1
|
Salsabila S, Khairinisa MA, Wathoni N, Sufiawati I, Mohd Fuad WE, Khairul Ikram NK, Muchtaridi M. In vivo toxicity of chitosan-based nanoparticles: a systematic review. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2025; 53:1-15. [PMID: 39924869 DOI: 10.1080/21691401.2025.2462328] [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: 09/24/2024] [Revised: 01/08/2025] [Accepted: 01/27/2025] [Indexed: 02/11/2025]
Abstract
Chitosan nanoparticles have been extensively utilised as polymeric drug carriers in nanoparticles formulations due to their potential to enhance drug delivery, efficacy, and safety. Numerous toxicity studies have been previously conducted to assess the safety profile of chitosan-based nanoparticles. These toxicity studies employed various methodologies, including test animals, interventions, and different routes of administration. This review aims to summarise research on the safety profile of chitosan-based nanoparticles in drug delivery, with a focus on general toxicity tests to determine LD50 and NOAEL values. It can serve as a repository and reference for chitosan-based nanoparticles, facilitating future research and further development of drugs delivery system using chitosan nanoparticles. Publications from 2014 to 2024 were obtained from PubMed, Scopus, Google Scholar, and ScienceDirect, in accordance with the inclusion and exclusion criteria.The ARRIVE 2.0 guidelines were employed to evaluate the quality and risk-of-bias in the in vivo toxicity studies. The results demonstrated favourable toxicity profiles, often exhibiting reduced toxicity compared to free drugs or substances. Acute toxicity studies consistently reported high LD50 values, frequently exceeding 5000 mg/kg body weight, while subacute studies typically revealed no significant adverse effects. Various routes of administration varied, including oral, intravenous, intraperitoneal, inhalation, and topical, each demonstrating promising safety profiles.
Collapse
Affiliation(s)
- Shela Salsabila
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Padjadjaran University, Sumedang, Indonesia
| | - Miski Aghnia Khairinisa
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Irna Sufiawati
- Department of Oral Medicine, Faculty of Dentistry, Universitas Padjadjaran, Sumedang, Indonesia
| | - Wan Ezumi Mohd Fuad
- Programme of Biomedicine, School of Health Sciences, USM Health Campus, Kubang Kerian, Kelantan, Malaysia
| | | | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Padjadjaran University, Sumedang, Indonesia
| |
Collapse
|
2
|
Noury H, Rahdar A, Romanholo Ferreira LF, Jamalpoor Z. AI-driven innovations in smart multifunctional nanocarriers for drug and gene delivery: A mini-review. Crit Rev Oncol Hematol 2025; 210:104701. [PMID: 40086770 DOI: 10.1016/j.critrevonc.2025.104701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
The convergence of artificial intelligence (AI) and nanomedicine has revolutionized the design of smart multifunctional nanocarriers (SMNs) for drug and gene delivery, offering unprecedented precision, efficiency, and personalization in therapeutic applications. AI-driven approaches enhance the development of these nanocarriers by accelerating their design, optimizing drug loading and release kinetics, improving biocompatibility, and predicting interactions with biological barriers. This review explores the transformative role of AI in the fabrication and functionalization of SMNs, emphasizing its impact on overcoming challenges in targeted drug delivery, controlled release, and theranostics. We discuss the integration of AI with advanced nanomaterials-such as polymeric, lipidic, and inorganic nanoparticles-highlighting their potential in oncology and hematology. Furthermore, we examine recent clinical and preclinical case studies demonstrating AI-assisted nanocarrier development for personalized medicine. The synergy between AI and nanotechnology paves the way for next-generation precision therapeutics, addressing critical limitations in traditional drug delivery systems. However, data standardization, regulatory compliance, and translational scalability challenges remain. This review underscores the need for interdisciplinary collaboration to unlock AI's potential in nanomedicine fully, ultimately advancing the clinical application of SMNs for more effective and safer patient care.
Collapse
Affiliation(s)
- Hamid Noury
- Health Research Center, Chamran Hospital, Tehran, Iran
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol 538-98615, Iran.
| | | | - Zahra Jamalpoor
- Trauma and Surgery Research Center, Aja University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Naradun N, Chueakwon P, Kamkaew A, Hsu KY, Lee MN, Chansaenpak K, Wang YS, Lai RY. Ferritin Fused with MiniSOG for In Vitro Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2025; 8:3909-3919. [PMID: 40260679 PMCID: PMC12093366 DOI: 10.1021/acsabm.5c00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
To avoid off-target effects, targeted cancer therapy offers a feasible alternative to traditional cancer therapies such as chemotherapy and radiotherapy. The human ferritin receptor (transferrin receptor, TfR1) is greatly overexpressed in several cancer types, including liver cancer. Therefore, human ferritin (HFn) has been used in drug encapsulation for targeted therapy. However, the drug encapsulation method is time-consuming and not applicable to all conditions. In this study, we effectively designed HFn fused with a photosensitizing protein called mini-singlet oxygen generator (miniSOG) to create HFn-miniSOG for targeted photodynamic therapy (PDT) applications. The fusion protein HFn-miniSOG self-assembled to form nanoparticles with an average size of 22.4 ± 1.3 nm and generated singlet oxygen (1O2) when activated by blue-light irradiation with ΦΔ = 0.30. To demonstrate its targeted PDT capability, phototoxicity was assessed in HepG2 and HeLa cells with varying TfR1 expression levels. The viability of HepG2 cells was reduced by 63% after light irradiation, compared to 34% in HeLa cells, because HepG2 cells exhibit greater levels of TfR1. As a result, our study provides a straightforward approach for creating ALL-IN-ONE protein nanoparticles for targeted PDT applications.
Collapse
Affiliation(s)
- Narathip Naradun
- School
of Chemistry, Institute of Science, Suranaree
University of Technology, Nakhon
Ratchasima 30000, Thailand
| | - Piyasiri Chueakwon
- School
of Chemistry, Institute of Science, Suranaree
University of Technology, Nakhon
Ratchasima 30000, Thailand
| | - Anyanee Kamkaew
- School
of Chemistry, Institute of Science, Suranaree
University of Technology, Nakhon
Ratchasima 30000, Thailand
| | - Kai-Yu Hsu
- Institute
of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Man Nee Lee
- Institute
of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Kantapat Chansaenpak
- National
Nanotechnology Center, National Science
and Technology Development Agency, Thailand Science Park, Pathum Thani 12120, Thailand
| | - Yane-Shih Wang
- Institute
of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
| | - Rung-Yi Lai
- School
of Chemistry, Institute of Science, Suranaree
University of Technology, Nakhon
Ratchasima 30000, Thailand
| |
Collapse
|
4
|
de Moura IA, Silva AJD, de Macêdo LS, de Melo KMTB, Leal LRS, Espinoza BCF, Invenção MDCV, de Pinho SS, de Freitas AC. Advances in the Functionalization of Vaccine Delivery Systems: Innovative Strategies and Translational Perspectives. Pharmaceutics 2025; 17:640. [PMID: 40430931 PMCID: PMC12115142 DOI: 10.3390/pharmaceutics17050640] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The development of effective vaccines requires a rational design that considers the interaction between antigens, their vectors, and the immune system in addition to the activation of pathways that induce a safe and specific immune response. The efficacy of a vaccine formulation depends on the nature of the antigen, the protection offered by the delivery system, the ability to potentiate the immune response, and the precise release of the immunogen. Carrier systems such as lipid nanoparticles, polymers, exosomes, and microorganisms can be functionalized by chemical, physical, or biological methods to generate selective and improved biodistribution profiles. These methods enhance interaction with target cells, thereby improving immunological efficacy. The conjugation of specific ligands or the modification of parameters such as shape, charge, and size of vectors can enhance the specificity, stability, and efficiency of antigen transport to cellular compartments, thereby facilitating a robust immune response. This study examines modifications in vaccine delivery systems, focusing on biomolecules and physicochemical changes that enhance antigen presentation. Additionally, we examine innovative methods, including microneedles, electroporation, and needle-free systems that show potential for enhancing the immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Avenida da Engenharia S/N, Recife 50740-600, Pernambuco, Brazil; (I.A.d.M.); (A.J.D.S.); (L.S.d.M.); (K.M.T.B.d.M.); (L.R.S.L.); (B.C.F.E.); (M.d.C.V.I.); (S.S.d.P.)
| |
Collapse
|
5
|
Jayalakshmi CS, Haider M, Sanpui P. Tyrosine kinase inhibitor-loaded zein nanoparticles with enhanced drug delivery and anticancer efficacy. Int J Biol Macromol 2025; 314:143898. [PMID: 40348249 DOI: 10.1016/j.ijbiomac.2025.143898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/19/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
Tyrosine kinase inhibitors (TKIs) including erlotinib (Er) and gefitinib (Gef) can restrict the uncontrolled proliferation of cancer cells by inhibiting tyrosine kinases. However, their clinical application is limited by poor solubility and bioavailability; necessitating high doses that cause toxicity. Herein, we report the preparation of gum arabic-stabilized zein nanoparticles (GA-ZNPs) for delivering Er and Gef. The formulations exhibited particle sizes of 136.34 nm for Er-GA-ZNPs and 163.65 nm for Gef-GA-ZNPs with 86.4 % and 82.9 % encapsulation efficiencies, respectively. The drug-loaded GA-ZNPs demonstrated sustained release, with cumulative drug release reaching 75.8 % for Er and 65.7 % for Gef over 72 h. Uptake studies revealed a 2.45-2.7-fold increase in intracellular drug accumulation for GA-ZNPs formulations compared to free drugs. GA-ZNPs formulations significantly enhanced the cytotoxicity of the drugs on cancer cells, reducing IC₅₀ of free drugs by 1.58-2.35-fold for Er and 1.8-1.9-fold for Gef. Also, Er-GA-ZNPs and Gef-GA-ZNPs induced 1.5-2.4-fold and 1.2-2.7-fold higher apoptosis than free drugs in tested cancer cells, respectively. Mechanistic studies revealed higher ROS generation and mitochondrial membrane depolarization in cells treated with GA-ZNPs formulations. These findings suggest that GA-ZNPs-based formulations significantly improve the anticancer efficacy of TKIs, offering a promising approach for targeted cancer therapy.
Collapse
Affiliation(s)
- C S Jayalakshmi
- Department of Biotechnology, Birla Institute of Technology and Science, Pilani, Dubai Campus, Dubai International Academic City, Dubai, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed Haider
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| | - Pallab Sanpui
- Department of Biotechnology, Birla Institute of Technology and Science, Pilani, Dubai Campus, Dubai International Academic City, Dubai, United Arab Emirates.
| |
Collapse
|
6
|
Serra A, Fratello M, Federico A, Greco D. An update on knowledge graphs and their current and potential applications in drug discovery. Expert Opin Drug Discov 2025; 20:599-619. [PMID: 40223439 DOI: 10.1080/17460441.2025.2490253] [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: 09/19/2024] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION Knowledge graphs are becoming prominent tools in computational drug discovery. They effectively integrate heterogeneous biomedical data and generate new hypotheses and knowledge. AREAS COVERED This article is based on a literature review using Google Scholar and PubMed to retrieve articles on existing knowledge graphs relevant to the drug discovery field. The authors compare the types of entities, relationships, and data sources they encompass. Additionally, the authors provide examples of their use in the drug discovery field and discuss potential strategies for advancing this research area. EXPERT OPINION Knowledge graphs are crucial in drug discovery, but their construction leads to challenges in data integration and consistency. Future research should prioritize the standardization of data sources and data modeling. More efforts are needed for the integration in knowledge graphs of diverse data types, such as chemical structures and epigenetic data, to enhance their effectiveness. Additionally, advancements in large language models should be pursued to aid the development of knowledge graphs, provide intuitive querying capabilities for non-expert users, and explain knowledge graphs -derived predictions, thereby making these tools more accessible and their insights more interpretable for a wider audience.
Collapse
Affiliation(s)
- Angela Serra
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Michele Fratello
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antonio Federico
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Tampere Institute for Advanced Study, Tampere University, Tampere, Finland
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Yu D, Liu M, Ding Q, Wu Y, Wang T, Song L, Li X, Qian K, Cheng Z, Gu M, Li Z. Molecular imaging-guided diagnosis and treatment integration for brain diseases. Biomaterials 2025; 316:123021. [PMID: 39705925 DOI: 10.1016/j.biomaterials.2024.123021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
In practical clinical scenarios, improved diagnostic methods have been developed for the precise visualization of molecular targets using molecular imaging in brain diseases. Recently, the introduction of innovative molecular imaging modalities across both macroscopic and mesoscopic dimensions, with remarkable specificity and spatial resolution, has expanded the scope of applications beyond diagnostic testing, with the potential to guide therapeutic interventions, offering real-time feedback in the context of brain therapy. The molecular imaging-guided integration of diagnosis and treatment holds the potential to revolutionize disease management by enabling the real-time monitoring of treatment responses and therapy adjustments. Given the vibrant and ever-evolving nature of this field, this review provides an integrated picture on molecular image-guided diagnosis and treatment integration for brain diseases involving the basic concepts, significant breakthroughs, and recent trends. In addition, based on the current achievements, some critical challenges are also discussed.
