1
|
Li B, Song S, Zhou Y, Chen X, Zhang Z, Liu X, Zhang R. Biopolymer hydrogels in biomedicine: Bridging chemistry, biology, and clinical translation. Int J Biol Macromol 2025; 318:145048. [PMID: 40490181 DOI: 10.1016/j.ijbiomac.2025.145048] [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: 03/10/2025] [Revised: 06/04/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
Biopolymer hydrogels are revolutionizing biomedicine by synergizing ecological sustainability with dynamic biofunctionality, offering transformative solutions for tissue regeneration and precision therapeutics. Derived from polysaccharides (e.g., chitosan, hyaluronic acid), proteins (e.g., collagen, silk fibroin), and nucleic acids, these hydrogels recapitulate critical extracellular matrix (ECM) features through tunable viscoelasticity (elastic modulus: 1-100 kPa), multi-stimuli responsiveness (enzyme/pH/ROS), and spatiotemporal bioactivity. Recent innovations leverage their structure-function relationships: protein-polysaccharide interpenetrating networks (e.g., gelatin-oxidized alginate) enable stage-specific cytokine release (e.g., IL-10/TGF-β dual delivery) and interfacial tissue regeneration, while glycosaminoglycan-mimetic hydrogels direct stem cell differentiation via stiffness-mediated YAP/TAZ mechanotransduction. However, clinical translation faces critical barriers: reconciling injectability with load-bearing resilience, synchronizing degradation kinetics with tissue remodeling phases, and modulating immune-microenvironment crosstalk to balance biointegration and fibrosis (e.g., M1-to-M2 macrophage polarization). Emerging strategies address these through covalent-noncovalent dual crosslinking, zwitterionic immunomodulatory interfaces, and 4D-bioprinted architectures with anisotropic bioactivity. Challenges persist in preventing phase separation in hybrid systems and matching metabolic kinetics in vivo. Future directions demand molecular-scale engineering of dynamic networks, machine learning-guided multi-omics customization, and standardized validation frameworks. This review delineates a roadmap for transforming natural hydrogels from bioactive scaffolds to intelligent, clinically viable systems for active tissue repair and disease modulation.
Collapse
Affiliation(s)
- Bailei Li
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, Zhejiang Province 314006, China; Zhejiang Key Laboratory of Multiomics and Molecular Enzymology, Yangtze Delta Region Institute of Tsinghua University, Zhejiang, Zhejiang 314006, China.
| | - Suxian Song
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, Zhejiang Province 314006, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Yu Zhou
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, Zhejiang Province 314006, China.
| | - Xinjiani Chen
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang 314001, China.
| | - Zhen Zhang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, Zhejiang Province 314006, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China.
| | - Xiaojun Liu
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, Zhejiang Province 314006, China; Zhejiang Key Laboratory of Multiomics and Molecular Enzymology, Yangtze Delta Region Institute of Tsinghua University, Zhejiang, Zhejiang 314006, China; Taizhou Innovation Center, Yangtze Delta Region Institute of Tsinghua University, Zhejiang 318000, China.
| | - Rongqing Zhang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, Zhejiang Province 314006, China; Zhejiang Key Laboratory of Multiomics and Molecular Enzymology, Yangtze Delta Region Institute of Tsinghua University, Zhejiang, Zhejiang 314006, China; Taizhou Innovation Center, Yangtze Delta Region Institute of Tsinghua University, Zhejiang 318000, China.
| |
Collapse
|
2
|
Abdel-Megeed RM. Biogenic nanoparticles as a promising drug delivery system. Toxicol Rep 2025; 14:101887. [PMID: 39867515 PMCID: PMC11762933 DOI: 10.1016/j.toxrep.2024.101887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/03/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Nanotechnology has significantly influenced the worldwide medical services sector during the past few decades. Biological collection approaches for nanoparticles are economical, non-toxic, and ecologically benign. This review provides up-to-date information on nanoparticle production processes and biological sources, including algae, plants, bacteria, fungus, actinomycetes, and yeast. The biological technique of generating nanoparticles has advantages over chemical, physical, and biological methods, including low-toxicity and friendly to the environment, thereby providing a viable option for therapeutic applications as s promising drug delivery system. In addition to aiding researchers, the bio-mediated, obtained nanoparticles also modify particles to promote both health and safety. We also looked at the important medicinal uses of nanoparticles, including their antifungal, antimicrobial, antiviral, antidiabetic, anti-inflammatory, and antioxidant properties. The current study highlights the findings of recent research in this field and discusses various methods proposed to describe the bio-mediated acquisition of novel nanoparticles.. The production of nanoparticles via biogenic sources possess various benefits, such as low cost, bioavailability, and environmental friendliness. In addition to the determination of the bioactive chemicals mediated by nanoparticle as well as the examination of the biochemical pathways and enzyme reactions. The major focus of this review is highlighting on the essential role of biogenic nanoparticles as promising drug delivery system.
Collapse
|
3
|
Russo S, Privitera A, Greco G, Di Pietro L, Cardaci V, Carota G, Sarpietro MG, Caruso G. Development and in vitro characterization of new carnosine-loaded liposomal formulations. J Liposome Res 2025; 35:117-124. [PMID: 39404506 DOI: 10.1080/08982104.2024.2415664] [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: 09/11/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 05/29/2025]
Abstract
Carnosine is an endogenous dipeptide characterized by a multimodal mechanism of action. However, its clinical potential is limited by serum and cytosolic carnosinases, which significantly reduce its bioavailability. Based on that, different research groups have worked on the development of new strategies able not only to prevent its rapid metabolization but also to improve its distribution and specific targeting. In the present study, the development and in vitro characterization of new liposomal formulations loaded with carnosine are described. Nanoliposomes, produced through Thin-Layer Hydration followed by Extrusion method, were first investigated for their physicochemical stability. Photon correlation spectroscopy and electrophoretic light scattering, assessing the stability of the formulations, showed a strong homogeneity-oriented tendency for up to two months. Particle size, polydispersity index, and zeta potential were determined through dynamic light scattering and electrophoretic light scattering, demonstrating an almost neutral charge of the formulation and an effective encapsulation of carnosine. The morphology assessment performed via scanning electron microscopy showed good conformity and polydispersity. Differential scanning calorimetry measurements suggest the ability of carnosine to stabilize the large unilamellar vesicles. Lastly, the newly developed carnosine-loaded liposomal formulations also showed a good safety profile in human microglia.
