanozymes, photodermal therapy, antibacterial metallic NPs, and traditional PD therapies have all been reviewed in this research. Medicinal herbs, antibiotics, photothermal therapy, nanozymes, antibacterial metallic NPs, and conventional therapies for PD have all been examined in this research. After that, we reviewed the key features of many innovative DDSs and how they worked for PD therapy. Finally, we have discussed the advantages and disadvantages of these DDSs.
Collapse
Affiliation(s)
- Mehrnaz Fayazi
- School of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Mitra Rostami
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kamyar Nasiri
- Department of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Azadeh Tadayonfard
- Department of Prosthodontics, Dental Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Behnam Roudsari
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Zahra Parhizgar
- Resident of Periodontology, Department of Periodontics, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
14
|
Pandya T, Joshi D, Presswala Z, Kulkarni M, Patel R, Patel S, Bhattacharya S, Prajapati BG. Advanced therapeutic strategies using Thermo-sensitive chitosan/pectin hydrogel in the treatment of multiple cancers. Carbohydr Polym 2025; 357:123454. [PMID: 40158985 DOI: 10.1016/j.carbpol.2025.123454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
Current cancer therapies including immunotherapy and chemotherapy produce adverse side effects that demand improved drug distribution methods. Research shows that thermosensitive chitosan/pectin-based hydrogels serve as an effective platform technology for drug delivery during cancer therapy because of their ability to control drug release at specific locations. The hydrogels perform temperature-triggered sol-gel phase shifts which enables prolonged drug delivery together with minimal toxic side effects. The biocompatible and biodegradable properties of these materials enable solutions against drug resistance and tumour heterogeneity challenges. Studies have demonstrated that these hydrogels enhance drug bioavailability, extend circulation time, and improve tumour targeting, leading to increased therapeutic efficacy and reduced systemic toxicity. Their ability to sustain drug release and penetrate tumour microenvironments makes them a promising strategy for overcoming drug resistance and tumour heterogeneity. Their ability to reproduce native tissue properties poses challenges that scientists must address through improved structural optimization approaches. The combination of latest nanotechnology innovations and interdisciplinary studies has sped up the creation of chitosan/pectin hydrogels for cancer treatment applications. This review highlights the significant advancements and demonstrated effectiveness of thermosensitive chitosan/pectin hydrogels in cancer treatment by exploring their design parameters alongside their drug release behaviour while discussing their potential medical applications.
Collapse
Affiliation(s)
- Tosha Pandya
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, L J University, Ahmedabad, Gujarat 382210, India
| | - Disha Joshi
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, L J University, Ahmedabad, Gujarat 382210, India
| | - Zenab Presswala
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, L J University, Ahmedabad, Gujarat 382210, India
| | - Mangesh Kulkarni
- Department of Pharmaceutics, Gandhinagar Institute of Pharmacy, Gandhinagar University, Moti Bhoyan, Khatraj-Kalol Road, Gujarat 382721, India
| | - Riya Patel
- School of Pharmacy, Indrashil University, Rajpur, Kadi, Gujarat 382715, India
| | - Shivani Patel
- Parul College of Pharmacy and Research, Parul University, P.O.Limda, Ta.Waghodia, Vadodara, Gujarat 391760, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India.
| | - Bhupendra G Prajapati
- Shree S.K. Patel College of Pharmaceutical Education & Research, Ganpat University, Gujarat 384012, India; Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
15
|
Uçar Baş K, Ağaçdiken A, Örs Demet ED, Tuğal Aslan D, Reçber T, Öztürk SC, Gulsun T, Çelebier M, Göktaş Z. Comparison of free vs. liposomal naringenin in white adipose tissue browning in C57BL/6j mice. J Liposome Res 2025; 35:94-104. [PMID: 39298118 DOI: 10.1080/08982104.2024.2405131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 05/29/2025]
Abstract
Naringenin may play a role in browning by increasing thermogenic gene expression. In this study, we encapsulated naringenin using a liposomal formulation and examined the effects of both free and liposomal naringenin on white adipose tissue browning in C57BL6/J mice. In the first phase of the study, naringenin was encapsulated by the liposome method, which is biocompatible and biodegradable. The physical and chemical properties of liposomal naringenin were tested. In the second phase, a total of 48 six-week-old mice were divided into two main groups: prevention and recovery. Each main group was divided into four subgroups: nano-naringenin, void, free-naringenin, and control. The prevention group received a high-fat diet for 10 weeks along with weekly intravenous injections of 20 µM naringenin. On the other hand, the recovery group was first subjected to a high-fat diet for 10 weeks, followed by an additional 10 weeks of the same diet, along with weekly intravenous injections of 20 µM naringenin. Body weight was measured once per week, and brown adipose tissue, inguinal white adipose tissue, and serum samples were collected from each mouse. The mean particle size, polydispersity index and zeta potential values of liposomal naringenin were ∼207 nm, 0.35, and -27 mV, respectively. The encapsulation and loading efficiencies of liposomal naringenin were 94.6 and 19.2%, respectively. Liposomal naringenin exhibited sustained-release behavior, while free naringenin showed a burst-release profile. Liposomal naringenin showed the best physical stability in light and at 4 °C, while free naringenin was more chemically stable in light and at 4 and 22 °C. Free and liposomal naringenin did not significantly reduce weight gain. In the prevention group, liposomal naringenin increased PRDM16 gene expression in inguinal white adipose tissue 4.29 times more than free naringenin (p = 0.010). However, neither formulation significantly altered the expression levels of other browning or adipogenesis markers in the tissues. The results suggest that free naringenin can be efficiently encapsulated in biocompatible and biodegradable nanoparticles. Further research is needed to better understand the physiological effects of liposomal naringenin.
Collapse
Affiliation(s)
- Kübra Uçar Baş
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Aslıhan Ağaçdiken
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Elif Didem Örs Demet
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Necmettin Erbakan University, Konya, Turkey
| | - Dilem Tuğal Aslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Süleyman Can Öztürk
- Research and Application Center for Animal Experiments, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Tugba Gulsun
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Mustafa Çelebier
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Zeynep Göktaş
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara, Turkey
| |
Collapse
|
16
|
Chen N, Yang Y, Fan L, Cai Y, Yin W, Yang Z, Zhao Y, Chen S, Zhi H, Xue L, Zhang X, An L, Li Y, Ren T. The STING-activating nanofactory relieves T cell exhaustion in Mn-based tumor immunotherapy by regulating mitochondrial dysfunction. J Nanobiotechnology 2025; 23:403. [PMID: 40450344 DOI: 10.1186/s12951-025-03469-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 05/16/2025] [Indexed: 06/03/2025] Open
Abstract
Manganese-based STING-activating tumor immunotherapy faces limitations due to T cell exhaustion. Mitochondrial dysfunction is a key factor contributing to T cell exhaustion. Modulating mitochondrial function during manganese-based immunotherapy offers a promising strategy to reverse T cell exhaustion. Spermidine (SPD) enhances mitochondrial function in T cells, making the co-delivery of Mn and SPD a potential therapeutic approach. However, intravenous co-delivery is hindered by the rapid formation of MnO(OH)₂ precipitates. In this study, liposomes were employed as nano-reactors to facilitate the reaction between pre-loaded Mn²⁺ and O₂ in the presence of SPD, forming MnO(OH)₂ precipitates within the liposomes. These liposomes function as nanofactories, further processing MnO(OH)₂ under the regulation of the tumor microenvironment (TME) and delivering Mn, SPD, and O₂. Beyond activating the STING pathway in dendritic cells, L@Mn@SPD alleviates TME hypoxia and effectively reverses CD8⁺ T cell exhaustion. In vivo, L@Mn@SPD achieved a 2.44-fold increase in tumor suppression compared to MnCl₂, along with a 47% rise in CD8⁺ T cell infiltration, a 62.1% reduction in PD-1 expression, and a 110% increase in IFN-γ secretion. This STING-activating nanofactory provides a promising strategy to enhance manganese-based tumor immunotherapy by addressing mitochondrial dysfunction in exhausted T cells.
Collapse
Affiliation(s)
- Nana Chen
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yushan Yang
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Limin Fan
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yanni Cai
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Weimin Yin
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zichen Yang
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yuge Zhao
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shiyu Chen
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Hui Zhi
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Liangyi Xue
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoyou Zhang
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Lulu An
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yongyong Li
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Tianbin Ren
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
17
|
Luo Q, Xing X, Song Y, Gu B, Hu Q, Liu W, Xiao Y, Wang Z. MiR-29a-3p ameliorate behavioral deficiency in hypoxia-ischemia brain damage in neonatal mice by inhibiting BTG2. Behav Brain Res 2025; 486:115552. [PMID: 40147793 DOI: 10.1016/j.bbr.2025.115552] [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: 01/13/2025] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
It has been reported that miR-29a-3p played a part in series neurological disorders. However, it remains unclear whether miR-29a-3p participate in the pathological mechanism in hypoxia-ischemia (HI) brain injury. In this study, we detected the change of miR-29a-3p level in the ipsilateral cortex following HI brain injury and found that miR-29a-3p was significantly increased at 3 days in the ipsilateral cortex following HI insult in neonatal mice. Therefore, we further explored the role of miR-29a-3p in HI brain injury and its molecular mechanism. The results showed that miR-29a-3p mimics attenuated and miR-29a-3p antagomir aggravated brain infarction volume at 3 days following HI insult. We further found that overexpression of miR-29a-3p also suppressed apoptosis and neuroinflammation, reduced synaptic loss and prevent HI-induced microglial morphological changes 3 days following HI insult. Neurobehavioral tests revealed that overexpression of miR-29a-3p could improve both short-term and long-term behavioral defects after HI injury. Furthermore, we proved that miR-29a-3p targets B-cell translocation gene 2 (BTG2) and further inhibits the expression of Bax by luciferase reporter assay and qRT-PCR. Moreover, overexpression of miR-29a-3p, by applying liposomes through intranasal route, could also achieve the same therapeutic effect in HI injury. Our data showed that by inhibiting BTG2/Bax, increasing level of miR-29a-3p might serve as a strategy to prevent brain damage and behavioral deficiency in HI.
Collapse
Affiliation(s)
- Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaohui Xing
- Department of Neurosurgery, Liaocheng Neuroscience Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China
| | - Yan Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Bing Gu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China; Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Quan Hu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong 271000, PR China
| | - Weiyang Liu
- Jinan Xicheng Experimental High School, Dezhou Road, Jinan, Shandong 1999, PR China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng Neuroscience Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China.