Collapse
Affiliation(s)
- Donghu Yu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Menghao Liu
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul, 02841, South Korea.
| | - Youxian Wu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tianqing Wang
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Litong Song
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaoyu Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kun Qian
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Meijia Gu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Zhiqiang Li
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Soans SH, Chonche MJ, Sharan K, Srinivasan A, Archer AC. Apoptotic and anti-inflammatory effect of nisin-loaded sodium alginate-gum arabic nanoparticles against colon cancer cells. Int J Biol Macromol 2025; 305:141747. [PMID: 40049503 PMCID: PMC7617643 DOI: 10.1016/j.ijbiomac.2025.141747] [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: 09/30/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Colon cancer is one of the leading causes of mortality and morbidity worldwide. Nisin, a polycyclic antibacterial peptide and food preservative has shown potential to combat cancer. However, it is susceptible to proteolytic cleavage in the gut. The current study investigates the protective and cytotoxic effects of nisin loaded sodium alginate gum arabic nanoparticles (Nis/ALG-GA NPs) in Caco2 cells. The physicochemical properties, loading efficiency and release kinetics were studied. Cytotoxicity (MTT assay), apoptotic effect (Ethidium bromide and acridine orange staining) and internalisation (FITC tagging) were evaluated. Gene expression of apoptotic markers and IL-10 were analysed by qPCR. The Nis/ALG-GA NPs were spherical, small with a smooth outer surface and mean size of 193 ± 4 nm. The loading efficacy was 88 ± 2 % exhibiting slow sustained release of the peptide under different gut pH conditions. The IC50 value obtained was 500 μg for 48 h and 80 μg for 72 h of incubation. The Nis/ALG-GA NPs were internalised into Caco2 cells and induced apoptosis with an increased expression of bax gene and converse decrease of bcl-2 gene. Anti-inflammatory gene IL10 was upregulated upon treatment with NPs. Thus, the Nis/ALG-GA NPs may be promising oral drug delivery systems against colon cancers.
Collapse
Affiliation(s)
- Sanya Hazel Soans
- Department of Microbiology, JSS Academy of Higher Education and Research, Mysuru, India
| | - Muzaffar Jahangir Chonche
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Kunal Sharan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Asha Srinivasan
- Division of Nanoscience and Technology, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, India
| | - Ann Catherine Archer
- Department of Microbiology, JSS Academy of Higher Education and Research, Mysuru, India.
| |
Collapse
|
9
|
Uti DE, Atangwho IJ, Alum EU, Ntaobeten E, Obeten UN, Bawa I, Agada SA, Ukam CIO, Egbung GE. Antioxidants in cancer therapy mitigating lipid peroxidation without compromising treatment through nanotechnology. DISCOVER NANO 2025; 20:70. [PMID: 40272665 PMCID: PMC12021792 DOI: 10.1186/s11671-025-04248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/27/2025]
Abstract
BACKGROUND Cancer treatments often exploit oxidative stress to selectively kill tumour cells by disrupting their lipid peroxidation membranes and inhibiting antioxidant enzymes. However, lipid peroxidation plays a dual role in cancer progression, acting as both a tumour promoter and a suppressor. Balancing oxidative stress through antioxidant therapy remains a challenge, as excessive antioxidant activity may compromise the efficacy of chemotherapy and radiotherapy. AIM This review explores the role of antioxidants in mitigating lipid peroxidation in cancer therapy while maintaining treatment efficacy. It highlights recent advancements in nanotechnology-based targeted antioxidant delivery to optimize therapeutic outcomes. METHODS A comprehensive literature review was conducted using reputable databases, including PubMed, Scopus, Web of Science, and ScienceDirect. The search focused on publications from the past five years (2020-2025), supplemented by relevant studies from earlier years. Keywords such as "antioxidants," "lipid peroxidation," "nanotechnology in cancer therapy," and "oxidative stress" were utilized. Relevant articles were critically analysed, and graphical illustrations were created. RESULTS Emerging evidence suggests that nanoparticles, including liposomes, polymeric nanoparticles, metal-organic frameworks, and others, can effectively encapsulate and control the release of antioxidants in tumour cells while minimizing systemic toxicity. Stimuli-responsive carriers with tumour-specific targeting mechanisms further enhance antioxidant delivery. Studies indicate that these strategies help preserve normal cells, mitigate oxidative stress-related damage, and improve treatment efficacy. However, challenges such as bioavailability, stability, and potential interactions with standard therapies remain. CONCLUSION Integrating nanotechnology with antioxidant-based interventions presents a promising approach for optimizing cancer therapy. Future research should focus on refining lipid peroxidation modulation strategies, assessing oxidative stress profiles during treatment, and employing biomarkers to determine optimal antioxidant dosing. A balanced approach to antioxidant use may enhance therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
- Daniel Ejim Uti
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria.
| | - Item Justin Atangwho
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Esther Ugo Alum
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda
| | - Emmanuella Ntaobeten
- Department of Cancer and Haematology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Uket Nta Obeten
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| | - Inalegwu Bawa
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | - Samuel A Agada
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | | | - Godwin Eneji Egbung
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
10
|
Yang YP, Nicol CJB, Chiang MC. A Review of the Neuroprotective Properties of Exosomes Derived from Stem Cells and Exosome-Coated Nanoparticles for Treating Neurodegenerative Diseases and Stroke. Int J Mol Sci 2025; 26:3915. [PMID: 40332773 PMCID: PMC12028030 DOI: 10.3390/ijms26083915] [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: 02/18/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Neurological diseases, including neurodegenerative disorders and stroke, represent significant medical challenges due to their complexity and the limitations of current treatment approaches. This review explores the potential of stem cell (SC)-derived exosomes (Exos) as a transformative therapeutic strategy for these diseases. Exos, especially those derived from SCs, exhibit natural targeting ability, biocompatibility, and the capacity to cross the blood-brain barrier (BBB), making them ideal vehicles for drug delivery. This review provides an in-depth discussion of the properties and advantages of SC-Exos. It highlights their potential synergistic benefits in therapeutic approaches to treat neurological diseases. This article discusses the mechanisms of action of SC-Exos, highlighting their ability to target specific cells, modulate disease pathways, and provide controlled release of therapeutic agents. Applications in specific neurological disorders have been investigated, demonstrating the potential to improve outcomes in conditions such as Alzheimer's Disease (AD), Parkinson's Disease (PD), and stroke. Moreover, Exos-coated nanoparticles (NPs) combine the natural properties of Exos with the multifunctionality of NPs. This integration takes advantage of exosome membrane biocompatibility and targeting capabilities while preserving NPs' beneficial features, such as drug loading and controlled release. As a result, Exos-coated NPs may enhance the precision, efficacy, and safety of therapeutic interventions. In conclusion, SC-Exos represent a promising and innovative approach to treating neurological diseases.
Collapse
Affiliation(s)
- Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Christopher J. B. Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, and Cancer Biology and Genetics Division, Sinclair Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
11
|
Zhra M, Akhund SA, Mohammad KS. Advancements in Osteosarcoma Therapy: Overcoming Chemotherapy Resistance and Exploring Novel Pharmacological Strategies. Pharmaceuticals (Basel) 2025; 18:520. [PMID: 40283955 PMCID: PMC12030420 DOI: 10.3390/ph18040520] [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: 02/28/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
Osteosarcoma is recognized as the most prevalent primary bone malignancy, primarily affecting children and adolescents. It is characterized by its aggressive behavior and high metastatic potential, which often leads to poor patient outcomes. Despite advancements in surgical techniques and chemotherapy regimens, the prognosis for patients with osteosarcoma remains unsatisfactory, with survival rates plateauing over the past few decades. A significant barrier to effective treatment is the development of chemotherapy resistance, which complicates the management of the disease and contributes to high rates of recurrence. This review article aims to provide a comprehensive overview of recent advancements in osteosarcoma therapy, particularly in overcoming chemotherapy resistance. We begin by discussing the current standard treatment modalities, including surgical resection and conventional chemotherapy agents such as methotrexate, doxorubicin, and cisplatin. While these approaches have been foundational in managing osteosarcoma, they are often limited by adverse effects and variability in efficacy among patients. To address these challenges, we explore novel pharmacological strategies that aim to enhance treatment outcomes. This includes targeted therapies focusing on specific molecular alterations in osteosarcoma cells and immunotherapeutic approaches designed to harness the body's immune system against tumors. Additionally, we review innovative drug delivery systems that aim to improve the bioavailability and efficacy of existing treatments while minimizing toxicity. The review also assesses the mechanisms underlying chemotherapy resistance, such as drug efflux mechanisms, altered metabolism, and enhanced DNA repair pathways. By synthesizing current research findings, we aim to highlight the potential of new therapeutic agents and strategies for overcoming these resistance mechanisms. Ultimately, this article seeks to inform future research directions and clinical practices, underscoring the need for continued innovation in treating osteosarcoma to improve patient outcomes and survival rates.
Collapse
Affiliation(s)
| | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.Z.); (S.A.A.)
| |
Collapse
|
12
|
George M, Boukherroub R, Sanyal A, Szunerits S. Treatment of lung diseases via nanoparticles and nanorobots: Are these viable alternatives to overcome current treatments? Mater Today Bio 2025; 31:101616. [PMID: 40124344 PMCID: PMC11930446 DOI: 10.1016/j.mtbio.2025.101616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Challenges Respiratory diseases remain challenging to treat, with current efforts primarily focused on managing symptoms rather than maintaining overall lung health. Traditional treatment methods, such as oral or parenteral administration of antiviral, antibacterial, and anti-inflammatory drugs, face limitations. These include difficulty in delivering therapeutic agents to pathogens residing deep in the airways and the risk of severe side effects due to high systemic drug concentrations. The growing threat of drug-resistant pathogens further complicates infection management. Advancements The lung's large surface area offers an attractive target for inhalation-based drug delivery. Nanoparticles (NP) enable uniform and sustained drug distribution across the alveolar network, overcoming challenges posed by complex lung anatomy. Recent breakthroughs in nanorobots (NR) have demonstrated precise navigation through biological environments, delivering therapies directly to affected lung areas with enhanced accuracy. Nanotechnology has also shown promise in treating lung cancer, with nanoparticles engineered to overcome biological barriers, improve drug solubility, and enable controlled drug release. Future scope This review explores the progress of NP and NR in addressing challenges in pulmonary drug delivery. These innovations allow targeted delivery of nucleic acids, drugs, or peptides to the pulmonary epithelium with unprecedented accuracy, offering significant potential for improving therapeutic effectiveness in respiratory disorders.
Collapse
Affiliation(s)
- Meekha George
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Bebek, 34342, Istanbul, Turkey
| | - Sabine Szunerits
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| |
Collapse
|
13
|
Mehta J, Kumar P, Pawar SV. Exploration of capsaicin-encapsulated lignin nanoparticles for alleviating non-alcoholic fatty liver disease: In-vitro study. Int J Biol Macromol 2025; 303:140616. [PMID: 39909244 DOI: 10.1016/j.ijbiomac.2025.140616] [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: 10/10/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a rising public health concern with limited effective therapeutic options, making it a significant risk factor for end-stage liver disease and cirrhosis, globally. This increasing prevalence of NAFLD underscores the development of innovative therapeutic approaches to confront the increasing prevalence of NAFLD. This research explores the potential of capsaicin-encapsulated lignin nanoparticles (Cap-LNPs) as a novel targeted therapeutic approach. Capsaicin, a bioactive compound has demonstrated anti-oxidant, anti-inflammatory, anti-steatotic, and anti-fibrotic properties that have a protective role against NAFLD. Lignin, recognized for its non-toxic, eco-friendly, multifunctional, and biodegradable attributes, has garnered significant attention as a versatile material for drug delivery systems. Incorporating these two natural compounds into nanoparticles offers a promising approach to enhance capsaicin's bioavailability, stability, and targeted delivery to hepatic cells. The Cap-LNPs were synthesized using the nanoprecipitation technique and characterized by a mean diameter of 200.2 ± 5.79 nm, polydispersity index (PDI) of 0.137 ± 0.0459 with spherical morphology, encapsulation efficiency of 96.85 ± 0.73 %, and drug loading capacity of 16.14 ± 0.12 %. In-vitro studies demonstrated that Cap-LNPs substantially reduced intracellular accumulation of triglyceride compared with free capsaicin and control groups, confirmed by Oil Red O staining and triglycerides (TG) quantification.