Collapse
Affiliation(s)
- Stefano Russo
- Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3), Mannheim Cancer Center (MCC), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anna Privitera
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuliana Greco
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Lucia Di Pietro
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Scuola Superiore di Catania, University of Catania, Catania, Italy
| | | | - Giuseppe Carota
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, Troina, Italy
| |
Collapse
|
4
|
Bacal CJO, Allardyce BJ, Valente F. Influence of material format and surface chemistry for the sustained delivery and efficacy of silk drug delivery systems in vivo. J Mater Chem B 2025. [PMID: 40400450 DOI: 10.1039/d4tb02756f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Silk fibroin materials are promising for use in controlled drug delivery in the field of tissue engineering and biomedical applications thanks to silk's generally established biocompatibility and tunable properties for implants and drug storage. Several factors must be considered in the materials design, including material format, drug properties and release kinetics, and the activity and stability of the drug after release. While numerous reviews described silk-based DDS that demonstrated controllable in vitro release, success in vivo has been limited, especially in some material formats. This review therefore aims to provide insight into the current material format and functionalization strategies to maximize in vivo performance by describing the in vivo activity of recently developed silk drug delivery systems. The review also aims to provide a fresh perspective on the suitable format and functionalization strategies for a target biomedical application. Based on the release behavior of drugs in various material formats, silk films, foams, and microneedles were better suited to serve as scaffolds for cell regeneration and improved recovery rate for biomedical applications involving wound healing and tissue engineering. Gels and particles could be incorporated within the films and foams but the purpose would be to serve as additional physical barriers towards drug diffusion in these types of application. For drugs or therapeutics that target internal organs (i.e. brain, liver, intestines, etc.), gels and particles were mainly used due to their size. In the event that the material format selection based on the target application does not contribute a lot to the prolonged release of drugs or therapeutic agents, hybrid functionalization strategies were adapted to make the surface chemistry of the material more responsive to the environmental stimuli for a more tunable silk DDS.
Collapse
Affiliation(s)
- Christine Jurene O Bacal
- Ear Science Institute Australia, Ear Sciences Centre, School of Medicine, The University of Western Australia, Nedlands 6009, Australia.
| | | | - Filippo Valente
- Ear Science Institute Australia, Ear Sciences Centre, School of Medicine, The University of Western Australia, Nedlands 6009, Australia.
| |
Collapse
|
5
|
Yadav PK, Chauhan D, Yadav P, Tiwari AK, Sultana N, Gupta D, Mishra K, Gayen JR, Wahajuddin M, Chourasia MK. Nanotechnology Assisted Drug Delivery Strategies for Chemotherapy: Recent Advances and Future Prospects. ACS APPLIED BIO MATERIALS 2025; 8:3601-3622. [PMID: 40318022 DOI: 10.1021/acsabm.5c00046] [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: 05/07/2025]
Abstract
In pursuit of the treatment of cancer, nanotechnology engineering has emerged as the simplest and most effective means, with the potential to deliver antitumor chemotherapeutics at the targeted site. Employing nanotechnology for drug delivery provides diverse nanosize particles ranging from one to a thousand nanometers. Reduced size improves drug bioavailability by increasing drug diffusion and decreasing the efflux rate. These nanocarriers offer an enormous scope for modification following the chemical and biological properties of both the drug and its disease. Moreover, these nanoformulations assist in targeting pharmaceutically active drug molecules to the desired site and have gained importance in recent years. Their modern use has revolutionized the antitumor action of many therapeutic agents. Higher drug loading efficiency, thermal stability, easy fabrication, low production cost, and large-scale industrial production draw attention to the application of nanotechnology as a better platform for the delivery of drug molecules. Furthermore, the interaction of nanocarrier technology-assisted agents lowers a drug's toxicity and therapeutic dosage, reduces drug tolerance, and enhances active drug concentration in neoplasm tissue, thus decreasing the concentration in healthy tissue. Nanotechnology-based medications are being widely explored and have depicted effective cancer management in vivo and in vitro systems, leading to many clinical trials with promising results. This review summarizes the innovative impact and application of different nanocarriers developed in recent years in cancer therapy. Subsequently, it also describes the essential findings and methodologies and their effects on cancer treatment. Compared with conventional therapy, nanomedicines can significantly improve the therapeutic effectiveness of antitumor drugs. Thus, the adverse effects associated with healthy tissues are decreased, and adverse effects are scaled back through enhanced permeability and retention effects. Lastly, future insights assisting nanotechnology in active therapeutics delivery and their scope in cancer chemotherapeutics have also been discussed.
Collapse
Affiliation(s)
- Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Chauhan
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pooja Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amrendra K Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nazneen Sultana
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Deepak Gupta
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Keerti Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Jiaur R Gayen
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Richmond Road, Bradford BD7 1DP, United Kingdom
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
6
|
Jiang J, Wang J, Fan P, Zhao Z, Deng H, Li J, Wang Y, Wang Y. Biomaterial-based strategies for bone cement: modulating the bone microenvironment and promoting regeneration. J Nanobiotechnology 2025; 23:343. [PMID: 40361125 PMCID: PMC12070552 DOI: 10.1186/s12951-025-03363-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoporotic bone defect and fracture healing remain significant challenges in clinical practice. While traditional therapeutic approaches provide some regulation of bone homeostasis, they often present limitations and adverse effects. In orthopedic procedures, bone cement serves as a crucial material for stabilizing osteoporotic bone and securing implants. However, with the exception of magnesium phosphate cement, most cement variants lack substantial bone regenerative properties. Recent developments in biomaterial science have opened new avenues for enhancing bone cement functionality through innovative modifications. These advanced materials demonstrate promising capabilities in modulating the bone microenvironment through their distinct physicochemical properties. This review provides a systematic analysis of contemporary biomaterial-based modifications of bone cement, focusing on their influence on the bone healing microenvironment. The discussion begins with an examination of bone microenvironment pathology, followed by an evaluation of various biomaterial modifications and their effects on cement properties. The review then explores regulatory strategies targeting specific microenvironmental elements, including inflammatory response, oxidative stress, osteoblast-osteoclast homeostasis, vascular network formation, and osteocyte-mediated processes. The concluding section addresses current technical challenges and emerging research directions, providing insights for the development of next-generation biomaterials with enhanced functionality and therapeutic potential.