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
18
|
Wang X, Wang H, Dai Y, Lu R, Chen J, Kong Q. Application potential of injectable hydrogels in the post-surgical window period following tumor surgery. Int J Pharm 2025; 679:125754. [PMID: 40425056 DOI: 10.1016/j.ijpharm.2025.125754] [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: 04/12/2025] [Revised: 05/17/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
Chemotherapy is one of the primary modalities for the treatment of malignant diseases. The outcomes, however, are different between tumors of various origins, which hinder clinical applications. The advantages of chemotherapies in patients with hematological lesions are more obvious than those seen in solid tumors. This might be attributed to the availability of drug concentration and exposure time. Based on this phenomenon, we hypothesis that localized drug administration is expected to be more potential for solid tumors, particularly for the residual tumors in post-operative "window period". The presence of residual tumors after surgical resection are the major factors leading to tumor recurrence after surgery. The methods of dealing with this problem are yet to be found. Conventional chemotherapies are scarcely applied in the post-surgical window period due to their unselected and unexpected side effects. This article studied the advantages and disadvantages of prominent formulations currently utilized in the field of local implantation in cancer treatment, with the notable superiority of injectable hydrogel platforms being most appealing. These platforms not only enhance wound healing of the patients with less side effects, during the "window period" following tumor surgery, but also effectively eradicate residual tumors by facilitating the establishment of a favorable microenvironment. Additionally, the challenges seen in this field and future directions are discussed, which is expected to provide insights for pharmaceutical professionals and clinical applications.
Collapse
Affiliation(s)
- Xilei Wang
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China; Taizhong Pharmaceutical Co., Ltd, No. 799 Yaocheng Avenue, Medical High-Tech Zone (Fudan University Taizhou Institute of Health Sciences), Taizhou 225326, China.
| | - Huan Wang
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Yue Dai
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Rong Lu
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Jingdi Chen
- Marine College, Shandong University, No. 180 Wenhua West Road, Weihai 264209, China.
| | - Qingzhong Kong
- Taizhong Pharmaceutical Co., Ltd, No. 799 Yaocheng Avenue, Medical High-Tech Zone (Fudan University Taizhou Institute of Health Sciences), Taizhou 225326, China.
| |
Collapse
|
19
|
Cimmino W, Angelillo A, Rea G, Kalligosfyri PM, Nele V, Campani V, De Rosa G, Cinti S. Development of an Electrochemical Biosensor to Detect miRNA Encapsulated in Lipid Nanoparticles. Anal Chem 2025. [PMID: 40411458 DOI: 10.1021/acs.analchem.5c00692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2025]
Abstract
Lipid nanoparticles (LNPs) represent a versatile delivery platform proposed for a wide range of therapies based on nucleic acids, including microRNA (miRNAs). The ability of LNPs to encapsulate and protect RNA from degradation, as well as their ability to promote cellular uptake, has led to their clinical use with the approval of RNA-based medicinal products, i.e., COVID vaccines. In this context, a growing number of LNP formulations with improved transfection and biocompatibility are under development, requiring rapid, sensitive, and robust quality control tests, e.g., for the quantification of the encapsulated RNA. Nowadays, classical analytical approaches such as fluorescence, ultraviolet-visible (UV-vis) spectrophotometry, and chromatography are mainly used for the quantification of the encapsulated drug. However, the user-friendly and cost-effective quantification of the encapsulation efficacy within LNPs represents an important research focus, as it would allow monitoring of the amount of encapsulated RNA, thus providing immediate quality control. In this work, we present the adaptation of an electrochemical strip to quantify the encapsulation of a miRNA, i.e., miR-218, whose antitumor effect has been widely reported in the literature within LNPs. We provide a rapid and sensitive method to assess the concentrations of miRNA actually encapsulated, obtaining satisfactory agreement compared to the traditional fluorimetric approach. Specifically, the platform is based on a commercial gold-screen-printed electrode modified with a DNA probe designed to be fully complementary to the target miRNA-218. The electrochemical system was successfully combined with a 3D-printed chamber that allowed the use of multiple electrodes simultaneously and the use of Triton X-100 surfactant to disrupt the LNPs and release the encapsulated miRNA-218 achieving a detection limit as low as 1 nM.
Collapse
Affiliation(s)
- Wanda Cimmino
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Alessia Angelillo
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppina Rea
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | | | - Valeria Nele
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Virginia Campani
- Department of Life Science, Health and Health Professions, Link Campus University, Rome 00165, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Stefano Cinti
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
- Bioelectronics Task Force at University of Naples Federico II, Via Cinthia 21, Naples 80126, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
20
|
Pang ZY, Zhu YB, Hu JX, Li XX, Gao YJ, Wang YY, Zhou Q, Li P. The impact of lipidome on intervertebral disk degeneration, low back pain, and sciatica: a Mendelian randomization study. Sci Rep 2025; 15:18045. [PMID: 40410311 PMCID: PMC12102376 DOI: 10.1038/s41598-025-99914-9] [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: 12/27/2024] [Accepted: 04/23/2025] [Indexed: 05/25/2025] Open
Abstract
Degeneration of intervertebral discs is a significant factor in chronic lower back pain, impacting millions annually. Existing studies propose a potential link between lipids and disc disease, though causal relationships remain unclear. The objective of this study is to explore the causal connections between lipids, lower back pain, disc degeneration, and the risk of sciatica In this research, we utilized a comprehensive GWAS dataset encompassing 179 lipid traits to explore the causal connections between lipids and the susceptibility to conditions such as lower back pain (LBP), intervertebral disc degeneration (IVDD), and sciatica. To establish causality, we employed two-sample Mendelian randomization, supplemented by Bayesian model averaging for verification. Our assessment of diversity and mutual influence involved Cochran's Q test, MR-Egger intercept assessment, and MR-PRESSO. Additionally, we performed a sensitivity analysis by systematically excluding individual elements to gauge their impact on outcomes in Mendelian randomization. Lastly, bidirectional Mendelian randomization was conducted to explore potential inverse associations between lipids and IVDD. Analyzing 179 lipidomic features as exposures and IVDD, LBP, and sciatica as outcomes, this study reveals significant causal relationships of glycerophospholipids, sterols, and glycerolipids with the risk of IVDD, LBP, and sciatica. Phosphatidylcholine, triglycerides, and sterols consistently exerted risk influences on IVDD, while phosphatidylethanolamine (O-16:1_18:2) among glycerophospholipids exhibited a protective effect (OR: 0.927-0.998, P < 0.05). Regarding LBP, sphingomyelin (d38:2) in sphingolipids demonstrated a protective effect (OR: 0.925-0.997, P < 0.05). For sciatica, triglycerides exhibited a risk influence, with varying effects observed for phosphatidylcholine and sterols with different molecular structures. Notably, sterol ester (27:1/16:1) consistently showed a risk effect across all three conditions. Our research provides valuable insights into how certain lipids are linked to the risks of LBP, IVDD, and sciatica. Our findings indicate that phosphatidylcholine and triglycerides may increase the incidence of IVDD, LBP, and sciatica, suggesting potential adverse effects. In contrast, sphingomyelin appears to reduce the occurrence of LBP and sciatica, indicating a protective role. Sterol esters also show a protective effect against sciatica; however, the sterol ester (27:1/16:1) consistently demonstrates a detrimental impact on IVDD, LBP, and sciatica. Additionally, our study underscores the intricate nature of lipid metabolism concerning IVDD, LBP, and sciatica. It uncovers a range of structural variations among lipids and explores how these variations may lead to different effects across various molecular subtypes.
Collapse
Affiliation(s)
- Ze-Yu Pang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yi-Bo Zhu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Jun-Xian Hu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Xiao-Xiao Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yong-Jian Gao
- Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yi-Yang Wang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China.
| | - Qiang Zhou
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China.
| | - Pei Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China.
| |
Collapse
|
21
|
Trusova V, Malovytsia U, Vus K, Zhytniakivska O, Gorbenko G. Amyloid-liposome composites as hybrid platforms for doxorubicin delivery. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184426. [PMID: 40414331 DOI: 10.1016/j.bbamem.2025.184426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/26/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
The feasibility of engineering the sophisticated hybrid drug delivery platforms through the integration of phospholipid vesicles within a matrix of amyloid suspensions has been evaluated. Utilizing the equilibrium dialysis methodology and spectrofluorometric technique, the quantitative analysis of doxorubicin (DOX) encapsulation capacity of diverse phospholipid assemblies, amyloid suspensions, and their corresponding composite systems has been performed. Our findings revealed that the incorporation of negatively charged cardiolipin (CL) into phosphatidylcholine (PC) lipid vesicles significantly enhances DOX encapsulation and retention, while the addition of amyloid fibrils to charged liposomes has minimal impact on the drug binding. The neutral PC liposomes modified with insulin and lysozyme fibrillar suspensions exhibited improved doxorubicin encapsulation and retention compared to unmodified liposomes, thereby displaying a potential for reduced toxicity and prolonged drug action in vivo. Notably, amyloid fibrils alone were found to demonstrate the lower degree of DOX encapsulation and retention as compared to liposomes. Fluorimetric analysis suggests that the presence of insulin and lysozyme fibrils alters the microenvironment of DOX towards a more hydrophobic which is consistent with deeper bilayer penetration. Cumulative data from release kinetics and retention studies along with fluorescence measurements suggest that PC liposome-insulin fibril composites represent the most promising DOX nanocarriers, combining enhanced drug encapsulation, structural stability, and optimal drug location within the bilayer. The results obtained provide valuable insights into the design of protein-lipid nanomaterials for enhanced drug delivery, offering promising avenues for the development of more effective and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Valeriya Trusova
- Department of Medical Physics and Biomedical Nanotechnologies, V.N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv 61022, Ukraine.
| | - Uliana Malovytsia
- Department of Medical Physics and Biomedical Nanotechnologies, V.N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv 61022, Ukraine
| | - Kateryna Vus
- Department of Medical Physics and Biomedical Nanotechnologies, V.N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv 61022, Ukraine
| | - Olga Zhytniakivska
- Department of Medical Physics and Biomedical Nanotechnologies, V.N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv 61022, Ukraine
| | - Galyna Gorbenko
- Department of Medical Physics and Biomedical Nanotechnologies, V.N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv 61022, Ukraine
| |
Collapse
|
22
|
Hildebrand LS, Jost T, Schindler M, Derer A, Fuhrmann G, Fietkau R, Distel LV. Inhibiting NHEJ in HNSCC cell lines by the ligase IV inhibitor SCR130 has limited radiosensitizing effects. Sci Rep 2025; 15:17871. [PMID: 40404928 PMCID: PMC12098888 DOI: 10.1038/s41598-025-03159-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
Radiotherapy (RT) is a relevant treatment for head and neck squamous cell carcinoma (HNSCC) patients but radioresistance, which depends on DNA damage response (DDR), restrains outcome. Therefore, manipulating DDR by small molecule inhibitors (SMI) is a promising treatment option. The main DNA double strand break (DSB) repair mechanisms in healthy mammalian cells are homologous recombination (HR) and non-homologous end joining (NHEJ). It is known that HR is already often impaired in tumors because of cancerous transitions. Therefore, additionally inhibiting NHEJ is a possibility to specifically target tumor cells and spare healthy tissue, which has the alternative DSB repair mechanism available. We treated HNSCC and healthy fibroblast cell lines with 30 µM of the ligase IV inhibitor SCR130 and a single dose of 2 Gy (Gy) ionizing radiation (IR) to investigate the inhibitor's radiosensitizing effect. In short, the effect of SCR130 in combination with IR on cell death, clonogenicity, and DNA damage is limited and highly cell line specific. Nevertheless, SCR130 increases the number of cells in G0/G1 phase concomitant with gained p21 expression consistently. We suggest that SCR130 in combination with IR has anti-proliferative effects, but an escape of the cells by upregulation of ligase IV resulting from the treatment is possible.