Collapse
Affiliation(s)
- Jhalak Mehta
- Pharmaceutical Biotechnology Research Lab, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Pankaj Kumar
- Pharmaceutical Biotechnology Research Lab, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Sandip V Pawar
- Pharmaceutical Biotechnology Research Lab, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
14
|
Sitarek P, Owczarek M, Kowalczyk T, Kukula-Koch W, Lasoń-Rydel M, Herczyńska L. Dual Biopolymer Layer Using Nanoparticles with Active Substance Enclosed in Microcapsules: Innovative Solution for Slow Release of Ginkgo biloba L. Extract for Potential Therapies. Int J Mol Sci 2025; 26:3066. [PMID: 40243724 PMCID: PMC11988488 DOI: 10.3390/ijms26073066] [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: 02/19/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The dynamic development of various branches of medicine and pharmacy, along with the emergence of new preventive and alternative therapies for various diseases, creates opportunities for new solutions utilizing carriers of active substances. Their therapeutic effect may occur through direct contact with skin lesions or indirectly, where medicinal substances penetrate the capillary network in the deeper layers of the skin and reach the bloodstream. The aim of the research was to obtain carriers with a matrix consisting of two renewable-source polymers (chitosan and ethylcellulose) and a core material derived from Ginkgo biloba green leaf extract (GBE). The obtained ethylcellulose microcapsules with encapsulated chitosan nanoparticles with extract {Et[Ch(GB)NP]} were characterized with respect to size, shape, surface morphology (SEM microscopy), and active substance release kinetics (UV-VIS and mathematical release models). The kinetics of active substance release were analyzed using UV-VIS spectroscopy and mathematical release models. The released active components were assessed microbiologically for activity against six bacterial strains and two fungal strains, as well as chromatographically using HPLC-ESI-QTOF-MS/MS fingerprinting. The microcapsules with a dual polymer layer exhibited a slow release of the core material, which demonstrated microbiological activity. The strongest antimicrobial effects were observed against Klebsiella pneumoniae and Salmonella enteritidis, with a minimum inhibitory concentration (MIC) of 410 µg/mL. The release of the core material from the double-layer polymer structures was more efficient in a physiological saline environment, with the best fit for the extract release kinetics following a zero-order model (regression coefficient R2 = 0.9939). The obtained microcapsules with a dual polymer layer show great potential for therapeutic applications in the medical industry. Their controlled release properties and antibacterial effectiveness make them a promising carrier for active substances in modern therapies.
Collapse
Affiliation(s)
- Przemysław Sitarek
- Department of Medical Biology, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Monika Owczarek
- Łukasiewicz Research Network—Lodz Institute of Technology, Skłodowskiej-Curie 19/27, 90-570 Lodz, Poland; (M.O.); (M.L.-R.)
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland;
| | - Magdalena Lasoń-Rydel
- Łukasiewicz Research Network—Lodz Institute of Technology, Skłodowskiej-Curie 19/27, 90-570 Lodz, Poland; (M.O.); (M.L.-R.)
| | - Lucyna Herczyńska
- Textile Institute, Faculty of Material Technologies and Textile Design, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland;
| |
Collapse
|
15
|
Altuwaijri N, Fitaihi R, Alkathiri FA, Bukhari SI, Altalal AM, Alsalhi A, Alsulaiman L, Alomran AO, Aldosari NS, Alqhafi SA, Alhamdan M, Alfaraj R. Assessing the Antibacterial Potential and Biofilm Inhibition Capability of Atorvastatin-Loaded Nanostructured Lipid Carriers via Crystal Violet Assay. Pharmaceuticals (Basel) 2025; 18:417. [PMID: 40143193 PMCID: PMC11944405 DOI: 10.3390/ph18030417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Atorvastatin (ATR), an antihyperlipidemic drug with a potential antibacterial effect, was investigated in this study. Like other statins, ATR has been repurposed for several uses, ranging from anti-inflammatory to antimicrobial applications, and has demonstrated successful results. However, the efficacy of ATR is limited by its low solubility, indicating an opportunity for its encapsulation in a nanotechnology-based drug delivery system. Methods: Nanostructured lipid carrier (NLC) formulations were prepared using high-pressure homogenization and ultrasonication. The formulations were characterized, including their particle size, polydispersity index, zeta potential, encapsulation efficiency, and in vitro release. Antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA), Escherichia coli (E. coli), and Staphylococcus aureus (S. aureus) was evaluated using the growth curve (bacterial growth over time) and well diffusion methods (zone of inhibition and minimum inhibitory concentration (MIC) determination). The crystal violet assay was employed to assess biofilm inhibition. Results: The NLC formulations were optimized, and the size and zeta potential of the blank nanoparticles were 130 ± 8.39 nm and -35 ± 0.5 mV, respectively. In comparison, the encapsulated NLCs had a size of 142 ± 52.20 nm and a zeta potential of -31 ± 1.41 mV. The average encapsulation efficiency was 94%, and 70% of the drug was released after 24 h. The ATR-loaded NLCs showed significantly enhanced antibacterial activity by reducing the minimum inhibitory concentration by 2.5-fold for E. coli, 1.8-fold for S. aureus, and 1.4-fold for MRSA, and promoting more effective bacterial growth inhibition. Notably, biofilm inhibition was significantly improved with ATR-NLCs, achieving 80% inhibition for S. aureus, 40% for E. coli, and 30% for MRSA, compared to free ATR (p < 0.001). These findings suggest that NLC encapsulation enhances ATR's antimicrobial efficacy and biofilm suppression. Conclusions: This study identified NLCs as successful carriers of ATR, significantly enhancing its antibacterial efficacy and biofilm inhibition capabilities. This formulation, which shows antimicrobial potential against both Gram-positive and Gram-negative bacteria, should be further studied and developed against different resistant microbial strains.
Collapse
Affiliation(s)
- Njoud Altuwaijri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Rawan Fitaihi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Fai A. Alkathiri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Sarah I. Bukhari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Alanoud M. Altalal
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Alyaa Alsalhi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Lama Alsulaiman
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Aljawhara O. Alomran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Noura S. Aldosari
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Safa A. Alqhafi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Majd Alhamdan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Rihaf Alfaraj
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| |
Collapse
|
16
|
Mohamed HRH, Elberry YA, Magdy H, Ismail M, Michael M, Eltayeb N, Safwat G. Erbium oxide nanoparticles induce potent cell death, genomic instability and ROS-mitochondrial dysfunction-mediated apoptosis in U937 lymphoma cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03962-x. [PMID: 40072553 DOI: 10.1007/s00210-025-03962-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
Erbium oxide nanoparticles (Er2O3-NPs) have attracted significant attention for their unique physicochemical properties, including high surface area, biocompatibility, and stability. However, the impact of Er2O3-NPs on lymphoma cells (LCs) has not been explored, making this an innovative avenue for exploration. Therefore, the current study aimed to explore the influence of Er2O3-NPs on cell viability, genomic and mitochondrial DNA integrity, reactive oxygen species (ROS) generation and apoptosis induction in human U937 LCs. Er2O3-NPs were characterized using X-ray diffraction (XRD) and transmission electron microscopy (TEM). The effect of Er2O3-NPs on cell viability and genomic DNA integrity was estimated after 48 h using the WST-1 cytotoxicity and alkaline Comet assays, respectively. The generation level of reactive oxygen species (ROS) and mitochondrial membrane potential were also analyzed. Flow Cytometry was used to assess apoptosis induction and quantitative RT-PCR was conducted to measure the apoptotic (p53), anti-apoptotic (Bcl2), and mitochondrial (ND3) gene expression. Our results demonstrated the purity and well distribution of Er2O3-NPs and revealed that Er2O3-NPs induce strong cytotoxicity on U937 cells, as evidenced by a concentration-dependent reduction in cell viability with an IC50 value of 3.20 µg/ml. Exposure of U937 LCs to the IC50 concentration (3.20 µg/ml) of Er2O3-NPs promoted excessive ROS generation, leading to dramatic damage to genomic DNA and mitochondrial membrane potential, as well as marked dysregulation of apoptotic (p53), anti-apoptotic (Bcl2) and mitochondrial ND3 gene expression. This cascade of events triggered both apoptosis and necrosis in Er2O3-NPs-treated U937 LCs. In conclusion, these findings highlight the strong in vitro cytotoxic potential of Er2O3-NPs against highly aggressive U937 LCs, mediated by excessive ROS production, which leads to severe genomic DNA and mitochondrial membrane damage, as well as profound alterations in apoptotic, anti-apoptotic and mitochondrial gene expression. Future research is needed to further investigate the potential use of Er2O3-NPs in treating lymphoma and to optimize their therapeutic efficacy.
Collapse
Affiliation(s)
- Hanan R H Mohamed
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Yusuf Ahmed Elberry
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Hagar Magdy
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Maryam Ismail
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Maivel Michael
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Nourhan Eltayeb
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| |
Collapse
|
17
|
Prananda AT, Halim P, Syahputra RA. Targeting miRNA with flavonoids: unlocking novel pathways in cardiovascular disease management. Front Pharmacol 2025; 16:1532986. [PMID: 40115258 PMCID: PMC11922852 DOI: 10.3389/fphar.2025.1532986] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/10/2025] [Indexed: 03/23/2025] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality worldwide, with complex pathophysiological mechanisms such as oxidative stress, inflammation, apoptosis, and endothelial dysfunction driving disease progression. MicroRNAs (miRNAs), a class of non-coding RNAs, have emerged as key regulators of gene expression involved in these processes, positioning them as potential biomarkers and therapeutic targets in CVD management. Simultaneously, flavonoids, naturally occurring polyphenolic compounds found in various plant-based foods, have gained attention for their cardioprotective properties, including antioxidant, anti-inflammatory, and anti-apoptotic effects. Recent studies suggest a novel intersection between flavonoids and miRNAs, where flavonoids may modulate the expression of specific miRNAs implicated in CVD pathogenesis. This review explores the potential of flavonoids as miRNA modulators, focusing on their ability to regulate miRNAs associated with cardiac fibrosis, hypertrophy, and vascular inflammation. By bridging the therapeutic potential of flavonoids with miRNA targeting, this review highlights innovative pathways for advancing CVD treatment strategies. Additionally, preclinical and clinical evidence supporting these interactions is discussed, alongside the challenges and opportunities in developing flavonoid-based miRNA therapies. Unlocking this synergy could pave the way for more effective, personalized approaches to CVD management, addressing unmet needs in contemporary cardiovascular care.
Collapse
Affiliation(s)
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
18
|
Patne AY, Mohapatra S, Mohapatra SS. Role of Nanomedicine in Transforming Pharmacotherapy for Substance Use Disorder (SUD). WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70008. [PMID: 40190158 PMCID: PMC11973540 DOI: 10.1002/wnan.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
The field of nanomedicine offers revolutionary potential to reshape the discovery and development of therapeutics for diverse human diseases. However, its application has been limited in improving Substance Use Disorders (SUDs), which represent a profound public health crisis, including major types such as opioid, alcohol, stimulant, and cannabis use disorders. Pharmacotherapy, a cornerstone of SUD management, has reduced morbidity, mortality, and the societal impact of addiction, though its efficacy has ranged from none to moderate. Thus, there is a major unmet need to transform SUD pharmacotherapy to curb the epidemic of addiction. This article explores the potential roles of nanomedicine-inspired precision-targeted drug delivery, sustained release, and combination therapies to increase therapeutic efficacy and minimize side effects. Additionally, it discusses innovative mechanisms that align with the neurobiological complexities of addiction and synergistic approaches that integrate nanomedicine with behavioral interventions, device-based therapies, and emerging modalities such as immunotherapy and neurostimulation. Despite these advancements, barriers such as treatment accessibility, adherence challenges, and inequitable resource distribution persist, particularly in underserved populations. By harnessing the transformative capabilities of nanomedicine and integrating it into holistic, equitable, and personalized care frameworks, this review highlights a path forward to revolutionize the SUD pharmacotherapy landscape. The article underscores the need for continued nano-SUD pharmacotherapy research and the development of strategies to alleviate the substantial burden of addiction on individuals, families, and society.
Collapse
Affiliation(s)
- Akshata Y. Patne
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
- Graduate Programs, Taneja College of Pharmacy, MDC30, 12908 USF Health DriveTampaFloridaUSA
| | - Subhra Mohapatra
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
- Department of Molecular Medicine, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
- Research ServiceJames A. Haley Veterans HospitalTampaFloridaUSA
| | - Shyam S. Mohapatra
- Center for Research and Education in Nanobioengineering, Department of Internal Medicine, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
- Graduate Programs, Taneja College of Pharmacy, MDC30, 12908 USF Health DriveTampaFloridaUSA
- Research ServiceJames A. Haley Veterans HospitalTampaFloridaUSA
| |
Collapse
|
19
|
Subramaniyan V, Hanim YU. Role of pancreatic lipase inhibition in obesity treatment: mechanisms and challenges towards current insights and future directions. Int J Obes (Lond) 2025; 49:492-506. [PMID: 40016558 PMCID: PMC11971044 DOI: 10.1038/s41366-025-01729-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/16/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025]
Abstract
The worldwide health emergency of obesity is closely connected to how dietary fats are metabolized, whereas the process is significantly influenced by pancreatic lipase (PL), an enzyme critical for lipid hydrolysis into fatty acids. This narrative review employs a methodological approach utilizing literature searches of PubMed data up to March 2024. The search term criteria encompasses keywords related to the role, mechanism, challenges, and current and future treatments of pancreatic lipase in obesity with an overall references is 106. This paper offers a comprehensive explanation of the role of PL, underlining its significance in the digestive process and lipid imbalances that contribute to obesity and by extension, its impact on obesity development and progression. Additionally, it delves into the dual functionality of the pancreas, emphasizing its impact on metabolism and energy utilization which, when dysregulated, promotes obesity. A focal point of this review is the investigation into the efficacy, challenges, and adverse effects of current pancreatic lipase inhibitors, with orlistat being highlighted as a primary current drug delivery. By discussing advanced obesity treatments, including the exploration of novel anti-obesity medications that target specific biological pathways, this review underscores the complexity of obesity treatment and the necessity for a multifaceted approach. In conclusion, this paper emphasizing the importance of understanding the role of enzymes like pancreatic lipase mechanistic and adopting a multidisciplinary approach to treatment and side effects of current obesity drugs and explore new emerging therapeutic strategies for more effective obesity management.