Collapse
Affiliation(s)
- Jiawei Jiang
- Medical School of Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Juan Wang
- Central Laboratory, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, Jiangsu, China
| | - Pan Fan
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Zhe Zhao
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China
| | - Hongjian Deng
- Department of Orthopaedics, The Affiliated 2 Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jian Li
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China.
| | - Yi Wang
- Department of Orthopaedics, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang, 332000, Jiangxi, China.
| | - Yuntao Wang
- Medical School of Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Orthopaedics, Xuyi People's Hospital, Xuyi, 211700, Jiangsu, China.
| |
Collapse
|
7
|
Kaňa M, Braunová A, Starenko D, Frejková M, Bouček J, Říhová B, Kovář M, Etrych T, Šírová M. Overcoming P-glycoprotein-mediated multidrug resistance in cancer cells through micelle-forming PHPMA-b-PPO diblock copolymers for doxorubicin delivery. J Control Release 2025; 381:113645. [PMID: 40112897 DOI: 10.1016/j.jconrel.2025.113645] [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: 12/18/2024] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Multidrug resistance (MDR) represents one of the major concerns in cancer therapy as it may cause reduced efficacy of chemotherapeutic drugs due to the overexpression of ABC transporters, particularly P-glycoprotein (P-gp). This study explores the potential of novel amphiphilic diblock (DB) copolymers composed of poly[N-(2-hydroxypropyl)methacrylamide]-based copolymers (PHPMA) and poly(propylene oxide) (PPO) to overcome MDR mechanisms. The DB copolymers and their doxorubicin (Dox) conjugates significantly increased Dox accumulation in P-gp positive cells, markedly sensitizing them to Dox cytotoxic activity. The underlying mechanisms included depletion of intracellular ATP with subsequent inhibition of P-gp mediated drug efflux, an altered mitochondrial membrane potential, and increased ROS production. Moreover, the DB-Dox conjugates inhibited tumor growth in vivo more effectively compared to the corresponding PHPMA-based drug delivery system. Copolymers with additionally loaded PPO in the micelle core demonstrated superior efficacy in terms of P-gp inhibition, ATP depletion, and chemosensitizing effect in vitro, as well as antitumor activity in vivo. DB copolymers effectively depleted ATP levels both in vitro and in vivo using patient-derived xenograft (PDX) models, underscoring their capacity to enhance the effectiveness of standard chemotherapy and translational potential.
Collapse
MESH Headings
- Doxorubicin/administration & dosage
- Doxorubicin/chemistry
- Doxorubicin/therapeutic use
- Micelles
- Humans
- Animals
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Multiple/drug effects
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/therapeutic use
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Cell Line, Tumor
- Propylene Glycols/chemistry
- Propylene Glycols/administration & dosage
- Mice, Nude
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Neoplasms/pathology
- Mice, Inbred BALB C
- Polypropylenes/chemistry
- Female
- Adenosine Triphosphate/metabolism
- Mice
- Drug Carriers/chemistry
- Polymers/chemistry
- Methacrylates/chemistry
Collapse
Affiliation(s)
- Martin Kaňa
- Institute of Microbiology, Czech Academy of Sciences, Czech Republic; Department of Otorhinolaryngology and Head and Neck Surgery, First faculty of Medicine, Charles University and University Hospital Motol, Czech Republic
| | - Alena Braunová
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Czech Republic
| | - Daniil Starenko
- Department of Otorhinolaryngology and Head and Neck Surgery, First faculty of Medicine, Charles University and University Hospital Motol, Czech Republic
| | - Markéta Frejková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Czech Republic
| | - Jan Bouček
- Institute of Microbiology, Czech Academy of Sciences, Czech Republic
| | - Blanka Říhová
- Department of Otorhinolaryngology and Head and Neck Surgery, First faculty of Medicine, Charles University and University Hospital Motol, Czech Republic
| | - Marek Kovář
- Department of Otorhinolaryngology and Head and Neck Surgery, First faculty of Medicine, Charles University and University Hospital Motol, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Czech Republic
| | - Milada Šírová
- Department of Otorhinolaryngology and Head and Neck Surgery, First faculty of Medicine, Charles University and University Hospital Motol, Czech Republic.
| |
Collapse
|
8
|
Yang JW, Yoon T, Kim H, Joo KI, Cha HJ. Acrylated Bioengineered Mussel Protein-Based Adhesive Nanoparticles for Locoregional and Sustained Drug Delivery. ACS Biomater Sci Eng 2025. [PMID: 40335274 DOI: 10.1021/acsbiomaterials.5c00390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Nanoparticles have emerged as promising drug carriers owing to their ability to permeate cell membranes and enhance drug stability. However, their clinical application faces significant challenges, including rapid diffusion, inefficient retention at target sites, and burst drug release. This study proposes the use of adhesive nanoparticles derived from acrylated bioengineered mussel adhesive proteins (MAPs). Acrylic groups were conjugated to lysine residues in MAPs to form polyacrylate-MAPs by photo-cross-linking, retaining sufficient 3,4-dihydroxyphenylalanine residues for strong tissue adhesion in aqueous environments. These nanoparticles were designed to adhere effectively to the administration sites and facilitate continuous drug release. In vitro and in vivo evaluations demonstrated that the acrylated MAP-based nanoparticles exhibited superior wet adhesive properties, sustained drug release, and long-term retention at the administration site and effectively suppressed tumor growth, ensuring that a single dose maintained a therapeutic concentration at the target site over extended periods. Thus, this approach could address the challenges of drug localization and retention, significantly improving therapeutic efficacy. This study emphasizes the versatility of bioengineered MAP-based adhesive nanoparticles for locoregional and sustained drug delivery, with promising applications in cancer therapy, regenerative medicine, and other biomedical fields.