Collapse
Affiliation(s)
- Laura S Hildebrand
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Tina Jost
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Radiation Oncology, Translational Radiobiology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
| | - Marion Schindler
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstraße 5, 91058, Erlangen, Germany
| | - Anja Derer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Radiation Oncology, Translational Radiobiology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
| | - Gregor Fuhrmann
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstraße 5, 91058, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Luitpold V Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.
| |
Collapse
|
23
|
Bacal CJO, Allardyce BJ, Valente F. Influence of material format and surface chemistry for the sustained delivery and efficacy of silk drug delivery systems in vivo. J Mater Chem B 2025. [PMID: 40400450 DOI: 10.1039/d4tb02756f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Silk fibroin materials are promising for use in controlled drug delivery in the field of tissue engineering and biomedical applications thanks to silk's generally established biocompatibility and tunable properties for implants and drug storage. Several factors must be considered in the materials design, including material format, drug properties and release kinetics, and the activity and stability of the drug after release. While numerous reviews described silk-based DDS that demonstrated controllable in vitro release, success in vivo has been limited, especially in some material formats. This review therefore aims to provide insight into the current material format and functionalization strategies to maximize in vivo performance by describing the in vivo activity of recently developed silk drug delivery systems. The review also aims to provide a fresh perspective on the suitable format and functionalization strategies for a target biomedical application. Based on the release behavior of drugs in various material formats, silk films, foams, and microneedles were better suited to serve as scaffolds for cell regeneration and improved recovery rate for biomedical applications involving wound healing and tissue engineering. Gels and particles could be incorporated within the films and foams but the purpose would be to serve as additional physical barriers towards drug diffusion in these types of application. For drugs or therapeutics that target internal organs (i.e. brain, liver, intestines, etc.), gels and particles were mainly used due to their size. In the event that the material format selection based on the target application does not contribute a lot to the prolonged release of drugs or therapeutic agents, hybrid functionalization strategies were adapted to make the surface chemistry of the material more responsive to the environmental stimuli for a more tunable silk DDS.
Collapse
Affiliation(s)
- Christine Jurene O Bacal
- Ear Science Institute Australia, Ear Sciences Centre, School of Medicine, The University of Western Australia, Nedlands 6009, Australia.
| | | | - Filippo Valente
- Ear Science Institute Australia, Ear Sciences Centre, School of Medicine, The University of Western Australia, Nedlands 6009, Australia.
| |
Collapse
|
24
|
Kapoor A, Hafeez A, Kushwaha P, Ara N. Chitosan-coated nanostructured lipid carriers of amantadine for nose-to-brain delivery: formulation optimization, in vitro-ex vivo characterization, and in vivo anti-parkinsonism assessment. Int J Biol Macromol 2025:144497. [PMID: 40409652 DOI: 10.1016/j.ijbiomac.2025.144497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 05/15/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Amantadine hydrochloride (AMH) is a repurposed moiety of anti-viral category having significant response against Parkinsonism. Nasal route is linked with the direct delivery of drugs to brain through olfactory and trigeminal nerves. The mucociliary clearance is the major obstacle for effective delivery of drugs. The optimized formulation was coated with chitosan (CH) to enhance the residence time of formulation in the nasal cavity. The optimized NLCs exhibited particle size of 174.2 ± 1.5 nm, PDI of 0.131 ± 0.019, zeta potential of -27.52 ± 3.29 mV, EE of 74.06 ± 2.3 %, and nasal flux of 66.03 ± 0.9 μg/cm2/h. FTIR analysis revealed AMH-excipient compatibility and encapsulation of AMH into NLCs. XRD analysis confirmed no polymorphic transition in the developed NLCs. Drug release exhibited sustained release of AMH (71.01 ± 2.19 %) up to 24 h. Ex vivo study (goat nasal mucosa) showed 3.22 folds enhanced permeability of AMH from AMH-NLCs in comparison to AMH-Sol. The recommended storage condition for AMH-NLCs was found to be 25 °C/60 % RH. AMH-NLCs showed high cell viability (88.23 ± 7.49 %) in comparison to AMH-Sol (62.14 ± 6.81 %) on H2O2 induced PC12 cells at a concentration level of 100 μg/ml. In vivo studies exhibited effectiveness of CH-AMH-NLCs and AMH-NLCs and showed increased response when compared with amantadine solution (pure form) and marketed formulation.
Collapse
Affiliation(s)
- Archita Kapoor
- Faculty of Pharmacy, Integral University, Lucknow- 226026, India.
| | - Abdul Hafeez
- Faculty of Pharmacy, Integral University, Lucknow- 226026, India.
| | - Poonam Kushwaha
- Faculty of Pharmacy, Integral University, Lucknow- 226026, India.
| | - Nargis Ara
- Faculty of Pharmacy, Integral University, Lucknow- 226026, India.
| |
Collapse
|
25
|
Huang Y, Li Q, He F, Yang T, Zhou Q, Zheng Y, Li Y. Cationic Azobenzene Tag to Enhance Liposomal Prodrug Retention and Tumor-Targeting Prodrug Activation for Improved Antitumor Efficacy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26323-26337. [PMID: 40285708 DOI: 10.1021/acsami.5c01634] [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: 04/29/2025]
Abstract
In this study, we reported a cationic azobenzene (Azo) tag to increase the retention of camptothecin (CPT) prodrugs in liposomes driven by π-π stacking interaction between Azo. Compared with a cationic CPT prodrug without Azo, the liposome-encapsulating Azo-linked CPT prodrugs (AzoCPT-Lips) exhibited slower prodrug leakage in plasma and a longer blood circulation time in mice. The AzoCPT-Lips had a high encapsulation efficiency (95%), loading capacity (20%, by weight), and good storage stability. The AzoCPT was efficiently taken up by 4T1 tumor cells (100-fold higher than CPT) and readily converted into active CPT in the cytoplasm to exert 10-fold higher cytotoxicity than free CPT. More importantly, AzoCPT-Lips resulted in 5-20 times higher tumor distribution of active CPT than that of CPT solution or those in other tissues, which further led to more potent antitumor activity and lower toxicities in the 4T1 breast cancer xenograft. Such a cationic Azo tag represents an effective strategy for developing liposomal antitumor drugs with improved antitumor efficacy.
Collapse
Affiliation(s)
- Yuanyuan Huang
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qiunan Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Fei He
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tao Yang
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu 610051, China
| | - Yaxin Zheng
- Key Laboratory of Structure-Specific Small Molecule Drugs at Chengdu Medical College of Sichuan Province, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yang Li
- Department of Pharmaceutics, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
26
|
Aly Abdelfattah MT, Kassem AA, Mohamed Shalaby HK, Awad Tantawy MA. Investigation of the effect of resveratrol lyo-nanograft on the bone regeneration process of osseous bony defects in rabbits. Int J Pharm 2025; 678:125679. [PMID: 40324606 DOI: 10.1016/j.ijpharm.2025.125679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 05/01/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Periodontal diseases affect tooth-supporting structure producing bone-defects. Well-known bone-regeneration techniques are expensive, sophisticated and maybe immunogenic. We aimed at emerging a cost-effective, easily manufactured and safe nano-bone graft containing the natural-polyphenol resveratrol (RSV) which was reported to have in-vivo bone-regeneration efficacy. However, RSV suffers from poor solubility, bioavailability and photosensitivity. Its incorporation into a nanocarrier system could resolve these problems. RSV liposomes were prepared using Phosal® in different ratios and the optimized formula showed multilamellar vesicles, PS (126.1 nm), PDI (0.3017), ZP (-34.5 mV), %EE (90.86 %), with a sustained-release pattern. Then it was lyophilized and recharacterized and the obtained pliable mass (RSV-Ph75-LyoMan) lyo-nanograft suited its application into bone-defects performed in rabbits' tibiae. Histomorphometric quantitative analyses of bone sections micrographs were performed to verify RSV anti-inflammatory action, by counting bone marrow-infiltrating lymphocytes, and to explore the phases of bone-regeneration process exerted by RSV, by counting osteocytes, osteoblasts and osteoclasts and further confirmed by assessing bone-thickness, using ImageJ-software. This was done after 3, 6, 9 weeks of applying either placebo or RSV-Ph75-LyoMan. A significant decrease in lymphocytes' counts in RSV-Ph75-LyoMan compared to control-sites at the assessed time points was obtained. Further, RSV-Ph75-LyoMan significantly accelerated bone-healing and regeneration processes as concluded from the differential osteocytes, osteoblasts and osteoclasts counts. Consequently, a significant increase in bone-thickness in (RSV-Ph75-LyoMan)-treated-sites; 68.25 ± 3.45, 109.5 ± 5.49, and 148.2 ± 7.25 µm after 3, 6, 9 weeks was obtained, with a significant difference when compared to control-sites; 40.05 ± 2.21, 45.23 ± 1.87 and 46.57 ± 1.45 µm, respectively. Histological studies confirmed trabecular-remolding after 9 weeks in (RSV-Ph75-LyoMan)-treated-sites. RSV-Ph75-LyoMan lyo-nanograft is promising for treatment of osseous bony-defects in rabbits.
Collapse
Affiliation(s)
| | - Abeer Ahmed Kassem
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| | | | - Mohamed Ali Awad Tantawy
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
27
|
Nurjis F, Sarwar U, Sarfraz Ali J, Fayyaz M, Munir F, Shahzad S. Synergistic combinatorial delivery system based on nanoliposome encapsulating doxorubicin and sorafenib for broad-spectrum cancer treatment. J Microencapsul 2025; 42:300-312. [PMID: 40126990 DOI: 10.1080/02652048.2025.2480597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 03/12/2025] [Indexed: 03/26/2025]
Abstract
A novel combination delivery approach entrapping Sorafenib inside a nanoliposome bilayer and Doxorubicin within the aqueous core to achieve the broad-spectrum synergistic chemotherapeutic effect. DOX-SOR liposomes were synthesized by thin film hydration and characterized using UV-visible spectroscopy, Fourier Transform Infrared Spectroscopy, Dynamic Light Scattering, Fluorescence, and Scanning Electron Microscopy, followed by cytotoxicity assessments. Nanoliposomes demonstrated effective loading and encapsulation of Doxorubicin (10.23% ± 0.65 and 89.65% ± 0.52) and Sorafenib (10.42% ± 0.50 and 85.35% ± 0.72) with a 165 nm ± 1.34 mean diameter, -15.2 ± 1.78 zeta potential, and 75% ± 1.92 of cumulative release. In vitro analysis of nanoliposomes demonstrated biocompatibility up to 250 µg/mL concentration (p < 0.05), enhanced intracellular localization in Hep2c cell lines, 91% ± 1.72 cytotoxic effects (p < 0.0001) with IC50 up to 127µg/mL, 21% ± 0.89 cell viability with 85% apoptosis (p < 0.0001) using flow cytometer. This study presents a promising treatment approach using a multidrug-loaded nanoliposomes for broad-spectrum synergistic chemotherapy.