Collapse
Affiliation(s)
- Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
- Division of Pharmacology, School of Medical and Life Sciences, Sunway University, Sunway, 47500, Malaysia.
| | - Yusoff Umul Hanim
- Division of Pharmacology, School of Medical and Life Sciences, Sunway University, Sunway, 47500, Malaysia
| |
Collapse
|
20
|
Nyandoro VO, Ismail EA, Tageldin A, Gafar MA, Peters XQ, Mautsoe R, Omolo CA, Govender T. Potential of nanocarrier-mediated delivery of vancomycin for MRSA infections. Expert Opin Drug Deliv 2025; 22:347-365. [PMID: 39949087 DOI: 10.1080/17425247.2025.2459756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/24/2025] [Indexed: 02/20/2025]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) threatens global health due to its resistance to vancomycin, which is the standard treatment despite limitations, including nephrotoxicity and low intracellular permeability. This necessitates the development of innovative strategies such as nanocarrier-mediated delivery to overcome such limitations. Nanocarriers serve as delivery systems for vancomycin and exhibit inherent antibacterial properties, potentially providing synergism and overcoming MRSA's resistance. Nanocarriers provide sustained release and targeted delivery of vancomycin to the infection site, achieving higher therapeutic concentrations and superior antibacterial activity with reduced doses, which minimizes systemic toxicity. Moreover, leveraging simulations techniques provides more insights on vancomycin-nanocarrier interactions, facilitating the optimization of nanosystems. AREAS COVERED The article discusses the potential of nanocarriers in delivering vancomycin to infection site, reducing systemic toxicity, and potentiating anti-MRSA activity. Additionally, it reviews modeling and simulation studies to provide a deeper understanding of vancomycin-nanocarrier interactions. The literature search included experimental articles from 2017 to 2024, searched in Web of Science, Google scholar, PubMed, and Scopus. EXPERT OPINION Nanocarrier-mediated delivery of vancomycin offers promising approaches to combat MRSA infections by enhancing therapeutic efficacy and reducing systemic toxicity. However, further research is required to optimize these nanoformulations and advance them to clinical trials and practical applications.
Collapse
Affiliation(s)
- Vincent O Nyandoro
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Pharmaceutics and Pharmaceutical Chemistry, School of Pharmacy, Kabarak University, Kabarak, Kenya
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Medani, Sudan
| | - Abdelrahman Tageldin
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Mohammed A Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Xylia Q Peters
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Relebohile Mautsoe
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- School of Pharmacy and Health Sciences, Department of Pharmaceutics, United States International University-Africa, Nairobi, Kenya
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
21
|
MOMEN-MESGIN RAZIEH, REZAIE JAFAR, NEJATI VAHID, MOGHADAM PEYMANNAJAFI. Magnetic melamine cross-linked polystyrene-alt-malic anhydride copolymer: Synthesis, characterization, paclitaxel delivery, cytotoxic effects on human ovarian and breast cancer cells. Oncol Res 2025; 33:665-674. [PMID: 40109872 PMCID: PMC11915072 DOI: 10.32604/or.2024.054487] [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: 05/29/2024] [Accepted: 09/13/2024] [Indexed: 03/22/2025] Open
Abstract
Objectives Due to systematic side effects, there is a growing interest in nanoparticle formulation of anticancer drugs. Here, we aimed to synthesize poly (styrene-alt-maleic anhydride) cross-linked by melamine (PSMA/Me) and coated with magnetite nanoparticles (MNPs) PSMA/Me/Fe3O4. In addition, we aimed to load paclitaxel (PTX) into PSMA/Me/Fe3O4 for drug delivery and anticancer investigations. Methods Novel PSMA/Me was synthesized via free radical copolymerization, coated with Fe3O4, and then used as a transporter for PTX delivery. Fabricated copolymer was characterized using SEM, TGA, and XRD techniques. Drug release rate and loading efficiency were investigated. Human ovarian cancer cells (Skov-3) and breast cancer cells (MCF-7 cells) were incubated with the serial concentration of either free PTX or PSMA/Me/Fe3O4/PTX for cell viability and IC50 analysis for 24 and 48 h. Results Characterization methods confirmed PSMA/Me copolymer formation. The results showed a significant encapsulation efficiency of 83%. The drug release analysis exhibited that PSMA/Me/Fe3O4/PTX may be considered pH-sensitive nanocarriers. PSMA/Me/Fe3O4/PTX reduced cell viability both dose and time-dependently (p < 0.05). IC50 values of PSMA/Me/Fe3O4/PTX were low when compared to free PTX either 24 or 48 h post-treatment. Conclusions Our results indicated that PSMA/Me/Fe3O4/PTX was more cytotoxic than PTX in both cancer cells. Findings indicated the potential of PSMA/Me/Fe3O4/PTX as an anticancer nanocarrier system.
Collapse
Affiliation(s)
| | - JAFAR REZAIE
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, 5714783734, Iran
| | - VAHID NEJATI
- Department of Biology, Urmia University, Urmia, 5756151818, Iran
| | | |
Collapse
|
22
|
Moni SS, Moshi JM, Matou-Nasri S, Alotaibi S, Hawsawi YM, Elmobark ME, Hakami AMS, Jeraiby MA, Sulayli AA, Moafa HN. Advances in Materials Science for Precision Melanoma Therapy: Nanotechnology-Enhanced Drug Delivery Systems. Pharmaceutics 2025; 17:296. [PMID: 40142960 PMCID: PMC11945159 DOI: 10.3390/pharmaceutics17030296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Melanoma, a highly aggressive form of skin cancer, poses a major therapeutic challenge due to its metastatic potential, resistance to conventional therapies, and the complexity of the tumor microenvironment (TME). Materials science and nanotechnology advances have led to using nanocarriers such as liposomes, dendrimers, polymeric nanoparticles, and metallic nanoparticles as transformative solutions for precision melanoma therapy. This review summarizes findings from Web of Science, PubMed, EMBASE, Scopus, and Google Scholar and highlights the role of nanotechnology in overcoming melanoma treatment barriers. Nanoparticles facilitate passive and active targeting through mechanisms such as the enhanced permeability and retention (EPR) effect and functionalization with tumor-specific ligands, thereby improving the accuracy of drug delivery and reducing systemic toxicity. Stimuli-responsive systems and multi-stage targeting further improve therapeutic precision and overcome challenges such as poor tumor penetration and drug resistance. Emerging therapeutic platforms combine diagnostic imaging with therapeutic delivery, paving the way for personalized medicine. However, there are still issues with scalability, biocompatibility, and regulatory compliance. This comprehensive review highlights the potential of integrating nanotechnology with advances in genetics and proteomics, scalable, and patient-specific therapies. These interdisciplinary innovations promise to redefine the treatment of melanoma and provide safer, more effective, and more accessible treatments. Continued research is essential to bridge the gap between evidence-based scientific advances and clinical applications.
Collapse
Affiliation(s)
- Sivakumar S. Moni
- College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Jobran M. Moshi
- Department of Medical Laboratory Technology, College of Nursing and Health Science, Jazan University, Jazan 45142, Saudi Arabia
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Sabine Matou-Nasri
- Blood and Cancer Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh 11481, Saudi Arabia;
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Shmoukh Alotaibi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah 23433, Saudi Arabia; (S.A.); (Y.M.H.)
| | - Yousef M. Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah 23433, Saudi Arabia; (S.A.); (Y.M.H.)
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | | | | | - Mohammed A. Jeraiby
- Department of Basic Medical Science, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Ahmed A. Sulayli
- Laboratory Department, Prince Mohammed bin Nasser Hospital, Jazan Health Cluster, Jazan 82734, Saudi Arabia;
| | - Hassan N. Moafa
- Department of Public Health, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Quality and Patients Safety, Jazan University Hospital, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
23
|
Harine A, Ranjani S, Hemalatha S. Antifungal efficacy of Citrusfusion mediated silver nanoparticles in Candida species. BMC Biotechnol 2025; 25:18. [PMID: 39979871 PMCID: PMC11841014 DOI: 10.1186/s12896-025-00952-y] [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: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Candida species are commensal fungi that can become opportunistic pathogens under specific host and environmental conditions. The emergence of multidrug-resistant Candida strains poses a significant challenge. Nanotechnology represents a cutting-edge field offering precise and targeted delivery systems for combating fungal infections, leveraging the unique properties of plant-derived bioactive compounds. This investigation employed a biogenic approach utilizing polyherbal leaf extracts from Citrus limon and Citrus medica, known for their abundant Citral content. RESULTS Citrus sp. extracts were used to synthesize Citrusfusion silver nanoparticles (CitAgNPs) through a green synthesis method. Characterization of CitAgNPs was carried out using advanced analytical methods ensuring the quality, uniformity, size, and charge. The synthesized CitAgNPs exhibited non toxic effect when tested on Vigna radiata and Danio rerio, highlighting their potential for sustainability and safe therapeutic use. Antifungal assays demonstrated the potent efficacy of CitAgNPs in various Candida strains, with low MIC and MFC. CitAgNPs exhibited remarkable biofilm inhibition capabilities and elucidated specific mechanisms of action in Candida species, surpassing the performance of fluconazole. CONCLUSION This study underscores the immense potential of nanotechnology-driven approaches harnessing Citrus leaf extract for synthesizing highly effective antifungal nanoparticles. The fusion of biogenic nanoparticles with Citrus bioactive compounds presents a sustainable strategy for addressing the escalating challenge of azole-resistant Candida infections. The research outcomes suggest that CitAgNPs have promising applications in inhibiting Candida biofilms, offering potential solutions for infections caused by diaper rashes and onychomycosis, providing safe and effective alternatives to antifungal therapies.
Collapse
Affiliation(s)
- A Harine
- School of Life Sciences, B. S. Abdur Rahman Crescent Institute of Science and Technology, Vandalur, Chennai, Tamil Nadu, 600048, India
| | - S Ranjani
- School of Life Sciences, B. S. Abdur Rahman Crescent Institute of Science and Technology, Vandalur, Chennai, Tamil Nadu, 600048, India
| | - S Hemalatha
- School of Life Sciences, B. S. Abdur Rahman Crescent Institute of Science and Technology, Vandalur, Chennai, Tamil Nadu, 600048, India.
| |
Collapse
|
24
|
Theivendren P, Kunjiappan S, Pavadai P, Ravi K, Murugavel A, Dayalan A, Santhana Krishna Kumar A. Revolutionizing Cancer Immunotherapy: Emerging Nanotechnology-Driven Drug Delivery Systems for Enhanced Therapeutic Efficacy. ACS MEASUREMENT SCIENCE AU 2025; 5:31-55. [PMID: 39991031 PMCID: PMC11843507 DOI: 10.1021/acsmeasuresciau.4c00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 02/25/2025]
Abstract
Cancer immunotherapy is an innovative way of treating cancer by stimulating individual cells to overcome cancer. Widespread biomedical studies were carried out with the aim of exploring immunotherapy cancer therapeutics, and this review spotlights some mechanisms in which it was developed, namely immune checkpoint inhibitors (E.G PD-1/PD-L1, CTLA-4), adoptive cell therapy (e.g., CAR T-cell therapy), and cancer vaccines. Although it has shown clinical benefit in a number of cancer types, including melanoma and non-small-cell lung cancer, several challenges have dampened enthusiasm for this approach, from the differing patient response rates to toxicities. Nanotechnology in drug delivery systems must play a role in overcoming the same. Nanotechnology enables increased specificity and controlled drug release, improved solubility and bioavailability, can treat the tumor specifically, and localized drug delivery at the disease site decreases systemic toxicity. The review also features advances in the construction of lipid-based, polymeric, and inorganic nanoparticles that improve drug stability and allow the delivery of cotherapeutic agents. Nanotechnology-based delivery systems can be used alone or in combination with immunotherapy to assist in improving the immune response, gaining access to the tumor microenvironment, and overcoming biological barriers. Thus, the nano-DDS were both safe and effective in preclinical studies, and ongoing clinical trials have shown that they are capable of increasing the therapeutic index of anticancer drugs. Lastly, the review also discusses current challenges and regulatory issues in advancing these technologies and highlights the importance of further research to devise appropriate methodology for efficient functionalization of nanotechnology for individualized cancer solutions in cancer treatment.