Collapse
Affiliation(s)
- Jang Woo Yang
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Taehee Yoon
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Haram Kim
- Division of Chemical Engineering and Materials Science, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kye Il Joo
- Division of Chemical Engineering and Materials Science, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hyung Joon Cha
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| |
Collapse
|
9
|
Liu H, Li Y, Wang Y, Zhang L, Liang X, Gao C, Yang Y. Red blood cells-derived components as biomimetic functional materials: Matching versatile delivery strategies based on structure and function. Bioact Mater 2025; 47:481-501. [PMID: 40034412 PMCID: PMC11872572 DOI: 10.1016/j.bioactmat.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 03/05/2025] Open
Abstract
Red blood cells (RBCs), often referred to as "intelligent delivery systems", can serve as biological or hybrid drug carriers due to their inherent advantages and characteristics. This innovative approach has the potential to enhance biocompatibility, pharmacokinetics, and provide targeting properties for drugs. By leveraging the unique structure and contents of RBCs, drug-loading pathways can be meticulously designed to align with these distinctive features. This review article primarily discusses the drug delivery strategies and their applications that are informed by the structural and functional properties of the main components of RBCs, including living RBCs, membranes, hollow RBCs, and hemoglobin. Overall, this review article would assist efforts to make better decisions on optimization and rational utilization of RBCs derivatives-based drug delivery strategies for the future direction in clinical translation.
Collapse
Affiliation(s)
- Hangbing Liu
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Yi Li
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| | - Yuli Wang
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| | - Liying Zhang
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Xiaoqing Liang
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| | - Chunsheng Gao
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, People's Republic of China
| |
Collapse
|
10
|
Cao Y, Hu X, Wu D, Jiang Y, Yu Y, Wang S, Chen W, Long Y, Xu L, Qu J, Yang B, Chakhabi B, Wang H, Deng Y, Chen L, Chen Z, Li Q. Targeting capacity, safety and efficacy of engineered extracellular vesicles delivered by transdermal microneedles to treat plasmacytoma in mice. Clin Transl Med 2025; 15:e70327. [PMID: 40317915 PMCID: PMC12048306 DOI: 10.1002/ctm2.70327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Engineered extracellular vesicles (EVs) are emerging as a highly potential platform for targeted drug delivery in cancer therapy. Although intravenous injection is commonly used in EV treatment, there is growing interest in using microneedles (MNs) for transdermal EV delivery; however, comprehensive studies comparing the tissue distribution, safety and antitumour efficacy of these two approaches for delivering engineered EVs remain scarce. METHODS We used EVs derived from umbilical cord mesenchymal stem cells, modified with phospholipid‒polyethylene glycol‒N-hydroxysuccinimide and conjugated with CD38 peptides (CD38-EVs), to target myeloma cells that highly express CD38 antigen, and tested their safety and antitumour efficacy in mice with subcutaneous plasmacytoma, administrated via dissolvable transdermal MNs or intravenous injection. Flow cytometry, immunofluorescence and fluorescence molecular projection imaging analysis were employed to evaluate the distribution of CD38-EVs at the cellular level and within living systems. Additionally, histopathological analysis and biochemical analyses were conducted to assess the antitumour effects and safety of CD38-EVs loaded with doxorubicin (CD38-EVs-Dox). RESULTS Compared to standard EVs, CD38-EVs exhibited enhanced uptake by CD38high tumour cells and reduced uptake by CD38-negative non-tumour cells in vitro. In plasmacytoma NOD/SCID mouse models, CD38-EVs encapsulated within MNs (CD38-EVsMNs) effectively targeted the tumour cells much more than the standard EVs encapsulated within MNs (EVsMNs) and CD38-EVs intravenously administrated (CD38-EVsi.v), with reduced distribution to the lungs and spleen. Additionally, CD38-EVs-Dox induced significantly greater cytotoxicity against the tumour cells than EVs-Dox in vitro, and CD38-EVs-DoxMNs significantly reduced tumour burden compared to both EVs-DoxMNs and CD38-EVs-Doxi.v, while maintaining favourable safety profiles. CONCLUSIONS CD38-EVs-DoxMNs have superior efficacy and safety in treating plasmacytoma mice, compared to CD38-EVs-Doxi.v, providing novel insights into the potential of MNs as a platform for delivering targeted engineered EVs in tumour therapy. HIGHLIGHTS Enhanced tumor targeting: CD38-modified EVs (CD38-EVs) showed increased uptake by CD38high tumor cells while reducing uptake by CD38-negative non-tumor cells. Optimized delivery: MN-loaded CD38-EVs targeted tumors more effectively than MN-loaded EVs and intravenously injected CD38-EVs, with lower lung and spleen accumulation. Superior antitumor efficacy: MN-delivered CD38-EVs-Dox significantly suppressed tumor growth, outperforming intravenous CD38-EVs-Dox and MN-delivered EVs-Dox.