Collapse
Affiliation(s)
- Farwa Nurjis
- NILOP Nanomedicine Research Laboratories, National Institute of Lasers and Optronics College (NILOP-C), Pakistan Institute of Engineering and Applied Sciences, Islamabad, Pakistan
| | - Usama Sarwar
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Joham Sarfraz Ali
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| | - Mahnoor Fayyaz
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Faiza Munir
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Shaheen Shahzad
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| |
Collapse
|
28
|
Lopes-Nunes J, Lopes M, Rosado B, Maocha IG, Rolo J, Gaspar C, Pires B, Rosado T, Gallardo E, Palmeira-de-Oliveira A, Martinez-de-Oliveira J, Ferreira C, Cabral Campello MP, Paulo A, Medeiros-Fonseca B, Félix L, Venâncio C, Pinto MDL, Oliveira PA, Palmeira-de-Oliveira R, Cruz C. Vaginal formulation development: A strategy based on aptamer-guided liposome for human papillomavirus-induced lesions. Eur J Pharm Biopharm 2025; 210:114693. [PMID: 40154893 DOI: 10.1016/j.ejpb.2025.114693] [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: 03/07/2024] [Revised: 12/29/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
Human Papillomavirus (HPV) is the main cause of cervical cancer, and formulations have been widely used to treat vaginal lesions caused by HPV. Herein, liposomes with acridine orange derivative C8 were produced and functionalized with AT11 aptamer. Subsequently, they were incorporated into a formulation, prepared based on the universal placebo formulation, which included Thymus vulgaris (TEO) or Origanum vulgare (OEO) essential oils. The formulation was technologically characterized and permeation of C8 into vaginal tissue was determined. To assess its biological effect, cell viability and internalization tests were carried out using the MTT assay and confocal microscopy, respectively, and antimicrobial susceptibility was also assessed. The prepared formulations were able to internalize cells and reduce cell viability, especially in cancer cell lines. Additionally, formulations showed promising antibacterial and antifungal effects. The effect of the formulation containing TEO and the C8 AT11 liposomes was also tested in vivo in HPV16 transgenic and wild type mice. Briefly, the formulation proved to be safe for animals and presented some therapeutic potential, namely through the reduction of ear epithelial cells' proliferation. Overall, results suggest that essential oils can increase the anticancer potential of liposomes with associated C8 and AT11 promotes their selectivity towards cancer cells.
Collapse
Affiliation(s)
- Jéssica Lopes-Nunes
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Melanie Lopes
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Beatriz Rosado
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Izamara Gomes Maocha
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Joana Rolo
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Labfit-Health Products Research and Development Lda, UBImedical, Estrada Nacional 506, 6200-284 Covilhã, Portugal
| | - Carlos Gaspar
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Bruno Pires
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Tiago Rosado
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Eugénia Gallardo
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Palmeira-de-Oliveira
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Labfit-Health Products Research and Development Lda, UBImedical, Estrada Nacional 506, 6200-284 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - José Martinez-de-Oliveira
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Catarina Ferreira
- Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Maria Paula Cabral Campello
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal; Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal
| | - António Paulo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal; Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal
| | - Beatriz Medeiros-Fonseca
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Inov4Agro, Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, Vila Real, Portugal
| | - Luís Félix
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Inov4Agro, Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, Vila Real, Portugal
| | - Carlos Venâncio
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Inov4Agro, Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, Vila Real, Portugal
| | - Maria de Lurdes Pinto
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Inov4Agro, Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, Vila Real, Portugal
| | - Paula A Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Inov4Agro, Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, Vila Real, Portugal
| | - Rita Palmeira-de-Oliveira
- RISE-Health, Departamento de Ciências Médicas, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Labfit-Health Products Research and Development Lda, UBImedical, Estrada Nacional 506, 6200-284 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - Carla Cruz
- Departamento de Química, Universidade da Beira Interior, Rua Marquês de Ávila e Bolama, 6201-001 Covilhã, Portugal; RISE-Health, Departamento de Química, Faculdade de Ciências, Universidade da Beira Interior, Rua Marquês d'Ávila e Bolama 6201-001 Covilhã, Portugal.
| |
Collapse
|
29
|
Heo JI, Kim MJ, Kim D, Seo J, Moon JH, Choi SH, Lee HJ, Oh TJ. Alpha-Tocopherol-Loaded Liposomes Reduce High Glucose Induced Oxidative Stress in Schwann Cells: A Proof of Concept Study. Diabetes Metab J 2025; 49:507-512. [PMID: 39908988 PMCID: PMC12086562 DOI: 10.4093/dmj.2024.0489] [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: 08/19/2024] [Accepted: 10/23/2024] [Indexed: 02/07/2025] Open
Abstract
Although oxidative stress is the main pathophysiology of the development of diabetic neuropathy, oral administration of antioxidants has given disappointing results. Here, we hypothesized that local delivery of antioxidants would provide protective effects on Schwann cells due to the high concentration of local lesions. We prepared alpha-tocopherol (ATF)-loaded liposomes and tested their skin penetration after sonication. An in vitro study using IMS-32 cells was conducted to determine the level of reactive oxygen species (ROS) scavenging effects of ATF-liposomes. ATF reduced ROS in high-glucose-exposed IMS-32 cells in a dosedependent manner. ATF-liposomes also reduced the ROS level in vitro and ultrasound irradiation enhanced delivery to the dermis in porcine ear skin. This study showed that it is feasible to deliver ATF through the skin and can effectively reduce ROS. This model is worthy of development for clinical use.
Collapse
Affiliation(s)
- Jee-In Heo
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Mi Jeong Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Daehyun Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Nano Science and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Jimin Seo
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Joon Ho Moon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hak Jong Lee
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
- Department of Medical Device Development, Seoul National University College of Medicine, Seoul, Korea
- Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Tae Jung Oh
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Guo X, Zhang Y, Liu Q, Xu M, Pang J, Yang B, Rong S, Yang X. Progress on liposome delivery systems in the treatment of bladder cancer. RSC Adv 2025; 15:14315-14336. [PMID: 40330044 PMCID: PMC12053965 DOI: 10.1039/d5ra00746a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Bladder cancer (BC) in the urinary system remains one of the most prevalent malignancies with high recurrence rate globally. Current treatment schemes against BC such as surgery, chemotherapy, and radiotherapy have substantial limitations including side effects, drug resistance, and poor tumor targeting. Considering the above-mentioned challenges, nanotechnology has become a current research hotspot, particularly liposome-based drug delivery systems, which offer promising novel therapeutic strategies aimed at reducing systemic toxicity, overcoming drug resistance, and enhancing drug targeting. This review systematically elaborates the current research progress on liposomal drug delivery systems in BC treatment, focusing on their application in chemotherapy, immunotherapy, and gene therapy. Additionally, we provide a comprehensive assessment of the benefits and limitations of liposome nanocarriers used in BC treatment. The advanced targeting strategies and combination treatments via liposomal therapies are also discussed, demonstrating that liposomal formulations have great potential application value in the treatment of BC owing to their superior bioavailability, stability, and targeting and minimal adverse effects.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of Urology Surgery, First Hospital of Shanxi Medical University Taiyuan Shanxi 030001 China
- The First Clinical Medical College of Shanxi Medical University Taiyuan Shanxi 030001 China
| | - Yan Zhang
- School of Optoelectronic Engineering, Xi'an Technological University Xi'an 710021 China
| | - Quanyong Liu
- Department of Urology Surgery, First Hospital of Shanxi Medical University Taiyuan Shanxi 030001 China
- The First Clinical Medical College of Shanxi Medical University Taiyuan Shanxi 030001 China
| | - Mingquan Xu
- Department of Urology Surgery, First Hospital of Shanxi Medical University Taiyuan Shanxi 030001 China
- The First Clinical Medical College of Shanxi Medical University Taiyuan Shanxi 030001 China
| | - Jianzhi Pang
- The First Clinical Medical College of Shanxi Medical University Taiyuan Shanxi 030001 China
| | - Bin Yang
- Department of Urology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital Taiyuan 030032 China
| | - Shuo Rong
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital Taiyuan 030032 China
| | - Xiaofeng Yang
- Department of Urology Surgery, First Hospital of Shanxi Medical University Taiyuan Shanxi 030001 China
- The First Clinical Medical College of Shanxi Medical University Taiyuan Shanxi 030001 China
| |
Collapse
|
31
|
Huang L, Ni W, Jia Y, Zhu M, Yang T, Yu M, Zhang J. Process Development for the Continuous Manufacturing of Carbamazepine-Nicotinamide Co-Crystals Utilizing Hot-Melt Extrusion Technology. Pharmaceutics 2025; 17:568. [PMID: 40430860 PMCID: PMC12115052 DOI: 10.3390/pharmaceutics17050568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Objectives: Hot-melt extrusion (HME) offers a solvent-free, scalable approach for manufacturing pharmaceutical co-crystals (CCs), aligning with the industry's shift to continuous manufacturing (CM). However, challenges like undefined yield optimization, insufficient risk management, and limited process analytical technology (PAT) integration hinder its industrial application. This study aimed to develop a proof-of-concept HME platform for CCs, assess process risks, and evaluate PAT-enabled monitoring to facilitate robust production. Methods: Using carbamazepine (CBZ) and nicotinamide (NIC) as model compounds, an HME platform compatible with PAT tools was established. A systematic risk assessment identified five key risk domains: materials, machinery, measurement, methods, and other factors. A Box-Behnken design of experiments (DoE) evaluated the impact of screw speed, temperature, and mixing sections on CC quality. Near-infrared (NIR) spectroscopy monitored CBZ-NIC co-crystal formation in real time during HME process. Results: DoE revealed temperature and number of mixing sections significantly influenced particle size (D50: 2.0-4.0 μm), while screw speed affected efficiency. NIR spectroscopy detected a unique CC absorption peak at 5008.3 cm⁻¹, enabling real-time structural monitoring with high accuracy (R² = 0.9999). Risk assessment highlighted material attributes, process parameters, and equipment design as critical factors affecting CC formation. All experimental batches yielded ≥ 94% pure CCs with no residual starting materials, demonstrating process reproducibility and robustness. Conclusions: Overall, this work successfully established a continuous hot-melt extrusion (HME) process for manufacturing CBZ-NIC co-crystals, offering critical insights into material, equipment, and process parameters while implementing robust in-line NIR monitoring for real-time quality control. Additionally, this work provides interpretable insights and serves as a basis for future machine learning (ML)-driven studies.