Collapse
Affiliation(s)
- Panneerselvam Theivendren
- Department
of Pharmaceutical Chemistry, Swamy Vivekanandha
College of Pharmacy, Elayampalayam 637205, Namakkal, Tamil Nadu, India
| | - Selvaraj Kunjiappan
- Department
of Biotechnology, Kalasalingam Academy of
Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Parasuraman Pavadai
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M. S. R. Nagar, Bengaluru 560054, Karnataka, India
| | - Kaveena Ravi
- Department
of Pharmaceutics, Swamy Vivekananda College
of Pharmacy, Elayampalayam 637205, Namakkal, Tamil Nadu, India
| | - Anusuya Murugavel
- Center
for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Avinash Dayalan
- Center
for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - A. Santhana Krishna Kumar
- Department
of Chemistry, National Sun Yat-sen University, No. 70, Lien-hai Road, Gushan District, Kaohsiung City 80424, Taiwan
- Department
of Chemistry, Saveetha School of Engineering, Saveetha Institute of
Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 602105, Tamil Nadu, India
| |
Collapse
|
25
|
Nsairat H, Lafi Z, Al-Najjar BO, Al-Samydai A, Saqallah FG, El-Tanani M, Oriquat GA, Sa’bi BM, Ibrahim AA, Dellinger AL, Alshaer W. How Advanced are Self-Assembled Nanomaterials for Targeted Drug Delivery? A Comprehensive Review of the Literature. Int J Nanomedicine 2025; 20:2133-2161. [PMID: 39990285 PMCID: PMC11847455 DOI: 10.2147/ijn.s490444] [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: 08/07/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
The development of effective drug delivery systems is a key focus in pharmaceutical research, aiming to enhance therapeutic efficacy while minimizing adverse effects. Self-assembled nanostructures present a promising solution due to their tunable properties, biocompatibility, and ability to encapsulate and deliver therapeutic agents to specific targets. This review examines recent advancements in drug-based self-assembled nanostructures for targeted delivery applications, including drug-drug conjugates, polymeric-based architectures, biomolecules, peptides, DNA, squalene conjugates and amphiphilic drugs. Various strategies for fabricating these nanostructures are discussed, with an emphasis on the design principles and mechanisms underlying their self-assembly and potential for targeted drug delivery to specific tissues or cells. Furthermore, the integration of targeting ligands, stimuli-responsive moieties and imaging agents into these nanostructures is explored for enhanced therapeutic outcomes and real-time monitoring. Challenges such as stability, scalability and regulatory hurdles in translating these nanostructures from bench to bedside are also addressed. Drug-based self-assembled nanostructures represent a promising platform for developing next-generation targeted drug delivery systems with improved therapeutic efficacy and reduced side effects.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Belal O Al-Najjar
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ali Al-Samydai
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Fadi G Saqallah
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Ghaleb Ali Oriquat
- Pharmacological and Diagnostic Research Center, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Bailasan Mohammad Sa’bi
- Pharmacological and Diagnostic Research Center, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Abed Alqader Ibrahim
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Anthony Lee Dellinger
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
26
|
Fung AKY, Chok KSH. Hepatic artery infusion chemotherapy: A resurgence. World J Gastrointest Oncol 2025; 17:99612. [PMID: 39958544 PMCID: PMC11755999 DOI: 10.4251/wjgo.v17.i2.99612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 01/18/2025] Open
Abstract
In this manuscript, we comment on the article by Zhou et al, who assessed the efficacy of hepatic arterial infusion chemotherapy (HAIC) and its combination strategies for advanced hepatocellular carcinoma (HCC) using network meta-analysis methodology. We focus specifically on the potential advantages and role of HAIC in the treatment algorithm for advanced HCC. However, there remains numerous knowledge gaps before the role of HAIC can be established. There is significant heterogeneity of HAIC regimes with difficult interpretation of the clinical outcomes. Additionally, there is a lack of direct comparative data between HAIC, systemic chemotherapy, novel immunotherapies and targeted therapies. The underlying biochemical mechanisms that might explain the efficacy of HAIC and its effect on the HCC microenvironment requires further research. In the developing era of nanotechnology and targeted drug delivery systems, there is potential for integration of HAIC with novel technologies to effectively treat advanced HCC whilst minimising systemic complications.
Collapse
Affiliation(s)
- Andrew Kai-Yip Fung
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China
| | - Kenneth Siu Ho Chok
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong 999077, China
| |
Collapse
|
27
|
Karunakar KK, Edwin ER, Gopalakrishnan M, Cheriyan BV, Ramaiyan V, Karthikha VS, Justin JP. Advances in nephroprotection: the therapeutic role of selenium, silver, and gold nanoparticles in renal health. Int Urol Nephrol 2025; 57:479-510. [PMID: 39312019 DOI: 10.1007/s11255-024-04212-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 01/29/2025]
Abstract
Renal toxicity is a disorder that causes considerable issues in healthcare systems world, highlighting the critical importance of creating alternative treatments. Metallic nanoparticles have recently emerged as promising therapeutic agents for nephroprotection because of their remarkable properties. Numerous disciplines, including medicine, biotechnology, and the food industry, are currently investigating and exploring metallic nanoparticles, such as selenium, silver, and gold, with promising outcomes. In this overview, we provide the most current findings on cutting-edge nephroprotection through metallic nanoparticles, especially selenium, silver, and gold nanoparticles. While outlining the benefits, we outline possible methods for developing metallic nanoparticles, characterization techniques, and nephroprotection therapies. Selenium nanoparticles (SeNPs) minimize oxidative stress, a primary cause of nephrotoxicity through cell regeneration which protects kidneys. Silver nanoparticles (AgNPs) have anti-inflammatory capabilities that help alleviate kidney damage and nephrotoxicity. Gold nanoparticles (AuNPs), which are biocompatible and immune-modifying, reduce inflammation and promote renal cell regeneration, indicating nephroprotective advantages. Renal protection via the use of metallic nanoparticles represents a promising new frontier in the fight against kidney disease and other renal disorders. Metallic nanoparticles of selenium, silver, and gold can protect the kidneys by lowering oxidative stress, reducing inflammation, and improving cell repair. Through their mechanisms, these nanoparticles effectively safeguard and repair kidney function, making them suitable for treating renal diseases. The potential applications of selenium, silver, and gold nanoparticles, as well as their complex modes of action and renal penetration, provide fresh hope for improving renal health and quality of life in patients with kidney disease. The current study highlights therapeutic ability, stability, nephroprotection, and toxicity profiles, as well as the importance of continuous research in this dynamic and evolving field.
Collapse
Affiliation(s)
- Karthik K Karunakar
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Elizabeth Rani Edwin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Meenaloshini Gopalakrishnan
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India.
| | - Velmurugan Ramaiyan
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - V S Karthikha
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Jerry Peliks Justin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| |
Collapse
|
28
|
Park M, Kim YS, Song H. Macrophages: a double-edged sword in female reproduction and disorders. Exp Mol Med 2025; 57:285-297. [PMID: 39894821 PMCID: PMC11873061 DOI: 10.1038/s12276-025-01392-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
Reproduction consists of sequential inflammation-like events, primarily within the endometrium, from ovulation to embryo implantation, decidualization and delivery. During the reproductive cycle, the endometrium repeatedly undergoes cyclic periods of proliferation, differentiation, tissue breakdown and repair without scarring. Owing to their phagocytic activity, macrophages, key players in innate immunity, are thought to play crucial roles in the endometrium. Endometrial macrophages actively participate in various stages of reproductive tissue remodeling, particularly during decidualization and pregnancy establishment. Traditionally considered simple bystanders that clear debris to prevent autoimmune responses in tissue homeostasis, macrophages are now recognized as main actors with broad functional plasticity that allows them to fine tune the balance between pro- and anti-inflammatory responses during tissue inflammation, remodeling and repair. Homeostatic balance is determined by the sum of various mediators produced by two distinctly polarized macrophage subpopulations. The biased polarization of tissue-resident macrophages may contribute to the pathogenesis of various diseases, such as inflammation and cancer. Thus, understanding how macrophages contribute to endometrial homeostasis is crucial for deciphering the underlying mechanisms of various reproductive disorders. Nanomedicines using extracellular vesicles, nanoparticles and noncoding RNAs have recently been applied to modulate macrophage polarization and alleviate disease phenotypes. Despite these advances, the functions of endometrial macrophages under physiological and pathophysiological conditions remain poorly understood, which complicates the development of targeted therapies. Here we update the current understanding of the homeostatic function of macrophages and the putative contribution of endometrial macrophage dysfunction to reproductive disorders in women, along with innovative molecular therapeutics to resolve this issue.
Collapse
Affiliation(s)
- Mira Park
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, Korea
| | - Yeon Sun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, Korea
| | - Haengseok Song
- Department of Biomedical Science, College of Life Science, CHA University, Pocheon, Korea.
- Division of Life Science, CHA University, Pocheon, Korea.
- Department of Life Science, Graduate School, CHA University, Pocheon, Korea.
- CHA Advanced Research Institute, Seongnam, Korea.
- KW-Bio Co., Chuncheon, Korea.
| |
Collapse
|
29
|
Chavda VP, Joshi D. Surface modified proteins and peptides for targeted drug delivery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 212:389-438. [PMID: 40122652 DOI: 10.1016/bs.pmbts.2024.12.001] [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: 03/25/2025]
Abstract
Surface modification of proteins and peptides has emerged as a promising strategy to enhance their therapeutic efficacy and target specificity. This chapter delves into the various techniques employed to modify the surface properties of these biomolecules, including chemical conjugation, site-specific mutagenesis, and peptide synthesis. The focus is on strategies that improve drug delivery to specific target sites, such as tumor cells or inflamed tissues. By modifying surface properties, it is possible to enhance drug stability, reduce immunogenicity, and prolong circulation time. This chapter explores the latest advancements in this field and discusses the potential applications of surface-modified proteins and peptides in the development of novel therapeutic agents.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India.
| | - Disha Joshi
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, L J University, Ahmedabad, Gujarat, India
| |
Collapse
|
30
|
Grimaldi MC, Bozzer S, Sjöström DJ, Andersson LI, Mollnes TE, Nilsson PH, De Maso L, Riccardi F, Dal Bo M, Sblattero D, Macor P. DNA-loaded targeted nanoparticles as a safe platform to produce exogenous proteins in tumor B cells. Front Immunol 2025; 15:1509322. [PMID: 39911576 PMCID: PMC11794205 DOI: 10.3389/fimmu.2024.1509322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025] Open
Abstract
Introduction The functionalization of nanoparticles (NPs) with an antiCD19 targeting mechanism represents a promising approach for the selective delivery of drugs and nucleic acids into normal and tumor B cells. This strategy has the advantage of minimizing off-target effects by restricting gene delivery to the desired cell population. However, the nanoplatform must guarantee both the local production of the protein and the safety of the treatment to allow an effective therapy with reduced systemic toxicity. Methods In order to ensure a selective delivery of nucleic acids, we developed poly(lactic-co-glycolic acid) (PLGA)-poly(vinyl alcohol) (PVA) NPs loaded with an Enhanced Green Fluorescent Protein (EGFP)-coding plasmid and covalently coated with antiCD19 recombinant antibody as a targeting mechanism. To assess the functionality of the NPs, physicochemical characterization, safety tests, and transfection assay were employed to evaluate the NPs' behavior in vitro and in vivo, in a human/zebrafish lymphoma xenograft model. Results The results demonstrated that the PLGA-PVA nanoplatform was capable of efficiently encapsulating and releasing the payload. These nanostructures demonstrated a favorable safety profile, as evidenced by the absence of significant cell cytotoxicity, coagulation activation, complement system activation, and the slight activation of endothelial cells and leukocytes. The targeting mechanism facilitated the interaction of NPs with target cells, thereby enhancing their internalization and subsequent exogenous plasmid DNA (pDNA) translation and protein expression. In the human/zebrafish lymphoma xenograft model, no evidence of toxicity was observed, and targeted NPs demonstrated the capacity to enhance exogenous pDNA expression. Conclusion Our findings provide a rationale for the use of targeted NPs as a DNA delivery system for the local expression of therapeutic proteins.