Collapse
Affiliation(s)
- Yulin Cao
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Xuan Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Di Wu
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yuxuan Jiang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhanChina
- MoE Key Laboratory for Biomedical Photonics, Department of Biomedical EngineeringHuazhong University of Science and TechnologyWuhanChina
| | - Yali Yu
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Shan Wang
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Wenlan Chen
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yaoying Long
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Liuyue Xu
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jiao Qu
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Bianlei Yang
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Blal Chakhabi
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Hongxiang Wang
- Department of HematologyThe Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yong Deng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhanChina
- MoE Key Laboratory for Biomedical Photonics, Department of Biomedical EngineeringHuazhong University of Science and TechnologyWuhanChina
| | - Lei Chen
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhichao Chen
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Qiubai Li
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Hubei Engineering Research Center for Application of Extracellular VesicleHubei University of Science and TechnologyXianningChina
| |
Collapse
|
11
|
Bhagat N, Nalawala Z, Patel J, Das D, Baldha R, Sarolia J, Rathod S. Self-Assembled systems for Nose-to-Brain delivery of Temozolamide (TMZ) in brain tumor therapy. Int J Pharm 2025; 675:125540. [PMID: 40174811 DOI: 10.1016/j.ijpharm.2025.125540] [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: 11/21/2024] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Glioblastoma multiforme (GBM) is an aggressive and highly invasive primary brain tumor with poor prognosis and resistance to conventional therapies. The therapeutic efficacy of existing treatments is significantly hampered by the presence of the blood-brain barrier (BBB), tumor heterogeneity, and intrinsic drug resistance mechanisms. Temozolomide (TMZ), the standard chemotherapeutic agent for GBM, suffers from low bioavailability, rapid systemic clearance, and enzymatic degradation, limiting its clinical success. This review highlights the potential of self-assembled nanocarrier-based drug delivery systems for enhancing the therapeutic index of TMZ through intranasal administration, which provides a direct and non-invasive route to the brain, circumventing the BBB and improving central nervous system (CNS) drug bioavailability. Self-assembled systems are highly customizable, allowing for precise control over particle size, surface charge, and release profiles, which can be tailored to improve the penetration and retention of TMZ in the brain. We comprehensively discuss recent advancements in polymeric nanoparticles, liposomes, micelles, niosomes, and solid lipid nanoparticles, emphasizing their physicochemical properties, pharmacokinetics, and mechanisms of targeted drug release. Additionally, we explore molecular and oxidative stress-related pathways contributing to GBM progression and TMZ resistance. Emerging research suggests that nanocarrier-based intranasal delivery of TMZ enhances drug stability, prolongs brain retention time, and minimizes systemic toxicity, offering a promising avenue for improving GBM treatment outcomes.
Collapse
Affiliation(s)
- Nishank Bhagat
- Parul Institute of Pharmacy & Research, Parul University, Wagodia 391760
| | - Zainab Nalawala
- Parul Institute of Pharmacy & Research, Parul University, Wagodia 391760
| | - Jemini Patel
- Parul Institute of Pharmacy & Research, Parul University, Wagodia 391760
| | - Diponkar Das
- Parul Institute of Pharmacy & Research, Parul University, Wagodia 391760
| | - Raj Baldha
- Parul Institute of Pharmacy & Research, Parul University, Wagodia 391760.
| | | | - Sachin Rathod
- Parul Institute of Pharmacy & Research, Parul University, Wagodia 391760; Maliba Pharmacy College, Maliba Campus, 394350.
| |
Collapse
|
12
|
Bolze H, Mc Carogher K, Kuhn S. Microfluidic generation of nanoparticles using standing wave induced ultrasonic spray drying. NANOSCALE ADVANCES 2025; 7:2568-2574. [PMID: 40092060 PMCID: PMC11905916 DOI: 10.1039/d4na01012d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Spray drying is a well-established process for generating particles for various applications, including pharmaceuticals. In this process, atomization plays a crucial role by defining the size of the droplets and, consequently, particle size. While ultrasound is commonly used to enhance atomization by reducing droplet size, a novel approach has been introduced that utilizes plug flow to generate plugs resonating with an applied ultrasound frequency, triggering surface atomization. This study investigates the applicability of this method for microfluidic atomization and spray drying, particular for pharmaceutical carrier particles. The generated droplets exhibit a size of 7.24 μm and a PDI of 0.18, indicating a monodisperse distribution. The droplets are produced in discrete burst events, enabling an energy-efficient pulsed process with an applied power of less than 1 W. This approach successfully generates lipid nanoparticles with an average size of 140 nm, underscoring its potential for nanoparticle production.
Collapse
Affiliation(s)
- Holger Bolze
- KU Leuven, Department of Chemical Engineering Celestijnenlaan 200F 3001 Leuven Belgium
- Institut für Medizintechnik, Otto von Guericke Universität Magdeburg Universitätsplatz 2 39106 Magdeburg Germany
| | - Keiran Mc Carogher
- KU Leuven, Department of Chemical Engineering Celestijnenlaan 200F 3001 Leuven Belgium
| | - Simon Kuhn
- KU Leuven, Department of Chemical Engineering Celestijnenlaan 200F 3001 Leuven Belgium
| |
Collapse
|
13
|
Sun J, Li HL, Zhou WJ, Ma ZX, Huang XP, Li C. Current status and recent progress of nanomaterials in transcatheter arterial chemoembolization therapy for hepatocellular carcinoma. World J Clin Oncol 2025; 16:104435. [PMID: 40290691 PMCID: PMC12019268 DOI: 10.5306/wjco.v16.i4.104435] [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: 12/23/2024] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common cancers worldwide. Transcatheter arterial chemoembolization has become a common treatment modality for some patients with unresectable advanced HCC. Since the introduction of nanomaterials in 1974, their use in various fields has evolved rapidly. In medical applications, nanomaterials can serve as carriers for the delivery of chemotherapeutic drugs to tumour tissues. Additionally, nanomaterials have potential for in vivo tumour imaging. This article covers the properties and uses of several kinds of nanomaterials, focusing on their use in transcatheter arterial chemoembolization for HCC treatment. This paper also discusses the limitations currently associated with the use of nanomaterials.