Collapse
Affiliation(s)
- Lianghao Huang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Pharmaceutical Products Research and Development Center, Marine Biomedical Research Institute of Qingdao, Qingdao 266137, China
| | - Wen Ni
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Pharmaceutical Products Research and Development Center, Marine Biomedical Research Institute of Qingdao, Qingdao 266137, China
| | - Yaru Jia
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Pharmaceutical Products Research and Development Center, Marine Biomedical Research Institute of Qingdao, Qingdao 266137, China
| | - Minqing Zhu
- Material Characterization, Thermo Fisher Scientific, Shanghai 201203, China;
| | - Tiantian Yang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Pharmaceutical Products Research and Development Center, Marine Biomedical Research Institute of Qingdao, Qingdao 266137, China
| | - Mingchao Yu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Pharmaceutical Products Research and Development Center, Marine Biomedical Research Institute of Qingdao, Qingdao 266137, China
| | - Jiaxiang Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Pharmaceutical Products Research and Development Center, Marine Biomedical Research Institute of Qingdao, Qingdao 266137, China
| |
Collapse
|
32
|
Asghar S, Iliescu R, Stiufiuc RI, Dragoi B. Co-Encapsulation of Multiple Antineoplastic Agents in Liposomes by Exploring Microfluidics. Int J Mol Sci 2025; 26:3820. [PMID: 40332493 PMCID: PMC12027889 DOI: 10.3390/ijms26083820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
The inherent complexity of cancer proliferation and malignancy cannot be addressed by the conventional approach of relying on high doses of a single powerful anticancer agent, which is associated with poor efficacy, higher toxicity, and the development of drug resistance. Multiple drug therapy (MDT) rationally designed to target tumor heterogeneity, block alternative survival pathways, modulate the tumor microenvironment, and reduce toxicities would be a viable solution against cancer. Liposomes are the most suitable carrier for anticancer MDT due to their ability to encapsulate both hydrophilic and hydrophobic agents, biocompatibility, and controlled release properties; however, an adequate manufacturing method is important for effective co-encapsulation. Microfluidics involves the manipulation of fluids at the microscale for the controlled synthesis of liposomes with desirable properties. This work critically reviews the use of microfluidics for the synthesis of anticancer MDT liposomes. MDT success not only relies on the identification of synergistic dose combinations of the anticancer modalities but also warrants the loading of multiple therapeutic entities within liposomes in optimal ratios, the protection of the drugs by the nanocarrier during systemic circulation, and the synchronous release at the target site in the same pattern as confirmed in preliminary efficacy studies. Prospects have been identified for the bench-to-bedside translation of anticancer MDT liposomes using microfluidics.
Collapse
Affiliation(s)
- Sajid Asghar
- Nanotechnology Laboratory, TRANSCEND Department, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot, 700483 Iași, Romania;
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Radu Iliescu
- Proteomics Laboratory, TRANSCEND Research Center, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot Street, 700483 Iași, Romania
- Department of Pharmacology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 16 University Street, 700115 Iași, Romania
| | - Rares-Ionut Stiufiuc
- Nanotechnology Laboratory, TRANSCEND Department, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot, 700483 Iași, Romania;
- Department of NanoSciences, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hatieganu” University of Medicine and Pharmacy, 4-6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Brindusa Dragoi
- Nanotechnology Laboratory, TRANSCEND Department, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot, 700483 Iași, Romania;
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iași, 11 Bd. Carol I, 700506 Iași, Romania
| |
Collapse
|
33
|
Honrath S, Burger M, Leroux JC. Hurdles to healing: Overcoming cellular barriers for viral and nonviral gene therapy. Int J Pharm 2025; 674:125470. [PMID: 40112901 DOI: 10.1016/j.ijpharm.2025.125470] [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: 01/20/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Gene delivery offers great potential for treating various diseases, yet its success requires overcoming several biological barriers. These hurdles span from extracellular degradation, reaching the target cells, and inefficient cellular uptake to endosomal entrapment, cytoplasmic transport, nuclear entry, and transcription limitations. Viruses and non-viral vectors deal with these barriers via different mechanisms. Viral vectors, such as adenoviruses, adeno-associated viruses, and lentiviruses use natural mechanisms to efficiently deliver genetic material but face limitations including immunogenicity, cargo capacity, and production complexity. Nonviral vectors, including lipid nanoparticles, polymers, and protein-based systems, offer scalable and safer alternatives but often fall short in overcoming intracellular barriers and achieving high transfection efficiencies. Recent advancements in vector engineering have partially overcome several of these challenges. Ionizable lipids improve endosomal escape while minimizing toxicity. Biodegradable polymers balance efficacy with safety, and engineered protein systems, inspired by viral or bacterial entry mechanisms, integrate multifunctionality for enhanced delivery. Despite these advances, challenges, particularly in achieving robust in vivo translatability, scalability, and reduced immunogenicity, remain. This review synthesizes current knowledge of cellular barriers and the approaches to overcome them, providing a roadmap for designing more efficient gene delivery systems. By addressing these barriers, the field can advance toward safer, and more effective therapies.
Collapse
Affiliation(s)
- Steffen Honrath
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Michael Burger
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland.
| | - Jean-Christophe Leroux
- ETH Zurich, Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland.
| |
Collapse
|
34
|
Yang S, Guo J, Huang S, Sun Z, Yang M, Peng Y. Edible ginseng-derived exosomes as drug delivery vehicles reduce the dose and improve the anti-cancer effect of CDDP. Biochem Biophys Res Commun 2025; 758:151658. [PMID: 40121965 DOI: 10.1016/j.bbrc.2025.151658] [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: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Nanotechnology and nanomaterials have emerged as promising tools for the delivery of anti-tumor drug cisplatin (CDDP). However, concerns exist regarding potential toxicity and cost-effectiveness, limiting their clinical applications. In contrast, plant-derived exosomes (PDEs), as natural nanovesicles, offer significant advantages as drug delivery carriers due to their large-scale production, biocompatibility, and ability to efficiently transport therapeutic drug across cellular barriers. In this work, we established a ginseng-derived exosome (G-Exo)-based CDDP delivery system (G-CDDP) and evaluated its anti-tumor efficacy both in vitro and in vivo. The results demonstrated that G-CDDP effectively targeted tumor site, inhibiting the proliferation and migration and promoting apoptosis in U-87MG tumor cells. Notably, the amount of CDDP in G-CDDP required for achieving the same cytotoxic effect on tumor cells was 12.66 times lower than that of free CDDP. In U-87MG tumor-bearing mice, G-CDDP effectively targeted tumor sites and exhibited significant therapeutic effect. Collectively, these findings highlight the potent anti-tumor activity of G-CDDP at reduced CDDP dosage, positioning it as a promising and efficient alternative to conventional drug treatments in clinical settings.
Collapse
Affiliation(s)
- Shuiyue Yang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, 130112, China; School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin, 130012, China
| | - Jia Guo
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, 130112, China
| | - Shan Huang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, 130112, China; School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin, 130012, China
| | - Zepeng Sun
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, 130112, China; School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin, 130012, China
| | - Min Yang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, 130112, China.
| | - Yinghua Peng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, 130112, China.
| |
Collapse
|
35
|
Khalid R, Mahmood S, Mohamed Sofian Z, Chik Z, Ge Y. Development of Rapidly Dissolving Microneedles Integrated with Valsartan-Loaded Nanoliposomes for Transdermal Drug Delivery: In Vitro and Ex Vivo Evaluation. Pharmaceutics 2025; 17:483. [PMID: 40284478 PMCID: PMC12030157 DOI: 10.3390/pharmaceutics17040483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Hypertension (HTN) is recognized as a major risk factor for cardiovascular disease, chronic kidney disease, and peripheral artery disease. Valsartan (VAL), an angiotensin receptor blocker drug for hypertension, has been limited due to its poor solubility and poor absorption from the GIT, which leads to low oral bioavailability. Objectives/Method: In the present research, firstly, VAL-loaded nanoliposomes were formulated and optimized using the Box-Behnken design (BBD). Optimized VAL-nanoliposomes were physically characterized and their fate was examined by scanning and transmission microscopy, DSC, FTIR, XRD, and ex vivo studies using rat skin. In vitro studies using human keratinocyte (HaCaT) cells showed a decrease in cell viability as the liposome concentration increased. Secondly, the formulation of VAL-loaded nanoliposomes was integrated into dissolvable microneedles (DMNs) to deliver the VAL transdermally, crossing the skin barrier for better systemic delivery. Results: The optimized nanoliposomes showed a vesicle size of 150.23 (0.47) nm, a ZP of -23.37 (0.50) mV, and an EE% of 94.72 (0.44)%. The DMNs were fabricated using a ratio of biodegradable polymers, sodium alginate (SA), and hydroxypropyl methylcellulose (HPMC). The resulting VAL-LP-DMNs exhibited sharp pyramidal microneedles, adequate mechanical properties, effective skin insertion capability, and rapid dissolution of the microneedles in rat skin. In the ex vivo analysis, the transdermal flux of VAL was significantly (5.36 (0.39) μg/cm2/h) improved by VAL-LP-DMNs. The enhancement ratio of the VAL-LP-DMNs was 1.85. In conclusion, liposomes combined with DMNs have shown high potential and bright prospects as carriers for the transdermal delivery of VAL. Conclusions: These DMNs can be explored in studies focused on in vivo evaluations to confirm their safety, pharmacokinetics profile, and pharmacodynamic efficacy.
Collapse
Affiliation(s)
- Ramsha Khalid
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (R.K.); (Z.M.S.)
| | - Syed Mahmood
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (R.K.); (Z.M.S.)
- Universiti Malaya-Research Centre for Biopharmaceuticals and Advanced Therapeutics (UBAT), Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Centre of Advanced Materials (CAM), Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Zarif Mohamed Sofian
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (R.K.); (Z.M.S.)