Collapse
Affiliation(s)
| | - Sara Bozzer
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Dick J. Sjöström
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Linnea I. Andersson
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodo, Norway
| | - Per H. Nilsson
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Luca De Maso
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | | | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
31
|
Khalid Salah Al-Sheakly B, Saheb Sharif-Askari F, Saheb Sharif-Askari N, Hundt JE, Halwani R. The potential role of nanobodies in asthma therapy. Front Pharmacol 2025; 15:1510806. [PMID: 39902079 PMCID: PMC11788342 DOI: 10.3389/fphar.2024.1510806] [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: 10/13/2024] [Accepted: 12/27/2024] [Indexed: 02/05/2025] Open
Abstract
Asthma is a chronic inflammatory disease of the airways characterized by bronchoconstriction, airway hyperresponsiveness, and mucus production. The pathophysiology of asthma involves a complex interplay of immune cells and mediators, including cytokines, chemokines, and other inflammatory molecules. Despite advances in asthma management, many patients continue to experience symptoms due to the limitations of current therapies. Monoclonal antibodies (mAbs) targeting specific inflammatory mediators have improved treatment outcomes for some patients, but challenges such as poor tissue penetration and high costs remain. Nanobodies (Nbs), a novel class of single-domain antibodies, offer a promising alternative due to their small size, stability, and potential for enhanced tissue penetration. This review discusses the key mediators involved in asthma, challenges in current treatments, and the potential of Nbs as a new therapeutic strategy. We also explore current studies and innovations in nanobody technology.
Collapse
Affiliation(s)
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jennifer E. Hundt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pediatrics, Faculty of Medicine, Prince Abdullah Ben Khaled Celiac Disease Chair, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
32
|
Dhandapani S, Ha Y, Wang R, Kwon TW, Cho IH, Kim YJ. Lipid-encapsulated gold nanoparticles: an advanced strategy for attenuating the inflammatory response in SARS-CoV-2 infection. J Nanobiotechnology 2025; 23:15. [PMID: 39815303 PMCID: PMC11734238 DOI: 10.1186/s12951-024-03064-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 12/05/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Nanodrugs play a crucial role in biomedical applications by enhancing drug delivery. To address safety and toxicity concerns associated with nanoparticles, lipid-nanocarrier-based drug delivery systems have emerged as a promising approach for developing next-generation smart nanomedicines. Ginseng has traditionally been used for various therapeutic purposes, including antiviral activity. This study aimed to prepare a biocompatible and therapeutically potent Korean ginseng nanoemulsion (KGS-NE) using ginseng seed oil (GSO), optimize its encapsulation and drug delivery efficiency, and evaluate its antiviral activity. RESULTS Various techniques were utilized to confirm the KGS-NE formation. Energy-dispersive X-ray spectroscopy identified gold nanoparticles with the highest Au peak at 2.1 keV. Selected area diffraction patterns revealed crystallographic structures. FT-IR spectrometry detected functional groups, with peaks at 2922.09 cm-1 (alkene C-H stretching), 1740.24 cm-1 (aldehyde C=O stretching), and 1098.07 cm-1 (C-O stretching in secondary alcohol). Storage stability studies showed that KGS-NE maintained its size and stability for 6 months at 4 °C. The KGS-NE exhibited a dose-dependent suppression of HCoV-OC43 viral replication in Vero E6 cells. RNA sequencing analysis unveiled differentially expressed genes (DEGs) specifically involved in the ABC transporters signaling pathway. KGS-NE oral administration facilitated the recovery of mice induced with the receptor binding domain (RBD) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, as confirmed by inflammatory markers expression in lung tissue. In the Syrian hamster infected with the SARS-CoV-2 model, the lungs dissected showed enlarged morphology and induced inflammatory cytokines. This effect was mitigated with KGS-NE oral administration, as observed through H&E and qRT-PCR analysis. Biochemical analysis at various oral administration concentrations demonstrated that KGS-NE had no adverse effects on the kidney and liver. CONCLUSIONS Our findings strongly suggest that oral administering KGS-NE in mice and Syrian hamster models has the potential to effectively mitigate lung inflammation against coronavirus. This indicates a promising new strategy for developing the antiviral nano-agent against SARS-CoV-2.
Collapse
Affiliation(s)
- Sanjeevram Dhandapani
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Republic of Korea
| | - Yujeong Ha
- Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Rongbo Wang
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Republic of Korea
| | - Tae Woo Kwon
- Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ik-Hyun Cho
- Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Yeon-Ju Kim
- Graduate School of Biotechnology, and College of Life Science, Kyung Hee University, Yongin-Si, 17104, Gyeonggi-Do, Republic of Korea.
| |
Collapse
|
33
|
Yu C, Fan CQ, Chen YX, Guo F, Rao HH, Che PY, Zuo CJ, Chen HW. Global research trends and emerging hotspots in nano-drug delivery systems for lung cancer: a comprehensive bibliometric analysis (1998-2024). Discov Oncol 2025; 16:33. [PMID: 39798040 PMCID: PMC11724832 DOI: 10.1007/s12672-025-01782-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025] Open
Abstract
PURPOSE Nano-drug delivery systems (NDDS) have become a promising alternative and adjunctive strategy for lung cancer (LC) treatment. However, comprehensive bibliometric analyses examining global research efforts on NDDS in LC are scarce. This study aims to fill this gap by identifying key research trends, emerging hotspots, and collaboration networks within the field of NDDS and LC. METHODS A total of 2452 publications, spanning from 1998 to 2024, were extracted from the Web of Science Core Collection. The data were analyzed using tools such as VOSviewer, CiteSpace, and the R package 'bibliometrix'. RESULTS The analysis covered contributions from 12,539 researchers affiliated with 2689 institutions across 55 countries, with their work published in 551 different journals. Research output has increased steadily, with China and the United States leading in both publication volume and impact. Major contributors include the Chinese Academy of Sciences and Shanghai Jiao Tong University. The International Journal of Nanomedicine published the most articles, while Journal of Controlled Release ranked highest in co-citations. Kamal Dua authored the most papers, and Maeda, H. was the most frequently co-cited author. Key research areas encompass "active targeting", "drug delivery optimization", "overcoming drug resistance", "nanocarriers", and "pulmonary drug delivery". Emerging hotspots include "epithelial mesenchymal transition", "mucus penetration", "lipid nanoparticles", "hydrogels", and "immune checkpoint inhibitors". CONCLUSION This bibliometric analysis, the first comprehensive study on NDDS in LC, identifies China and the United States as leading contributors in publication volume and impact. Key research areas include "active targeting" and "drug delivery optimization", with emerging hotspots such as "lipid nanoparticles" and "immune checkpoint inhibitors". These findings provide essential insights to guide future research and optimize treatment strategies.
Collapse
Affiliation(s)
- Cao Yu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing, 400010, China
- Department of Cardiothoracic Surgery, Chongqing University Jiangjin Hospital, Chongqing, 402260, China
| | - Chong-Qi Fan
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing, 400010, China
| | - Yao-Xuan Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing, 400010, China
| | - Feng Guo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing, 400010, China
| | - Hao-Han Rao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing, 400010, China
| | - Peng-Yu Che
- Department of Cardiothoracic Surgery, The People's Hospital of Chongqing Hechuan, Chongqing, 410520, China
| | - Chun-Jian Zuo
- Department of Thoracic Surgery, Army Medical Center of People's Liberation Army of China (PLA), 10# Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.
| | - Huan-Wen Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
34
|
Shen Y, Cai R, Wu L, Han K, Yang Y, Mao D. Programmable Intelligent DNA Nanoreactors (iDNRs) for in vivo Tumor Diagnosis and Therapy. ChemMedChem 2025; 20:e202400531. [PMID: 39377119 DOI: 10.1002/cmdc.202400531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
With the rapid advancement of DNA technology, intelligent DNA nanoreactors (iDNRs) have emerged as sophisticated tools that harness the structural versatility and programmability of DNA. Due to their structural and functional programmability, iDNRs play an important and unique role in in vivo tumor diagnosis and therapy. This review provides an overview of the structural design methods for iDNRs based on advanced DNA technology, including enzymatic reaction-mediated and enzyme-free strategies. This review also focuses on how iDNRs achieve intelligence through functional design, as well as the applications of iDNRs for in vivo tumor diagnosis and therapy. In summary, this review summarizes current advances in iDNRs technology, discusses existing challenges, and proposes future directions for expanding their applications, which are expected to provide insights into the development of the field of in vivo tumor diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Ying Shen
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, P. R. China
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Rongkai Cai
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Liang Wu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Kun Han
- CAS Key Lab of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, P. R. China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Dongsheng Mao
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| |
Collapse
|
35
|
Almutary AG, Chauhan P, Baldaniya L, Menon SV, Kumar MR, Chaturvedi B, Sharma N, Chauhan AS, Abomughaid MM, M D, Paiva-Santos AC, Lakhanpal S, Jha NK. Overcoming challenges in the design of drug delivery systems targeting the central nervous system. Nanomedicine (Lond) 2025; 20:5-8. [PMID: 39564781 DOI: 10.1080/17435889.2024.2421157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, P.O. Box 59911, United Arab Emirates
| | - Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanad University, Rohtak, Haryana 124001, India
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot, 360003, Gujrat, India
| | - Soumya V Menon
- Department of Chemistry & Biochemistry, School of Sciences, JAIN (Deemed to be University), Banglore, Karnataka, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh- 531162, India
| | - Bhumi Chaturvedi
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research & Innovation, Uttaranchal University, Dehradun, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Dhivyadharshni M
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences & Technology (SBT), Galgotias University, Greater Noida, India
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
- Department of Biotechnology Engineering & Food Technology, Chandigarh University, Mohali, 140413, India
| |
Collapse
|
36
|
Luiz Correa J, Kikuchi L, Ferreira DG, Gomes da Silva F, de Oliveira KMP, de Souza M, Baesso ML, Yamanishi G, Urbano A, Negri M. Antifungal potential of silver nanoparticles stabilized with the flavonoid naringenin. J Med Microbiol 2025; 74. [PMID: 39836546 DOI: 10.1099/jmm.0.001945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025] Open
Abstract
Introduction. Fungal infections caused by yeast have increased in recent decades, becoming a major threat to public health.Hypothesis/Gap Statement. Antifungal therapy represents a challenging problem because, in addition to presenting many side effects, fungal resistance has been increasing in recent years. As a result, the search for new therapeutic agents has advanced with the use of new technologies such as nanoparticles (NPs).Aim. Synthesize, characterize and evaluate the antifungal potential of naringenin (NAR)-stabilized silver NPs.Methodology. The biosynthesis of NPs was stabilized using the NAR molecule and an aqueous solution of silver nitrate. The characterization of silver nanoparticles (AgNPs) was performed using different methods, which include UV-visible spectroscopy, powder X-ray diffraction (XRD), transmission electron microscopy, zeta potential measurements and Fourier transform infrared (FTIR) spectroscopy. Antifungal activity was evaluated against clinical isolates of Candida albicans by determining the MIC and the minimum fungicidal concentration (MFC).Results. The AgNP NAR showed a colloidal appearance with an average size of 14.71 nm and zeta potential measured at -33.3 mV, indicating a highly stable suspension. XRD analysis confirmed the crystal structure. FTIR spectra showed the presence of several functional groups of plant compounds, which play an important role in the coating and bioreduction processes. The antifungal activity against C. albicans showed an MIC of 3.55 µg ml-1 and an MFC of 7.1 µg ml-1. According to the growth kinetic assay in 12 h, there was a reduction of ~50% (<3 log10). Furthermore, AgNP NAR did not show mutagenic potential.Conclusion. The AgNP NAR obtained presented ideal characteristics for biomedical applications, good stability and promising antimicrobial activity.
Collapse
Affiliation(s)
- Jakeline Luiz Correa
- Graduate Program in Health Sciences, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
- Medical Mycology Laboratory, Department of Clinical Analysis and Biomedicine, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
| | - Larissa Kikuchi
- Graduate Program in Health Sciences, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
- Medical Mycology Laboratory, Department of Clinical Analysis and Biomedicine, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
| | - Deisiany Gomes Ferreira
- Graduate Program in Health Sciences, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
- Medical Mycology Laboratory, Department of Clinical Analysis and Biomedicine, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
| | - Fabiana Gomes da Silva
- Applied Microbiology Laboratory, Faculty of Biological and Environmental Sciences, Federal University of Grande Dourados, Street Joo Rosa Ges, 1761, Dourados, MS, Brazil
| | - Kelly Mari P de Oliveira
- Applied Microbiology Laboratory, Faculty of Biological and Environmental Sciences, Federal University of Grande Dourados, Street Joo Rosa Ges, 1761, Dourados, MS, Brazil
| | - Monique de Souza
- Department of Physics, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
| | - Mauro Luciano Baesso
- Department of Physics, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
| | - Gustavo Yamanishi
- Department of Physics, State University of Londrina, Celso Garcia Cid Highway, Km 380 - University Campus, Londrina, PR, Brazil
| | - Alexandre Urbano
- Department of Physics, State University of Londrina, Celso Garcia Cid Highway, Km 380 - University Campus, Londrina, PR, Brazil
| | - Melyssa Negri
- Medical Mycology Laboratory, Department of Clinical Analysis and Biomedicine, State University of Maring, Colombo Avenue, 5790, Maring, PR, Brazil
| |
Collapse
|
37
|
Mazumdar H, Khondakar KR, Das S, Halder A, Kaushik A. Artificial intelligence for personalized nanomedicine; from material selection to patient outcomes. Expert Opin Drug Deliv 2025; 22:85-108. [PMID: 39645588 DOI: 10.1080/17425247.2024.2440618] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/15/2024] [Accepted: 12/06/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Artificial intelligence (AI) is changing the field of nanomedicine by exploring novel nanomaterials for developing therapies of high efficacy. AI works on larger datasets, finding sought-after nano-properties for different therapeutic aims and eventually enhancing nanomaterials' safety and effectiveness. AI leverages patient clinical and genetic data to predict outcomes, guide treatments, and optimize drug dosages and forms, enhancing benefits while minimizing side effects. AI-supported nanomedicine faces challenges like data fusion, ethics, and regulation, requiring better tools and interdisciplinary collaboration. This review highlights the importance of AI regarding patient care and urges scientists, medical professionals, and regulators to adopt AI for better outcomes. AREAS COVERED Personalized Nanomedicine, Material Discovery, AI-Driven Therapeutics, Data Integration, Drug Delivery, Patient Centric Care. EXPERT OPINION Today, AI can improve personalized health wellness through the discovery of new types of drug nanocarriers, nanomedicine of specific properties to tackle targeted medical needs, and an increment in efficacy along with safety. Nevertheless, problems such as ethical issues, data security, or unbalanced data sets need to be addressed. Potential future developments involve using AI and quantum computing together and exploring telemedicine i.e. the Internet-of-Medical-Things (IoMT) approach can enhance the quality of patient care in a personalized manner by timely decision-making.