Collapse
Affiliation(s)
- Jia Sun
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Hai-Liang Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Wen-Jun Zhou
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Zeng-Xin Ma
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Xiao-Pei Huang
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Cheng Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| |
Collapse
|
14
|
Petty RM, Rangan RS, Curry S, Brooks CD, Sabnis N, Clark AF, Lacko AG, Krishnamoorthy RR. Biodistribution of Reconstituted High-Density Lipoprotein Nanoparticles for Targeted Delivery to Retinal Ganglion Cells. J Ocul Pharmacol Ther 2025. [PMID: 40248840 DOI: 10.1089/jop.2024.0191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025] Open
Abstract
Purpose: Nanoparticle-based drug delivery systems offer a promising approach for overcoming the challenges of ocular drug delivery. Our study evaluated the biodistribution and potential targeting of reconstituted high-density lipoprotein nanoparticles (rHDL NPs) loaded with near-infrared dye IR780 to retinal ganglion cells (RGCs) and optic nerve head astrocytes (ONHAs) as a model for neuroprotective drug delivery in glaucoma. Methods: A stable rHDL-payload complex was formulated using IR780, phosphatidylcholine, and apolipoprotein A-I (Apo A-I) by using a novel preparation method. Fluorescent rHDL (rHDL-IR780) was assessed for cellular uptake in primary human ONHAs in vitro, whereas scavenger receptor class B1 (SR-B1) expression was confirmed by Western blot. Receptor-mediated uptake was examined by SR-B1 receptor blocking. Ex vivo biodistribution was evaluated by intravitreal injection of rHDL into postmortem human donor eyes. Results: Spectroscopic analysis confirmed IR780 encapsulation in rHDL NPs. Blocking SR-B1 receptors significantly reduced IR780 uptake by ONHAs, supporting an SR-B1-mediated delivery mechanism, in addition to confirming SR-B1 expression in human retinal lysates. In ex vivo experiments, 4 h postinjection, IR780 localized in the retinal nerve fiber and ganglion cell layers. By 24 h, IR780 penetrated deeper retinal layers, achieving RGC uptake. Conclusions: Our findings demonstrate that rHDL NPs facilitate targeted delivery to retinal tissues through an Apo A-I/SR-B1 pathway, overcoming ocular barriers to reach RGCs. This study supports the potential of rHDL NPs as a platform for neuroprotective drug delivery to treat glaucoma, enhancing both pharmacokinetics and targeted cellular uptake.
Collapse
Affiliation(s)
- R Max Petty
- Department of Pharmacology and Neuroscience, College of Biomedical and Translational Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
- North Texas Eye Research Institute, Fort Worth, Texas, USA
| | - Rajiv S Rangan
- Department of Pharmacology and Neuroscience, College of Biomedical and Translational Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
- North Texas Eye Research Institute, Fort Worth, Texas, USA
| | - Stacy Curry
- North Texas Eye Research Institute, Fort Worth, Texas, USA
| | - Calvin D Brooks
- Department of Pharmacology and Neuroscience, College of Biomedical and Translational Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
- North Texas Eye Research Institute, Fort Worth, Texas, USA
| | - Nirupama Sabnis
- Department of Microbiology, Immunology and Genetics, College of Biomedical and Translational Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Abbot F Clark
- Department of Pharmacology and Neuroscience, College of Biomedical and Translational Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
- North Texas Eye Research Institute, Fort Worth, Texas, USA
| | - Andras G Lacko
- Department of Physiology and Anatomy, College of Biomedical and Translational Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Raghu R Krishnamoorthy
- Department of Pharmacology and Neuroscience, College of Biomedical and Translational Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
- North Texas Eye Research Institute, Fort Worth, Texas, USA
| |
Collapse
|
15
|
Obeidat WM, Lahlouh IK. Chitosan Nanoparticles: Approaches to Preparation, Key Properties, Drug Delivery Systems, and Developments in Therapeutic Efficacy. AAPS PharmSciTech 2025; 26:108. [PMID: 40244367 DOI: 10.1208/s12249-025-03100-z] [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: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
The integration of nanotechnology into drug delivery systems holds great promise for enhancing pharmaceutical effectiveness. This approach enables precise targeting, controlled release, improved patient compliance, reduced side effects, and increased bioavailability. Nanoparticles are vital for transporting biomolecules-such as proteins, enzymes, genes, and vaccines-through various administration routes, including oral, intranasal, vaginal, buccal, and pulmonary. Among biodegradable polymers, chitosan, a linear polysaccharide derived from chitin, stands out due to its biocompatibility, safety, biodegradability, mucoadhesive properties, and ability to enhance permeation. Its cationic nature supports strong molecular interactions and provides antimicrobial, anti-inflammatory, and hemostatic benefits. However, its solubility, influenced by pH and ionic sensitivity, poses challenges requiring effective solutions. This review explores chitosan, its modified derivatives and chitosan nanoparticles mainly, focusing on nanoparticles physicochemical properties, drug release mechanisms, preparation methods, and factors affecting their mean hydrodynamic diameter (particle size). It highlights their application in drug delivery systems and disease treatments across various routes. Key considerations include drug loading capacity, zeta potential, and stability, alongside the impact of molecular weight, degree of deacetylation, and drug solubility on nanoparticle properties. Recent advancements and studies underscore chitosan's potential, emphasizing its modified derivatives'versatility in improving therapeutic outcomes.
Collapse
Affiliation(s)
- Wasfy M Obeidat
- Jordan University of Science and Technology, 3030, Irbid, 22110, Jordan.
| | - Ishraq K Lahlouh
- Jordan University of Science and Technology, 3030, Irbid, 22110, Jordan
| |
Collapse
|
16
|
Ghosh B, McCarley RL. Redox-responsive liposomes aimed at nitroreductase for contents release. J Liposome Res 2025:1-10. [PMID: 40241482 DOI: 10.1080/08982104.2025.2490537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
Here, we report a novel stimuli-responsive N-DOPE liposome where the redox-active 4-nitrobenzyl formate head group of liposomes would respond with respect to the presence of nitroreductase present in the environment of tumor tissues to release the payload. Our main emphasis is related to the construction of redox-sensitive liposomes that would function as the liposomal drug carriers to malignant tumors. Our N-DOPE liposome contains a nitro group (-NO₂) in the modified lipid, and we expect the reduction of the nitro group (-NO₂) to amine (-NH₂) would release the calcein (drug) through the 1,6 elimination as per our hypothesis. But we found no release after waiting for almost 20 hours with the use of Na₂S₂O₄, nitroreductase (NTR) and changes of different external environmental conditions, i.e. temperature, aerobic and anaerobic, etc. due to the formation of an azo (R-N = N-R) bond that stops the complete reduction of (-NO2) all the way down to form amine (-NH2) to stop releasing the payloads. However, adding an organic group containing nitro during the reduction process with the Na₂S₂O₄ resulted in a 45% release of liposomal content. A detailed study & explanation of the formation of azo bond in our N-DOPE liposome has been shown in a stepwise manner in through various spectroscopic methods, and we have discussed future directions.