- Universiti Malaya-Research Centre for Biopharmaceuticals and Advanced Therapeutics (UBAT), Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Zamri Chik
- Universiti Malaya-Research Centre for Biopharmaceuticals and Advanced Therapeutics (UBAT), Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Yi Ge
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK
| |
Collapse
|
36
|
Li SS, Guo M, Zhao Y, Fan F, Huang S, Yang H, Chen X, Jin X. Intranasal delivery of R8-modified circNFXL1 liposomes ameliorates Su5416-induced pulmonary arterial hypertension in C57BL/6 mice. Respir Res 2025; 26:127. [PMID: 40189516 PMCID: PMC11972480 DOI: 10.1186/s12931-025-03203-y] [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: 12/03/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a progressive, life-threatening condition characterized by increased pulmonary vascular resistance and right ventricular hypertrophy (RVH). Current treatments primarily alleviate symptoms but do not effectively target the underlying molecular mechanisms driving the disease. This study aimed to evaluate the therapeutic potential of R8-modified liposomal delivery of circNFXL1, a circular RNA, in a mouse model of PAH. METHODS R8-circNFXL1 liposomes were synthesized and characterized for their physicochemical properties, including encapsulation efficiency. PAH was induced in C57BL/6 mice using a combination of subcutaneous Su5416 administration and hypoxic exposure. Intranasal delivery of R8-circNFXL1 was performed, and therapeutic effects were assessed using echocardiography and hemodynamic measurements. Molecular mechanisms were explored through analysis of the miR-29b/Kcnb1 axis, a regulatory pathway in PAH. RESULTS The R8-circNFXL1 liposomes demonstrated optimal physicochemical properties, including high encapsulation efficiency. Treatment with R8-circNFXL1 significantly reduced RVH, improved cardiac function, and mitigated pulmonary vascular remodeling compared to untreated PAH controls. Molecular analysis revealed that R8-circNFXL1 modulated the miR-29b/Kcnb1 axis, providing insights into its mechanism of action. CONCLUSIONS R8-circNFXL1 liposomes offer a promising, targeted therapeutic strategy for PAH by addressing underlying molecular mechanisms. This approach has potential implications for developing alternative treatments to improve disease management and outcomes in PAH.
Collapse
Affiliation(s)
- Shan-Shan Li
- School of Medicine, Nankai University, Tianjin, China.
| | - Miao Guo
- School of Medicine, Nankai University, Tianjin, China
| | - Ying Zhao
- School of Medicine, Nankai University, Tianjin, China
| | - Feifei Fan
- School of Medicine, Nankai University, Tianjin, China
| | | | | | - Xu Chen
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
| | - Xin Jin
- School of Medicine, Nankai University, Tianjin, China.
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China.
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China.
| |
Collapse
|
37
|
Lin M, Tang K, Zheng W, Zheng S, Hu K. Curcumin delivery system based on a chitosan-liposome encapsulated zeolitic imidazolate framework-8: a potential treatment antioxidant and antibacterial treatment after phacoemulsification. Biomed Mater 2025; 20:035013. [PMID: 40081008 DOI: 10.1088/1748-605x/adc05c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/13/2025] [Indexed: 03/15/2025]
Abstract
Curcumin is a natural polyphenol extracted from plants that can interact with various molecular targets, including antioxidant, antibacterial, anticancer, and anti-aging activities. Due to its variety of pharmacological activities and large margin pf safety, curcumin has been used in the prevention and treatment of various diseases, such as Alzheimer's, heart, and rheumatic immune diseases. To develop curcumin eye drops that can be used as antioxidant and antibacterial agents after phacoemulsification, we have designed a nano-based drug delivery system to improve curcumin bioavailability and duration of action. We successfully prepared zeolitic imidazolate framework-8 (ZIF-8) coated with chitosan-liposome (Cur@ZIF-8/CS-Lip) for curcumin delivery. It can release curcumin for over 20 hin vitroand exhibits excellent biosafety, antioxidant, and antibacterial activities. Therefore, we hypothesized that Cur@ZIF-8/CS-Lip could reduce the incidence of oxidative stress and infection after cataract surgery.
Collapse
Affiliation(s)
- Meiting Lin
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Kunyuan Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Wendi Zheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Shijie Zheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on major blinding diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing 400016, People's Republic of China
| |
Collapse
|
38
|
Nguyen TT, Dao TK, Nguyen VT, Pham DT. Designing Effective Drug Therapies Using a Multiobjective Spider-Wasp Optimizer. Biomimetics (Basel) 2025; 10:219. [PMID: 40277618 PMCID: PMC12025240 DOI: 10.3390/biomimetics10040219] [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: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Designing effective drug therapies requires balancing competing objectives, such as therapeutic efficacy, safety, and cost efficiency-a task that poses significant challenges for conventional optimization methods. To address this, we propose the multi-objective spider-wasp optimizer (MOSWO), a novel approach uniquely emulating the cooperative predation dynamics between spiders and wasps observed in nature. MOSWO integrates adaptive mechanisms for exploration and exploitation to resolve complex trade-offs in multiobjective drug design. Unlike existing approaches, the algorithm employs a dynamic population-partitioning strategy inspired by predator-prey interactions, enabling efficient Pareto frontier discovery. We validate MOSWO's performance through extensive experiments on synthetic benchmarks and real-world case studies spanning antiviral and antibiotic therapies. Results demonstrate that MOSWO surpasses state-of-the-art methods (NSGA-II, MOEA/D, MOGWO, and MOPSO), achieving 11% higher hypervolume scores, 8% lower inverted generational distance scores, 9% higher spread scores, a 30% faster convergence, and superior robustness against noisy biological datasets. The framework's adaptability to diverse therapeutic scenarios underscores its potential as a transformative tool for computational pharmacology.
Collapse
Affiliation(s)
- Trong-The Nguyen
- School of Electronic Engineering, Fuzhou Institute of Technology, Fuzhou 350506, China;
| | - Thi-Kien Dao
- School of Electronic Engineering, Fuzhou Institute of Technology, Fuzhou 350506, China;
| | - Van-Thien Nguyen
- School of Information and Communication Technology, Hanoi University of Industry, Hanoi 10000, Vietnam; (V.-T.N.); (D.-T.P.)
| | - Duc-Tinh Pham
- School of Information and Communication Technology, Hanoi University of Industry, Hanoi 10000, Vietnam; (V.-T.N.); (D.-T.P.)
| |
Collapse
|
39
|
Patle RY, Dongre RS. Recent advances in PAMAM mediated nano-vehicles for targeted drug delivery in cancer therapy. J Drug Target 2025; 33:437-457. [PMID: 39530737 DOI: 10.1080/1061186x.2024.2428966] [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: 02/27/2024] [Revised: 10/02/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
3-D multi-faceted, nano-globular PAMAM dendritic skeleton is a highly significant polymer that offers applications in biomedical, industrial, environmental and agricultural fields. This is mainly due to its enhanced properties, including adjustable surface functionalities, biocompatibility, non-toxicity, high uniformity and reduced cytotoxicity, as well as its numerous internal cavities. This trait inspires further exploration and advancements in tailoring approaches. The implementation of deliberate strategic modifications in the morphological characteristics of PAMAM is crucial through chemical and biological interventions, in addition to its therapeutic advancements. Thus, the production of peripheral groups remains a prominent and highly advanced technique in molecular fabrication, aimed at boosting the potential of PAMAM conjugates. Currently, there exist numerous dendritic-hybrid materials, despite the widespread use of PAMAM-conjugated frameworks as drug delivery systems, which are well regarded for their efficacy in enhancing potency through the incorporation of surface functions. This paper provides a comprehensive review of recent progress in the design and assembly of various components of PAMAM conjugates, focusing on their unique formulations. The review encompasses synthetic methodologies, a thorough evaluation of their applicability, and an analysis of their potential functions in the context of Drug Delivery Systems (DDS) in the current period.
Collapse
Affiliation(s)
- Ramkrishna Y Patle
- PGTD Chemistry, RTM Nagpur University, Nagpur, India
- Mahatma Gandhi College of Science, Chandrapur, India
| | | |
Collapse
|
40
|
Samad MA, Ahmad I, Hasan A, Alhashmi MH, Ayub A, Al‐Abbasi FA, Kumer A, Tabrez S. STAT3 Signaling Pathway in Health and Disease. MedComm (Beijing) 2025; 6:e70152. [PMID: 40166646 PMCID: PMC11955304 DOI: 10.1002/mco2.70152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a critical transcription factor involved in multiple physiological and pathological processes. While STAT3 plays an essential role in homeostasis, its persistent activation has been implicated in the pathogenesis of various diseases, particularly cancer, bone-related diseases, autoimmune disorders, inflammatory diseases, cardiovascular diseases, and neurodegenerative conditions. The interleukin-6/Janus kinase (JAK)/STAT3 signaling axis is central to STAT3 activation, influencing tumor microenvironment remodeling, angiogenesis, immune evasion, and therapy resistance. Despite extensive research, the precise mechanisms underlying dysregulated STAT3 signaling in disease progression remain incompletely understood, and no United States Food and Drug Administration (USFDA)-approved direct STAT3 inhibitors currently exist. This review provides a comprehensive evaluation of STAT3's role in health and disease, emphasizing its involvement in cancer stem cell maintenance, metastasis, inflammation, and drug resistance. We systematically discuss therapeutic strategies, including JAK inhibitors (tofacitinib, ruxolitinib), Src Homology 2 domain inhibitors (S3I-201, STATTIC), antisense oligonucleotides (AZD9150), and nanomedicine-based drug delivery systems, which enhance specificity and bioavailability while reducing toxicity. By integrating molecular mechanisms, disease pathology, and emerging therapeutic interventions, this review fills a critical knowledge gap in STAT3-targeted therapy. Our insights into STAT3 signaling crosstalk, epigenetic regulation, and resistance mechanisms offer a foundation for developing next-generation STAT3 inhibitors with greater clinical efficacy and translational potential.
Collapse
Affiliation(s)
- Md Abdus Samad
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Iftikhar Ahmad
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Aakifah Hasan
- Department of BiochemistryFaculty of Life ScienceAligarh Muslim UniversityAligarhIndia
| | - Mohammad Hassan Alhashmi
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
- Department of Medical Laboratory SciencesFaculty of Applied Medical SciencesKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Arusha Ayub
- Department of MedicineCollege of Health SciencesUniversity of GeorgiaGeorgiaUSA
| | - Fahad A. Al‐Abbasi
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Ajoy Kumer
- Department of ChemistryCollege of Arts and SciencesInternational University of Business Agriculture & Technology (IUBAT)DhakaBangladesh
| | - Shams Tabrez
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
- Department of Medical Laboratory SciencesFaculty of Applied Medical SciencesKing Abdulaziz UniversityJeddahSaudi Arabia
| |
Collapse
|
41
|
Gu P, Liu J, Gao T, Ma Q, Gao S, Li N, Zhang W, Xia Z, Yang Q, Mu W, Liang S, Fu S, Yuan S, Wei S, Liu J, Yang Y, Yan X, Liu Y, Wang C, Zhang N. Temperature-Sensitive Nano-GOx Combined with Downregulation of Tumor Stemness to Initiate Robust Antitumor Efficacy. ACS NANO 2025; 19:11738-11755. [PMID: 40105115 DOI: 10.1021/acsnano.4c12962] [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: 03/20/2025]
Abstract
GOx-mediated glucose depletion offers an alternative noninvasive strategy for tumor therapy, but its lower catalytic activity in vivo limits its clinical application. Herein, we designed a temperature-sensitive nano-GOx (NG) that was constructed by gold nanorods chemically modified with GOx (AuNRs-GOx) and coated with temperature-sensitive lipids. The chemical linkage could maintain the natural conformation of GOx, ensuring that NG exerted powerful catalytic activity within the tumor and initiated antitumor immune response through moderate starvation and mild photothermal therapy (mPTT) to coregulating dendritic cells (DCs) and tumor-associated macrophages (TAMs). Ulteriorly, VTNG was obtained by NG coloading with verteporfin (VP) and evofosfamide (TH-302). VTNG demonstrated temperature-sensitive triggered drug release when exposed to near-infrared laser irradiation. NG exacerbated the degree of TME hypoxia and facilitated the activation of TH-302. Meanwhile, VP enhanced tumor cell sensitivity by decreasing the stemness of the tumor cells, thus realizing the effective killing of tumor cells and further enhancing the therapeutic effect of NG. Notably, VTNG had a significant antitumor effect in melanoma models compared with first-line melanoma therapy and formed an immune memory effect. In conclusion, VTNG provided an effective approach to enhance the therapeutic effect of GOx for tumor treatment.