Collapse
Affiliation(s)
- Hirak Mazumdar
- Department of Computer Science and Engineering, Adamas University, Kolkata, India
| | | | - Suparna Das
- Department of Computer Science and Engineering, BVRIT HYDERABAD College of Engineering for Women, Hyderabad, India
| | - Animesh Halder
- Department of Electrical and Electronics Engineering, Adamas University, Kolkata, India
| | - Ajeet Kaushik
- Nano Biotech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL, USA
| |
Collapse
|
38
|
Hashim GM, Shahgolzari M, Hefferon K, Yavari A, Venkataraman S. Plant-Derived Anti-Cancer Therapeutics and Biopharmaceuticals. Bioengineering (Basel) 2024; 12:7. [PMID: 39851281 PMCID: PMC11759177 DOI: 10.3390/bioengineering12010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/26/2025] Open
Abstract
In spite of significant advancements in diagnosis and treatment, cancer remains one of the major threats to human health due to its ability to cause disease with high morbidity and mortality. A multifactorial and multitargeted approach is required towards intervention of the multitude of signaling pathways associated with carcinogenesis inclusive of angiogenesis and metastasis. In this context, plants provide an immense source of phytotherapeutics that show great promise as anticancer drugs. There is increasing epidemiological data indicating that diets rich in vegetables and fruits could decrease the risks of certain cancers. Several studies have proved that natural plant polyphenols, such as flavonoids, lignans, phenolic acids, alkaloids, phenylpropanoids, isoprenoids, terpenes, and stilbenes, could be used in anticancer prophylaxis and therapeutics by recruitment of mechanisms inclusive of antioxidant and anti-inflammatory activities and modulation of several molecular events associated with carcinogenesis. The current review discusses the anticancer activities of principal phytochemicals with focus on signaling circuits towards targeted cancer prophylaxis and therapy. Also addressed are plant-derived anti-cancer vaccines, nanoparticles, monoclonal antibodies, and immunotherapies. This review article brings to light the importance of plants and plant-based platforms as invaluable, low-cost sources of anti-cancer molecules of particular applicability in resource-poor developing countries.
Collapse
Affiliation(s)
- Ghyda Murad Hashim
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Mehdi Shahgolzari
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan 65175-4171, Iran
| | - Kathleen Hefferon
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Afagh Yavari
- Department of Biology, Payame Noor University, Tehran P.O. Box 19395-3697, Iran
| | - Srividhya Venkataraman
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| |
Collapse
|
39
|
Sanati M, Figueroa-Espada CG, Han EL, Mitchell MJ, Yavari SA. Bioengineered Nanomaterials for siRNA Therapy of Chemoresistant Cancers. ACS NANO 2024; 18:34425-34463. [PMID: 39666006 DOI: 10.1021/acsnano.4c11259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Chemoresistance remains a long-standing challenge after cancer treatment. Over the last two decades, RNA interference (RNAi) has emerged as a gene therapy modality to sensitize cancer cells to chemotherapy. However, the use of RNAi, specifically small-interfering RNA (siRNA), is hindered by biological barriers that limit its intracellular delivery. Nanoparticles can overcome these barriers by protecting siRNA in physiological environments and facilitating its delivery to cancer cells. In this review, we discuss the development of nanomaterials for siRNA delivery in cancer therapy, current challenges, and future perspectives for their implementation to overcome cancer chemoresistance.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 97178, Iran
| | - Christian G Figueroa-Espada
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Emily L Han
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| |
Collapse
|
40
|
Adjei-Sowah E, Rangasami V, Loiselle AE, Benoit DSW. Optimizing Ligand Valency to Maximize Tendon Accumulation of Peptide-Targeted Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68864-68876. [PMID: 39630483 DOI: 10.1021/acsami.4c13388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
In many tissues, including musculoskeletal tissues such as tendon, systemic delivery typically results in poor targeting of free drugs. Hence, we previously developed a targeted drug delivery nanoparticle (NP) system for tendon healing, leveraging a tartrate resistant acid phosphatase (TRAP) binding peptide (TBP) ligand. The greatest tendon targeting was observed with NPs functionalized with 30 000 TBP ligands per NP at day 7 during the proliferative healing phase, relative to the inflammatory (day 3) and early remodeling (day 14) phases of healing. Nevertheless, TRAP activity varies throughout healing and, therefore, may offer an opportunity for optimizing temporal therapeutic targeting through multivalent interactions. Hence, in this study, we hypothesized that the ligand density (9000-55,000 TBPs per NP) can optimize tendon accumulation on the basis of variable TRAP levels. The multivalent nanoparticles were loaded with three different fluorophores. In vitro, the ligand density and fluorophore had no effect on the physicochemical properties of the NPs, including size, charge, polydispersity index, or dye loading efficiency; however, the TRAP binding affinity correlated positively with the ligand density. In vivo, the ligand density correlated positively with NP homing and retention in the tendon, establishing opportunities to leverage ligand density for tendon targeting across the tendon healing cascade, during aging, and in other tendon pathologies, including tendinopathies.
Collapse
Affiliation(s)
- Emmanuela Adjei-Sowah
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14623, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Vigneshkumar Rangasami
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, Oregon 97403, United States
| | - Alayna E Loiselle
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14623, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
- Department of Orthopedics & Physical Performance, University of Rochester Medical Center, Rochester, New York 14642, United States
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14623, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, Oregon 97403, United States
- Department of Orthopedics & Physical Performance, University of Rochester Medical Center, Rochester, New York 14642, United States
| |
Collapse
|
41
|
Lee MY, Lee D, Choi D, Kim KS, Kang PM. Targeting Reactive Oxygen Species for Diagnosis of Various Diseases. J Funct Biomater 2024; 15:378. [PMID: 39728178 DOI: 10.3390/jfb15120378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024] Open
Abstract
Reactive oxygen species (ROS) are generated predominantly during cellular respiration and play a significant role in signaling within the cell and between cells. However, excessive accumulation of ROS can lead to cellular dysfunction, disease progression, and apoptosis that can lead to organ dysfunction. To overcome the short half-life of ROS and the relatively small amount produced, various imaging methods have been developed, using both endogenous and exogenous means to monitor ROS in disease settings. In this review, we discuss the molecular mechanisms underlying ROS production and explore the methods and materials that could be used to detect ROS overproduction, including iron-based materials, ROS-responsive chemical bond containing polymers, and ROS-responsive molecule containing biomaterials. We also discuss various imaging and imaging techniques that could be used to target and detect ROS overproduction. We discuss the ROS imaging potentials of established clinical imaging methods, such as magnetic resonance imaging (MRI), sonographic imaging, and fluorescence imaging. ROS imaging potentials of other imaging methods, such as photoacoustic imaging (PAI) and Raman imaging (RI) that are currently in preclinical stage are also discussed. Finally, this paper focuses on various diseases that are associated with ROS overproduction, and the current and the future clinical applications of ROS-targeted imaging. While the most widely used clinical condition is cardiovascular diseases, its potential extends into non-cardiovascular clinical conditions, such as neurovascular, neurodegenerative, and other ROS-associated conditions, such as cancers, skin aging, acute kidney injury, and inflammatory arthritis.
Collapse
Affiliation(s)
- Moung Young Lee
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Centers for Research in ICT based Infectious Diseases, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Donguk Lee
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| | - Dayun Choi
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| | - Kye S Kim
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Peter M Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
42
|
Naskar A, Kilari S, Baranwal G, Kane J, Misra S. Nanoparticle-Based Drug Delivery for Vascular Applications. Bioengineering (Basel) 2024; 11:1222. [PMID: 39768040 PMCID: PMC11673055 DOI: 10.3390/bioengineering11121222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 01/05/2025] Open
Abstract
Nanoparticle (NP)-based drug delivery systems have received widespread attention due to the excellent physicochemical properties of nanomaterials. Different types of NPs such as lipid NPs, poly(lactic-co-glycolic) acid (PLGA) NPs, inorganic NPs (e.g., iron oxide and Au), carbon NPs (graphene and carbon nanodots), 2D nanomaterials, and biomimetic NPs have found favor as drug delivery vehicles. In this review, we discuss the different types of customized NPs for intravascular drug delivery, nanoparticle behaviors (margination, adhesion, and endothelium uptake) in blood vessels, and nanomaterial compatibility for successful drug delivery. Additionally, cell surface protein targets play an important role in targeted drug delivery, and various vascular drug delivery studies using nanoparticles conjugated to these proteins are reviewed. Finally, limitations, challenges, and potential solutions for translational research regarding NP-based vascular drug delivery are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (A.N.); (S.K.); (G.B.); (J.K.)
| |
Collapse
|
43
|
Kaur G, Bisen S, Singh NK. Nanotechnology in retinal diseases: From disease diagnosis to therapeutic applications. BIOPHYSICS REVIEWS 2024; 5:041305. [PMID: 39512331 PMCID: PMC11540445 DOI: 10.1063/5.0214899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
Nanotechnology has demonstrated tremendous promise in the realm of ocular illnesses, with applications for disease detection and therapeutic interventions. The nanoscale features of nanoparticles enable their precise interactions with retinal tissues, allowing for more efficient and effective treatments. Because biological organs are compatible with diverse nanomaterials, such as nanoparticles, nanowires, nanoscaffolds, and hybrid nanostructures, their usage in biomedical applications, particularly in retinal illnesses, has increased. The use of nanotechnology in medicine is advancing rapidly, and recent advances in nanomedicine-based diagnosis and therapy techniques may provide considerable benefits in addressing the primary causes of blindness related to retinal illnesses. The current state, prospects, and challenges of nanotechnology in monitoring nanostructures or cells in the eye and their application to regenerative ophthalmology have been discussed and thoroughly reviewed. In this review, we build on our previously published review article in 2021, where we discussed the impact of nano-biomaterials in retinal regeneration. However, in this review, we extended our focus to incorporate and discuss the application of nano-biomaterials on all retinal diseases, with a highlight on nanomedicine-based diagnostic and therapeutic research studies.
Collapse
|
44
|
Thiruvengadam R, Dareowolabi BO, Moon EY, Kim JH. Nanotherapeutic strategy against glioblastoma using enzyme inhibitors. Biomed Pharmacother 2024; 181:117713. [PMID: 39615164 DOI: 10.1016/j.biopha.2024.117713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Glioblastoma is the most aggressive brain cancer and thus patients with glioblastoma have a severely low 5-year survival rate (<5 %). Glioblastoma damages neural centers, causing severe depression, anxiety, and cognitive disorders. Glioblastoma is highly resistant to most of available anti-tumor medications, due to heterogeneity of glioblastoma as well as the presence of stem-like cells. To overcome the challenges in the current medications against glioblastoma, novel medications that are effective in treating the aggressive and heterogenous glioblastoma should be developed. Enzyme inhibitor and nanomedicine have been getting attention because of effective anticancer efficacies of enzyme inhibitors and a role of nanomedicine as effective carrier of chemotherapeutic drugs by targeting specific tumor areas. Furthermore, a tumor-initiating neuroinflammatory microenvironment, which is crucial for glioblastoma progression, was linked with several carcinogenesis pathways. Therefore, in this review, first we summarize neuroinflammation and glioblastoma-related neuropathways. Second, we discuss the importance of enzyme inhibitors targeting specific proteins in relation with neuroinflammation and glioblastoma-related molecular mechanisms. Third, we summarize recent findings on the significance of nanotherapeutic anticancer drugs developed using natural or synthetic enzyme inhibitors against glioblastoma as well as currently available Food and Drug Administration (FDA)-approved drugs against glioblastoma.