Collapse
Affiliation(s)
- Brajadulal Ghosh
- Department of Chemistry, Louisiana State University, Baton Rouge, LA, USA
| | - Robin L McCarley
- Department of Chemistry, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
17
|
Yin H, Gao Y, Zhou X, Chen X, Hu Z, Zhang L, Li L, Wang C. Oral multistage nanomedicine for synergistic chemo/chemodynamic/near-infrared-II photothermal cancer therapy. J Colloid Interface Sci 2025; 684:244-253. [PMID: 39793431 DOI: 10.1016/j.jcis.2025.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
The oral administration of drugs for cancer therapy can maintain optimal blood concentrations, is biologically safe and simple, and is preferred by many patients. However, the complex lumen environment, mucus layer, and intestinal epithelial cells are biological barriers that hinder the absorption of orally administered drugs. In this study, sea urchin-like manganese-doped copper selenide nanoparticles (Mn-Cu2-xSe NPs) were designed using an anion exchange method and coated with calcium alginate and chitosan (AC) to form Mn-Cu2-xSe@AC capsules. The pH-responsive swelling behavior of the AC protective layer aided doxorubicin (DOX)-loaded Mn-Cu2-xSe NPs in overcoming multiple biological barriers, maintained their stability in gastric acid, and facilitated the release of the NPs in the small intestine. The intestinal epithelial cell permeability of DOX/Mn-Cu2-xSe NPs was confirmed using a monolayer absorption model involving Caco-2 human epithelial cells. The released DOX/Mn-Cu2-xSe NPs smoothly passed through the mucus layer, and were absorbed by intestinal epithelial cells. In mice, the NPs circulated in the blood and passively targeted the tumors through blood circulation by enhancing the permeability and retention effect to achieve significant tumor suppression and reduce damage to normal tissues. In addition, the unique sea urchin-like morphology of Mn-Cu2-xSe NPs enhanced the absorption in the near-infrared-II (NIR-II) window for photothermal therapy, realized the near-infrared-stimulated response release of DOX for increased chemotherapy, and promoted the Fenton-like effect because of the doping of manganese ions for chemodynamic therapy. These effects could permit the development of various synergistic cancer treatments. The use of DOX/Mn-Cu2-xSe@AC capsules as a multistage oral drug delivery system may overcome the sequential absorption barriers that currently hinder chemotherapy, chemodynamic, and photothermal therapies.
Collapse
Affiliation(s)
- Hongxiu Yin
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Ying Gao
- Department of Stomatology, No. 964 Hospital, Changchun, Jilin 130021, PR China
| | - Xue Zhou
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Xiangjun Chen
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, Yantai 264003, PR China
| | - Zhichao Hu
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Lingyu Zhang
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China.
| | - Lu Li
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Chungang Wang
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China.
| |
Collapse
|
18
|
Samari M, Kashanian S, Zinadini S, Derakhshankhah H. Enhanced delivery of azithromycin using asymmetric polyethersulfone membrane modified with KIT-6 mesoporous material: Optimization and mechanistic studies. Eur J Pharm Sci 2025; 207:107038. [PMID: 39933630 DOI: 10.1016/j.ejps.2025.107038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/13/2025]
Abstract
This study presents the development of a novel drug delivery system designed for improving the release profile and sustained delivery of azithromycin (AZI), particularly aimed at applications requiring localized infection control and improved tissue compatibility. The system employs an asymmetric polyethersulfone (PES) membrane modified with KIT-6 mesoporous material, offering improved drug release performance and biocompatibility over conventional delivery platforms. Membrane optimization was achieved by systematically varying parameters such as thickness (150-600 µm), drug concentration (500-1500 mg/L), polymer content (13-21 % PES), pore maker percentage (0-4 % polyvinylpyrrolidone), and KIT-6 modifier percentage (0.5-2 %). Characterization included scanning electron microscopy, water contact angle measurements, porosity, tensile strength evaluation, and comprehensive bioactivity testing (cytotoxicity, antimicrobial efficacy, blood compatibility, and a novel tissue integrity assay). The optimized formulation (17 % PES, 2 % PVP, 1 % KIT-6) achieved a controlled and sustained release profile with improved drug availability (464 mg/L) compared to unmodified membranes (252 mg/L), with a sustained release profile governed by the Higuchi model. Additionally, the membrane demonstrated superior biocompatibility (-90 % cell viability, low hemolysis at 1.2 %) and preserved tissue integrity better than unmodified counterparts, as evidenced by in vitro and ex vivo studies. Notably, the system showed robust reusability over prolonged use, indicating its potential as an effective, sustainable, and biocompatible solution for localized AZI delivery. These advantages position this system as a promising alternative for medical applications requiring precise drug release and minimal tissue disruption.