Collapse
Affiliation(s)
- Panpan Gu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Jinhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Tong Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Shuying Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Nan Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Weihan Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Zhenxing Xia
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Qinglin Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Shuang Liang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Shijun Yuan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Suyun Wei
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Jie Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Yuxin Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Xiaoxin Yan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| |
Collapse
|
42
|
Szecskó A, Mészáros M, Simões B, Cavaco M, Chaparro C, Porkoláb G, Castanho MARB, Deli MA, Neves V, Veszelka S. PepH3-modified nanocarriers for delivery of therapeutics across the blood-brain barrier. Fluids Barriers CNS 2025; 22:31. [PMID: 40170024 PMCID: PMC11959756 DOI: 10.1186/s12987-025-00641-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/14/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Nanocarriers targeting the blood-brain barrier (BBB) are promising drug delivery systems to enhance the penetration of therapeutic molecules into the brain. Immunotherapy, particularly monoclonal antibodies designed to bind amyloid-beta peptides have become a promising strategy for Alzheimer's disease, but ensuring efficacy and safety is challenging and crucial for these therapies. Our aim was to develop an innovative nanocarriers conjugated with PepH3, a cationic peptide derived from Dengue virus type-2 capsid protein that crosses the BBB and acts as a shuttle peptide for the encapsulated single domain antibody (sdAb) recognizing Aβ oligomers. RESULTS PepH3 peptide enhanced the uptake of the nanoparticles (NPs) into brain endothelial cells, and transcytosis of sdAb, as a potential therapeutic molecule, across both rat and human BBB culture models. The cargo uptake was a temperature dependent active process that was reduced by metabolic and endocytosis inhibitors. The cellular uptake of the cationic PepH3-tagged NPs decreased when the negative surface charge of brain endothelial cells became more positive after treatments with a cationic lipid or with neuraminidase by digesting the glycocalyx. The NPs colocalized mostly with endoplasmic reticulum and Golgi apparatus and not with lysosomes, indicating the cargo may avoid cellular degradation. CONCLUSIONS Our results support that combination of NPs with a potential brain shuttle peptide such as PepH3 peptide can improve the delivery of antibody fragments across the BBB.
Collapse
Affiliation(s)
- Anikó Szecskó
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Mária Mészáros
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
- One Health Institute, Faculty of Health Sciences, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Beatriz Simões
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marco Cavaco
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Chaparro
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Gergő Porkoláb
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mária A Deli
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Vera Neves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Szilvia Veszelka
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
43
|
Chen M, Liu X, Kong L, Yu Y, Zang J, Li X, Guo R, Zhang L, Liu Y. Efficacy assessment of glycyrrhetinic acid-modified liposomes loaded with doxorubicin hydrochloride and cucurbitine B for synergistic treatment of hepatocellular carcinoma. Int J Pharm 2025; 673:125360. [PMID: 39954971 DOI: 10.1016/j.ijpharm.2025.125360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/22/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is characterized by a high incidence rate, aggressive invasion and metastasis, and a significant postoperative recurrence rate. Targeted therapy plays a crucial role in the precise treatment of HCC. Studies have demonstrated that Glycyrrhetinic acid (GA) specific receptors are overexpressed on the surface of HCC cells. Doxorubicin hydrochloride (Dox), a widely used chemotherapy agent for anti-tumor treatment, but is associated with substantial toxic side effects. Cucurbitacin B (CuB) also demonstrates promising anti-tumor activity, but its poor water solubility and low bioavailability limit its clinical application. The combination of Dox and CuB can exert a synergistic effect, thereby enhancing the overall anti-tumor efficacy. Therefore, we have developed GA-modified liposomes loaded with Dox and CuB (GA-Dox/CuB-Lips) to achieve synergistic therapy for HCC. METHOD In this study, GA-Dox/CuB-Lips were prepared using the thin film dispersion method and ammonium sulfate gradient method. In vitro, we evaluated the cellular uptake and cytotoxicity of the liposomes, as well as their anti-tumor effects in inhibiting tumor proliferation, promoting tumor apoptosis, and suppressing invasion and metastasis. In vivo, the targeting properties of GA-Dox/CuB-Lips were assessed through in vivo imaging. A tumor growth curve was generated by establishing a heterotopic nude mouse model. Additionally, an in-situ HCC model was established and the anti-tumor effects of liposomes were evaluated using HE staining, histological analysis and immunofluorescence staining. RESULTS We successfully prepared GA-Dox/CuB-Lips with a smooth, spherical morphology and uniform distribution. Both drugs exhibited high encapsulation efficiency, significantly enhancing the solubility of CuB. In vitro, GA-Dox/CuB-Lips demonstrated excellent targeting properties and exerted cytotoxic effects on Hepa1-6 cells, effectively inhibiting tumor cell proliferation, invasion, and metastasis while promoting tumor cell apoptosis. In vivo, GA-Dox/CuB-Lips selectively targeted tumor sites, disrupted tumor structures, inhibited tumor growth and proliferation, and promoted apoptosis. CONCLUSION GA-Dox/CuB-Lips exhibited excellent anti-HCC activity and represent a promising therapeutic approach for the treatment of HCC.
Collapse
Affiliation(s)
- Muhan Chen
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Xinze Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China; Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Shenyang 110000, China
| | - Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Xuetao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Ruibo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China.
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Shenyang Key Laboratory of Targeted Delivery of Chinese medicine, Shenyang 110000, China.
| |
Collapse
|
44
|
Mamidi N, Franco De Silva F, Orash Mahmoudsalehi A. Advanced disease therapeutics using engineered living drug delivery systems. NANOSCALE 2025; 17:7673-7696. [PMID: 40040419 DOI: 10.1039/d4nr05298f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Biological barriers significantly impede the delivery of nanotherapeutics to diseased tissues, diminishing therapeutic efficacy across pathologies such as cancer and inflammatory disorders. Although conventional strategies integrate multifunctional designs and molecular components into nanomaterials (NMs), many approaches remain insufficient to overcome these barriers. Key challenges, including inadequate drug accumulation at target sites and nonspecific biodistribution, persist in nanotherapeutic development. NMs, which harness the ability to precisely modulate drug delivery spatiotemporally and control release kinetics, represent a transformative platform for targeted cancer therapy. In this review, we highlight the biological obstacles limiting effective cancer treatment and evaluate how stimuli-responsive NMs address these constraints. By leveraging exogenous and endogenous stimuli, such NMs improve therapeutic specificity, reduce off-target effects, and amplify drug activity within pathological microenvironments. We systematically analyze the rational design and synthesis of stimuli-responsive NMs, driven by advances in oncology, biomaterials science, and nanoscale engineering. Furthermore, we highlight advances across NM classes-including polymeric, lipid-based, inorganic, and hybrid systems and explore functionalization approaches using targeting ligands, antibodies, and biomimetic coatings. Diverse delivery strategies are evaluated, such as small-molecule prodrug activation, peptide- and protein-based targeting, nucleic acid payloads, and engineered cell-mediated transport. Despite the promise of stimuli-responsive NMs, challenges such as biocompatibility, scalable fabrication, and clinical translation barriers must be addressed. By elucidating structure-function relationships and refining stimulus-triggered mechanisms, these NMs pave the way for transformative precision oncology strategies, enabling patient-specific therapies with enhanced efficacy and safety. This synthesis of interdisciplinary insights aims to catalyze innovation in next-generation nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Wisconsin Center for Nanobiosystems, School of Pharmacy, University of Wisconsin-Madison, Wisconsin-53705, USA.
| | - Fátima Franco De Silva
- Department of Food Engineering, Tecnologico de Monterrey, Monterrey, Nuevo Leon-64849, Mexico
| | - Amin Orash Mahmoudsalehi
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo Leon-64849, Mexico
| |
Collapse
|
45
|
Beheshtkhoo N, Jadidi Kouhbanani MA, Daghighi SM, Shakouri Nikjeh M, Esmaeili Z, Khosravani M, Adabi M. Effect of oral resveratrol-loaded nanoliposomes on hyperlipidemia via toll-like receptor 3 and TIR domain-containing adaptor inducing interferon-β protein expression in an animal model. J Liposome Res 2025:1-27. [PMID: 40098438 DOI: 10.1080/08982104.2025.2476529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/20/2025] [Accepted: 03/02/2025] [Indexed: 03/19/2025]
Abstract
Hyperlipidemia, a critical risk factor for various health conditions, necessitates innovative therapeutic strategies. Investigating the effectiveness of liposomal formulations in managing hyperlipidemia is essential. Resveratrol (RES)-loaded nanoliposomes present a promising new approach for hyperlipidemia treatment. In this study, we investigated the anti-hyperlipidemic potential of RES-loaded nanoliposomes in high-fat diet (HFD)-fed rats. The nanoliposomes were prepared using a thin-film hydration method. According to transmission electron microscopy (TEM) and dynamic light scattering (DLS) results, the mean size of prepared RES-loaded nanoliposomes were about 42 nm and 68 nm, respectively, with a zeta potential of -65.6 mV. The entrapment efficiency and loading content were 83.78% and 14.25%, respectively. Additionally, the RES-loaded nanoliposomes exhibited controlled release kinetics compared to the free RES form. Moreover, in a hyperlipidemic rat model induced by an HFD, orally administered RES-loaded nanoliposomes significantly reduced total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), and triglycerides (TG), while concurrently increasing high-density lipoprotein cholesterol (HDL-C) levels. Additionally, liver damage induced by HFD was alleviated by RES-loaded nanoliposomes. The expression levels of Toll-like receptor 3 (TLR3) and TIR domain-containing adaptor-inducing interferon-β (TRIF) were assessed using fluorescence immunohistochemistry. Notably, RES-loaded nanoliposomes significantly reduced the expression of these protein. The effect of RES-loaded nanoliposomes was measured on body weight of HFD rats, demonstrting RES-loaded nanoliposomes hold promise for weight management. These findings underscore the potential of RES-loaded nanoliposomes as a safe and effective therapeutic option for hyperlipidemia.