Collapse
Affiliation(s)
- Rekha Thiruvengadam
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | | | - Eun-Yi Moon
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea.
| |
Collapse
|
45
|
Urbaniak T, Milasheuski Y, Musiał W. Zero-Order Kinetics Release of Lamivudine from Layer-by-Layer Coated Macromolecular Prodrug Particles. Int J Mol Sci 2024; 25:12921. [PMID: 39684632 DOI: 10.3390/ijms252312921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
To reduce the risk of side effects and enhance therapeutic efficiency, drug delivery systems that offer precise control over active ingredient release while minimizing burst effects are considered advantageous. In this study, a novel approach for the controlled release of lamivudine (LV) was explored through the fabrication of polyelectrolyte-coated microparticles. LV was covalently attached to poly(ε-caprolactone) via ring-opening polymerization, resulting in a macromolecular prodrug (LV-PCL) with a hydrolytic release mechanism. The LV-PCL particles were subsequently coated using the layer-by-layer (LbL) technique, with polyelectrolyte multilayers assembled to potentially modify the carrier's properties. The LbL assembly process was comprehensively analyzed, including assessments of shell thickness, changes in ζ-potential, and thermodynamic properties, to provide insights into the multilayer structure and interactions. The sustained LV release over 7 weeks was observed, following zero-order kinetics (R2 > 0.99), indicating a controlled and predictable release mechanism. Carriers coated with polyethylene imine/heparin and chitosan/heparin tetralayers exhibited a distinct increase in the release rate after 6 weeks and 10 weeks, respectively, suggesting that this coating can facilitate the autocatalytic degradation of the polyester microparticles. These findings indicate the potential of this system for long-term, localized drug delivery applications, requiring sustained release with minimal burst effects.
Collapse
Affiliation(s)
- Tomasz Urbaniak
- Department of Physical Chemistry and Biophysics, Pharmaceutical Faculty, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| | - Yauheni Milasheuski
- Department of Physical Chemistry and Biophysics, Pharmaceutical Faculty, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| | - Witold Musiał
- Department of Physical Chemistry and Biophysics, Pharmaceutical Faculty, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| |
Collapse
|
46
|
Almutairy B, Alharthi S, Ziora ZM, Ebrahimi Shahmabadi H. Synthesis, radiolabeling, and biodistribution of 99 m-technetium-labeled zif-8 nanoparticles for targeted imaging applications. 3 Biotech 2024; 14:293. [PMID: 39525365 PMCID: PMC11549264 DOI: 10.1007/s13205-024-04145-w] [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: 09/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
This study investigates the synthesis and radiolabeling of zeolitic imidazolate frameworks (ZIF-8) with the radioisotope technetium-99 m (99mTc) using a solvothermal method in methanol. The methanolic medium facilitated the formation of nanoparticles with favorable characteristics, including a smaller particle size (198 ± 9.8 nm) and a low polydispersity index (PDI = 0.219 ± 0.011). Radiolabeling efficiency (RE%) and radiochemical purity (RCP%) were optimized by employing SnCl2 as a reducing agent, resulting in an RE% of 95.2 ± 1.9% and an RCP% of 96.1 ± 1.7% in triplicate (n = 3) at 65 °C. The nanoparticles exhibited high serum stability, retaining 99.05% of RCP% after 24 h, and demonstrated hemocompatibility, with hemolysis rates below 5% across all tested concentrations. In vitro biocompatibility assessments using NIH-3T3 cells indicated cell viability above 70% at concentrations up to 40 μg/mL. Biodistribution studies in rabbits (n = 6) revealed predominant accumulation in the bladder, with radiotracer uptake in the bladder being 6.3, 7.2, and 36.2 times higher than in the liver, kidneys, and heart (p < 0.0001), respectively, suggesting renal clearance. These results underscore the potential of 99mTc-(ZIF-8) nanoparticles for biomedical applications, particularly in targeted imaging and drug delivery. Future research will focus on improving targeting specificity and enhancing therapeutic efficacy in disease models. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04145-w.
Collapse
Affiliation(s)
- Bandar Almutairy
- Department of Pharmacology, College of Pharmacy, Shaqra University, 11961 Shaqra, Saudi Arabia
| | - Sitah Alharthi
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Al-Dawadmi Campus, Al-Dawadmi, 11961 Shaqra, Saudi Arabia
| | - Zyta M. Ziora
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Hasan Ebrahimi Shahmabadi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, 7718175911 Iran
| |
Collapse
|
47
|
Patel H, Patel A, Vats M, Patel K. Albumin and Polysorbate-80 Coated Sterile Nanosuspensions of Mebendazole for Glioblastoma Therapy. AAPS PharmSciTech 2024; 25:271. [PMID: 39586850 DOI: 10.1208/s12249-024-02978-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/17/2024] [Indexed: 11/27/2024] Open
Abstract
The scarcity of existing and novel therapies for brain cancer has significantly affected the survival rate of glioblastoma patients. Mebendazole (MBZ), an antiparasitic agent demonstrated promising activity against brain cancer. However, poor solubility, multiple polymorphs, and insufficient permeability through blood-brain barrier (BBB) restricts its therapeutic efficacy through parenteral administration. The current study aimed to develop, optimize, and characterize sterile, injectable nanosuspension of mebendazole using parenterally acceptable stabilizers. Albumin and polysorbate 80 (PS-80) coated MBZ Nanosuspension (NS) was prepared using wet media milling technique. Design of experiment (DoE) approach was used to understand effect of drug loading versus stabilizer concentration. The optimized MBZ NS showed hydrodynamic diameter of 208.36 ± 0.24 nm with a poly dispersibility index (PDI) of 0.210 ± 0.03 and zeta potential of -20.41 ± 0.36 mV. The IC50 value of MBZ NS in U-87 MG and LN-229 cell lines were found to be 0.49 ± 0.02 μM and 0.48 ± 0.05 μM, respectively. Additionally, MBZ NS demonstrated a 2.65-fold decrease in colony-forming efficiency and a 1.16-fold reduction in migration of the bridging area compared to MBZ. In 3D spheroids of the U-87 MG glioma cell line, MBZ NS exhibited a 50% reduction in tumor growth and increased cell apoptosis compared to the control. MBZ NS formulations were sterilized by gamma irradiation and tested as per the USP sterility test. Albumin-PS 80 coated NS is rendered to be useful parenteral delivery of mebendazole for the treatment of brain cancer.
Collapse
Affiliation(s)
- Himaxi Patel
- College of Pharmacy and Health Sciences, St. John's University, St. Albert Hall, B-49, 8000 Utopia Parkway, Queens, New York, 11439, USA
| | - Akanksha Patel
- College of Pharmacy and Health Sciences, St. John's University, St. Albert Hall, B-49, 8000 Utopia Parkway, Queens, New York, 11439, USA
| | - Mukti Vats
- College of Pharmacy and Health Sciences, St. John's University, St. Albert Hall, B-49, 8000 Utopia Parkway, Queens, New York, 11439, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, St. Albert Hall, B-49, 8000 Utopia Parkway, Queens, New York, 11439, USA.
| |
Collapse
|
48
|
Abarova S, Grancharova T, Zagorchev P, Tenchov B, Pilicheva B. Novel Spectroscopic Studies of the Interaction of Three Different Types of Iron Oxide Nanoparticles with Albumin. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1861. [PMID: 39683250 DOI: 10.3390/nano14231861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024]
Abstract
In the present work, we studied the interactions of three types of iron oxide nanoparticles (IONPs) with human serum albumin (HSA) by fluorescence and UV-Vis spectroscopy. The determined binding parameters of the reactions and the thermodynamic parameters, including ΔHo, ΔSo, and ΔGo indicated that electrostatic forces play a major role in the interaction of IONPs with HSA. These measurements indicate a fluorescent quenching mechanism based on IONPs-HSA static complex formation. Our study shows that the interaction between HSA and IONPs depends on the nanoparticle structure. The interaction between IONPs and HSA was found to be spontaneous, exothermic, and entropy-driven. HSA was shown to interact moderately with IONPs obtained with plant extracts of Uncaria tomentosa L. (IONP@UT) and Clinopodium vulgare L. (IONP@CV), and firmly with IONPs prepared with Ganoderma lingzhi (Reishi) extract (IONP@GL), via ground-state association. Analysis by modified Stern-Volmer approximation indicates that the quenching mechanism is static. Our study significantly improves our understanding of the mechanisms of interaction, distribution, and transport involved in the interaction between proteins and IONPs. It provides crucial insights into the functional perturbations of albumin binding capacity and the effects of IONPs on the stability and structural modifications of plasma carrier proteins.
Collapse
Affiliation(s)
- Silviya Abarova
- Department of Medical Physics and Biophysics, Faculty of Medicine, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Tsenka Grancharova
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Plamen Zagorchev
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Boris Tenchov
- Department of Medical Physics and Biophysics, Faculty of Medicine, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Bissera Pilicheva
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| |
Collapse
|
49
|
van Staden D, Gerber M, Lemmer HJR. The Application of Nano Drug Delivery Systems in Female Upper Genital Tract Disorders. Pharmaceutics 2024; 16:1475. [PMID: 39598598 PMCID: PMC11597179 DOI: 10.3390/pharmaceutics16111475] [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: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
The prevalence of female reproductive system disorders is increasing, especially among women of reproductive age, significantly impacting their quality of life and overall health. Managing these diseases effectively is challenging due to the complex nature of the female reproductive system, characterized by dynamic physiological environments and intricate anatomical structures. Innovative drug delivery approaches are necessary to facilitate the precise regulation and manipulation of biological tissues. Nanotechnology is increasingly considered to manage reproductive system disorders, for example, nanomaterial imaging allows for early detection and enhances diagnostic precision to determine disease severity and progression. Additionally, nano drug delivery systems are gaining attention for their ability to target the reproductive system successfully, thereby increasing therapeutic efficacy and decreasing side effects. This comprehensive review outlines the anatomy of the female upper genital tract by highlighting the complex mucosal barriers and their impact on systemic and local drug delivery. Advances in nano drug delivery are described for their sustainable therapeutic action and increased biocompatibility to highlight the potential of nano drug delivery strategies in managing female upper genital tract disorders.
Collapse
Affiliation(s)
| | | | - Hendrik J. R. Lemmer
- Centre of Excellence for Pharmaceutical Sciences (PharmacenTM), North-West University, Potchefstroom 2531, South Africa; (D.v.S.); (M.G.)
| |
Collapse
|
50
|
Moulton C, Baroni A, Quagliarini E, Leone L, Digiacomo L, Morotti M, Caracciolo G, Podda MV, Tasciotti E. Navigating the nano-bio immune interface: advancements and challenges in CNS nanotherapeutics. Front Immunol 2024; 15:1447567. [PMID: 39600701 PMCID: PMC11588692 DOI: 10.3389/fimmu.2024.1447567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
In recent years, significant advancements have been made in utilizing nanoparticles (NPs) to modulate immune responses within the central nervous system (CNS), offering new opportunities for nanotherapeutic interventions in neurological disorders. NPs can serve as carriers for immunomodulatory agents or platforms for delivering nucleic acid-based therapeutics to regulate gene expression and modulate immune responses. Several studies have demonstrated the efficacy of NP-mediated immune modulation in preclinical models of neurological diseases, including multiple sclerosis, stroke, Alzheimer's disease, and Parkinson's disease. While challenges remain, advancements in NPs engineering and design have led to the development of NPs using diverse strategies to overcome these challenges. The nano-bio interface with the immune system is key in the conceptualization of NPs to efficiently act as nanotherapeutics in the CNS. The biomolecular corona plays a pivotal role in dictating NPs behavior and immune recognition within the CNS, giving researchers the opportunity to optimize NPs design and surface modifications to minimize immunogenicity and enhance biocompatibility. Here, we review how NPs interact with the CNS immune system, focusing on immunosurveillance of NPs, NP-induced immune reprogramming and the impact of the biomolecular corona on NPs behavior in CNS immune responses. The integration of NPs into CNS nanotherapeutics offers promising opportunities for addressing the complex challenges of acute and chronic neurological conditions and pathologies, also in the context of preventive and rehabilitative medicine. By harnessing the nano-bio immune interface and understanding the significance of the biomolecular corona, researchers can develop targeted, safe, and effective nanotherapeutic interventions for a wide range of CNS disorders to improve treatment and rehabilitation. These advancements have the potential to revolutionize the treatment landscape of neurological diseases, offering promising solutions for improved patient care and quality of life in the future.
Collapse
Affiliation(s)
| | - Anna Baroni
- Human Longevity Program, IRCCS San Raffaele Roma, Rome, Italy
| | - Erica Quagliarini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Leone
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marta Morotti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Vittoria Podda
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ennio Tasciotti
- Human Longevity Program, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, Università telematica San Raffaele, Rome, Italy
| |
Collapse
|