Collapse
Affiliation(s)
- Nasrin Beheshtkhoo
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Jadidi Kouhbanani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mojtaba Daghighi
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shakouri Nikjeh
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Esmaeili
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Gu X, Du L, Lin R, Ding Z, Guo Z, Wei J, Li Y. How Advanced Is Nanomedicine for Atherosclerosis? Int J Nanomedicine 2025; 20:3445-3470. [PMID: 40125442 PMCID: PMC11928726 DOI: 10.2147/ijn.s508757] [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: 11/26/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in nanotechnology have opened new avenues for precision therapy, personalized medicine, and multifunctional theranostics in atherosclerosis (AS). This review provides a comprehensive overview of the role of nanoparticles (NPs) in precision medicine for AS, discussing their applications, challenges, and future prospects. The review first analyzes the current treatment landscape of AS and outlines potential biological targets for therapy. Various nanocarriers, including organic, inorganic, and hybrid systems, are evaluated for their therapeutic potential, with a focus on targeted drug delivery, anti-inflammatory therapy, vascular repair, plaque stabilization, and lipid clearance. Additionally, the review explores NP preparation methods, emphasizing strategies to enhance drug loading, stability, and controlled release. Finally, the translational challenges of NP-based therapies, including biocompatibility, large-scale production, regulatory hurdles, and clinical implementation, are critically analyzed. Future directions highlight the importance of interdisciplinary collaboration and technological innovation in advancing nanoparticle-based precision medicine for AS.
Collapse
Affiliation(s)
- Xiang Gu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Lixin Du
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ruifang Lin
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zehui Ding
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhihua Guo
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiaming Wei
- School of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ya Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
47
|
Liao T, Chen X, Qiu F, Zhang X, Wu F, Zhao Z, Xu M, Chen M, Shen JW, Shen Q, Ji J. Regulation of cancer-associated fibroblasts for enhanced cancer immunotherapy using advanced functional nanomedicines: an updated review. J Nanobiotechnology 2025; 23:166. [PMID: 40038745 PMCID: PMC11877876 DOI: 10.1186/s12951-025-03217-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The tumor microenvironment (TME) is a complex and dynamic ecosystem that plays a critical role in cancer progression. It comprises various cell types, including immune cells, tumor cells, and stromal cells. Among these, cancer-associated fibroblasts (CAFs) represent a heterogeneous population with diverse origins, phenotypes, and functions. Activated CAFs secrete multiple factors that promote tumor growth, migration, angiogenesis, and contribute to chemoresistance. Additionally, CAFs secrete extracellular matrix (ECM) components, such as collagen, which form a physical barrier that hinders the penetration of chemotherapeutic and immunotherapeutic agents. This ECM also influences immune cell infiltration, impeding their ability to effectively target tumor cells. As a result, modulating the activity of CAFs has emerged as a promising strategy to enhance the efficacy of tumor immunotherapy. Nano-delivery systems, constructed from various nanomaterials with high targeting specificity and biocompatibility, offer a compelling approach to deliver therapeutic agents or immunomodulatory factors directly to CAFs. This modulation can alter CAF function, reduce their tumor-promoting effects, and thereby improve the outcomes of immunotherapy. This review provides an in-depth exploration of the origins, functions, and interactions of CAFs within the TME, particularly in the context of immune suppression. Furthermore, it discusses the potential applications of functional nanocarrifers in modulating CAFs and enhancing the effectiveness of tumor immunotherapy, highlighting the significant progress and potential of nanotechnology in this area.
Collapse
Affiliation(s)
- Tingting Liao
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China
| | - Xiaoxiao Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Fengkai Qiu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Xinyu Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| | - Fazong Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Ming Xu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| | - Jia-Wei Shen
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Qiying Shen
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Jiansong Ji
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China.
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|
48
|
Baruah N, Midya J, Gompper G, Dasanna AK, Auth T. Adhesion-driven vesicle translocation through membrane-covered pores. Biophys J 2025; 124:740-752. [PMID: 39863923 PMCID: PMC11897550 DOI: 10.1016/j.bpj.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/24/2024] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
Translocation across barriers and through constrictions is a mechanism that is often used in vivo for transporting material between compartments. A specific example is apicomplexan parasites invading host cells through the tight junction that acts as a pore, and a similar barrier crossing is involved in drug delivery using lipid vesicles penetrating intact skin. Here, we use triangulated membranes and energy minimization to study the translocation of vesicles through pores with fixed radii. The vesicles bind to a lipid bilayer spanning the pore, the adhesion-energy gain drives the translocation, and the vesicle deformation induces an energy barrier. In addition, the deformation-energy cost for deforming the pore-spanning membrane hinders the translocation. Increasing the bending rigidity of the pore-spanning membrane and decreasing the pore size both increase the barrier height and shift the maximum to smaller fractions of translocated vesicle membrane. We compare the translocation of initially spherical vesicles with fixed membrane area and freely adjustable volume to that of initially prolate vesicles with fixed membrane area and volume. In the latter case, translocation can be entirely suppressed. Our predictions may help rationalize the invasion of apicomplexan parasites into host cells and design measures to combat the diseases they transmit.
Collapse
Affiliation(s)
- Nishant Baruah
- Theoretical Physics of Living Matter, Institute of Biological Information Processing and Institute for Advanced Simulation, Forschungszentrum Jülich, Jülich, Germany.
| | - Jiarul Midya
- Theoretical Physics of Living Matter, Institute of Biological Information Processing and Institute for Advanced Simulation, Forschungszentrum Jülich, Jülich, Germany; School of Basic Sciences, Indian Institute of Technology, Bhubaneswar, India.
| | - Gerhard Gompper
- Theoretical Physics of Living Matter, Institute of Biological Information Processing and Institute for Advanced Simulation, Forschungszentrum Jülich, Jülich, Germany.
| | - Anil Kumar Dasanna
- Department of Theoretical Physics and Center for Biophysics, Saarland University, Saarbrücken, Germany; INM-Leibniz Institute for New Materials, Saarbrücken, Germany; Department of Physical Sciences, Indian Institute of Science Education and Research Mohali, Knowledge City, Manauli, India.
| | - Thorsten Auth
- Theoretical Physics of Living Matter, Institute of Biological Information Processing and Institute for Advanced Simulation, Forschungszentrum Jülich, Jülich, Germany.
| |
Collapse
|
49
|
Zhang J, Jiang S, Jiang J, Liu Y. Global research landscape on nanotechnology in acute lung injury: a bibliometric analysis. Front Digit Health 2025; 7:1472753. [PMID: 40103738 PMCID: PMC11913875 DOI: 10.3389/fdgth.2025.1472753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
Background Acute lung injury is a common respiratory emergency that seriously affects the life, health and quality of life of patients, especially after the global COVID-19 pneumonia. The application of nanotechnology in acute lung injury is promising. In response to the knowledge explosion resulting from rapid publication growth, we applied bibliometric analysis to explore the research profile and thematic trends in the field. Methods Articles and reviews related to nanotechnology in acute lung injury from 2004 to 2023 were searched. Java-based Citespace, VOSviewer, and R software-based Bibiometrix were used to systematically evaluate publications by spatiotemporal distribution, author distribution, subject categories, topic distribution, references, and keywords. Results A total of 1,347 publications were included. The number of papers related to nanotechnology in acute lung injury has grown exponentially over the past 20 years. China was the most productive country out of all 53 countries, followed by the United States. The Chinese Academy of Sciences was the most productive institution with 76 papers. PARTICLE AND FIBRE TOXICOLOGY was the most productive journal. The top five high-frequency keywords were inflammation, oxidative stress, toxicity, in vitro, respiratory-distress-syndrome. And the top five emerging keywords were delivery, covid-19, extracellular vesicles, therapy, sars-cov-2. Drug delivery are the focus of current research. Two emerging research areas represented the development trends: novel nanocarriers with higher efficiency and lower biotoxicity, and the other is research related to impact of nanomaterials in the progression of acute lung injury. Conclusion The field of nanotechnology in acute lung injury has been in a period of rapid development in the last three years. Delivery,targeted delivery and exosm have been the focus of current research in this field. Two emerging research areas represented the development trends:novel nanocarriers with higher efficiency and lower biotoxicity such as extracellular vesicles, exosomes and solid lipid nanoparticles, and the other is research related to impact of nanomaterials in the progression of acute lung injury.
Collapse
Affiliation(s)
- Jian Zhang
- School of Medicine, Nankai University, Tianjin, China
- Department of Thoracic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shasha Jiang
- Department of Thoracic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Postgraduate School, Medical School of Chinese PLA, Beijing, China
| | - Jipeng Jiang
- Department of Thoracic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yang Liu
- School of Medicine, Nankai University, Tianjin, China
- Department of Thoracic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
50
|
Rastegar-Pouyani N, Farzin MA, Zafari J, Haji Abdolvahab M, Hassani S. Repurposing the anti-parasitic agent pentamidine for cancer therapy; a novel approach with promising anti-tumor properties. J Transl Med 2025; 23:258. [PMID: 40033361 PMCID: PMC11877826 DOI: 10.1186/s12967-025-06293-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/23/2025] [Indexed: 03/05/2025] Open
Abstract
Pentamidine (PTM) is an aromatic diamidine administered for infectious diseases, e.g. sleeping sickness, malaria, and Pneumocystis jirovecii pneumonia. Due to similarities of cellular mechanisms between human cells and such infections, PTM has also been proposed for repurposing in non-infectious diseases such as cancer. Indeed, by modulating different signaling pathways such as PI3K/AKT, MAPK/ERK, p53, PD-1/PD-L1, etc., PTM has been shown to inhibit different properties of cancer, including proliferation, invasion, migration, hypoxia, and angiogenesis, while inducing anti-tumor immune responses and apoptosis. Given the promising implications of PTM for cancer treatment, however, the clinical translation of PTM in cancer is not without certain challenges. In fact, clinical trials have shown that systemic administration of PTM can be concurrent with serious adverse effects, e.g. hypoglycemia. Therefore, to reduce the administered doses of PTM, lower the risk of adverse effects, and prevent any potential drug resistance, while maintaining the anti-tumor efficacy, two main strategies have been suggested. One is combination therapy that employs PTM in conjunction with other anti-cancer modalities, such as chemotherapy and radiotherapy, and attacks tumor cells with significant additive or synergistic anti-tumor effects. The other is developing PTM-loaded nanocarrier drug delivery systems e.g. pegylated liposomes, chitosan-coated niosomes, squalene-based nanoparticles, hyaluronated lipid-polymer hybrid nanoparticles, etc., that offer enhanced pharmacokinetic characteristics, including increased bioavailability, sit-targeting, and controlled/sustained drug release. This review highlights the anti-tumor properties of PTM that favor its repurposing for cancer treatment, as well as, PTM-based combination therapies and nanocarrier delivery systems which can enhance therapeutic efficacy and simultaneously reduce toxicity.
Collapse
Affiliation(s)
- Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Tehran University of Medical Sciences, Tehran, Iran.
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Mohammad Amin Farzin
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Jaber Zafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohadeseh Haji Abdolvahab
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Shokoufeh Hassani
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|