Collapse
Affiliation(s)
- Mahya Samari
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran
| | - Soheila Kashanian
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran; Nanobiotechnology Department, Faculty of Innovative Science and Technology, Razi University, Kermanshah, Iran.
| | - Sirus Zinadini
- Department of Applied Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran; Environmental Research Center (ERC), Razi University, Kermanshah, Iran
| | - Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Ohshima K, Mizomichi K, Ohsaki S, Nakamura H, Watano S. Influence of Solvents on Drug Loading Capacity of Metal-Organic Frameworks Focusing on Solvent Dipole Moment. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:8117-8124. [PMID: 40100142 DOI: 10.1021/acs.langmuir.4c04896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
The application of metal-organic frameworks (MOFs) as drug delivery systems with a high drug-loading capacity and targeted delivery is advancing rapidly. This study is the first to elucidate the mechanism of drug-loading in MOFs. It focused on the crucial role of solvents in drug-loading capacity. Ibuprofen, which is widely used as a nonsteroidal antiflammatory drug, was selected as a model drug. The drug-loading capacities of zeolitic imidazolate framework-8 (ZIF-8) and Universitetet i Oslo-66-NH2 (UiO-66-NH2) were investigated in various solvents. For ZIF-8, an increase in the solvent dipole moment corresponded to an increase in the drug-loading capacity. Intriguingly, the converse trend was observed for UiO-66-NH2. Therein, a decrease in the solvent dipole moment caused an increase in the drug-loading. These observations indicated that the solvent dipole moment plays a critical role in the drug-loading mechanism of the MOFs. Furthermore, Raman spectroscopy in the solvents with different polarities revealed significant variations in the molecular vibrations of ZIF-8 and UiO-66-NH2. It was indicated that in both the MOFs, the drug-loading amount increased in the solvents when the molecular vibrations of the MOF were constrained. This study revealed that the solvent plays a crucial role in the drug-loading in MOFs, and the polarity of the solvents contributes significantly to the molecular vibration of MOFs during drug-loading, thereby affecting the drug-loading capacity.
Collapse
Affiliation(s)
- Kazuki Ohshima
- Department of Chemical Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Keisuke Mizomichi
- Analytical & Testing Technology Department, Horiba Techno Service, Co., Ltd., Miyanohigashi-cho, Kisshoin Minami-ku, Kyoto 601-8305, Japan
| | - Shuji Ohsaki
- Department of Chemical Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Hideya Nakamura
- Department of Chemical Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Satoru Watano
- Department of Chemical Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
20
|
Zhu W, Zhao Z, Yuwen W, Qu L, Duan Z, Zhu C, Fan D. Chondrocalcin: Insights into its regulation and multi-function in cartilage and bone. Differentiation 2025; 143:100861. [PMID: 40157027 DOI: 10.1016/j.diff.2025.100861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Type Ⅱ collagen (COLⅡ) is the primary constituent of the cartilage matrix, specifically present in vitreous bodies, cartilage, bone, and other skeletal elements. Therefore, the normal expression of COLⅡ is crucial for the normal development, linear growth, mechanical properties, and self-repairing ability of cartilage. Chondrocalcin, the C-propeptide of type Ⅱ procollagen, is not only a marker of COLⅡ synthesis but also one of the most abundant polypeptides in cartilage. This work examines the pivotal role of chondrocalcin in the synthesis of COLⅡ, comprehensively examining its regulation and multi-functions in cartilage and bone related diseases. Our findings suggest that mutations in the chondrocalcin-encoding domain of COL2A1 affect cartilage and bone development in clinical conditions.
Collapse
Affiliation(s)
- Wensha Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Zilong Zhao
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Weigang Yuwen
- Xi 'an Giant Biotechnology Co., Ltd., Xi'an, 710069, China
| | - Linlin Qu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an, 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
21
|
Zhang X, Zhuo J, Wang D, Zhu X. Supramolecular Polymers for Drug Delivery. Chemistry 2025; 31:e202404617. [PMID: 39961052 DOI: 10.1002/chem.202404617] [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: 12/15/2024] [Indexed: 03/21/2025]
Abstract
Supramolecular polymers are constructed through highly reversible and directionally specific non-covalent interactions between monomer units. This unique feature enables supramolecular polymers to undergo controlled structural reconfiguration and functional transformation in response to external stimuli, imparting them with high environmental responsiveness and self-healing properties. In particular, supramolecular polymers exhibit several specific advantages compared to conventional polymers, such as inherent degradability, the ease of preparation and the incorporation of functional units, and smart responsiveness to various biological stimuli. These characters make supramolecular polymers promising candidates for intelligent drug delivery systems in complex biological environments. In this review, we comprehensively summarize the latest developments and representative achievements of supramolecular polymers in drug delivery fields, focusing primarily on the design and synthesis, the properties and functionalities, and the practical applications of supramolecular polymers in small molecule drug delivery, gene therapy, and protein delivery. Finally, we highlight future research directions, focusing on multifunctionality, adaptability, and personalized therapy. We focus on recent studies that address key challenges in the field, providing rational polymer design, important properties, functionality, and understanding delivery strategies. These developments are expected to advance supramolecular polymers as new platforms of intelligent drug delivery systems, offering innovative solutions for the treatment of complex diseases.
Collapse
Affiliation(s)
- Xinyue Zhang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jiaxin Zhuo
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| | - Dali Wang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
22
|
Jeong HS, Choi Y, Kim DW, Je JY, Lee SJ, Choi CH. Multicompartmental Hydrogel Microspheres with a Concentric Thin Oil Layer: Protecting and Targeting Therapeutic Agents for Inflammatory Bowel Disease. ACS APPLIED BIO MATERIALS 2025; 8:2251-2262. [PMID: 40030207 DOI: 10.1021/acsabm.4c01763] [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: 03/18/2025]
Abstract
Although oral delivery of therapeutic agents offers numerous benefits, its application is limited due to the digestive tract's harsh conditions (e.g., strong acidity and high osmolarity), which impair activity and create challenges in achieving targeted release into the intestine. Here, we present multicompartmental hydrogel microspheres equipped with a concentric oil layer to significantly enhance the oral drug delivery efficiency for treating inflammatory bowel disease (IBD). These microspheres are created through the utilization of triple-emulsion droplets, featuring intermediate oil layers that distinctively separate two prepolymer phases, allowing us to fine-tune the composition of each compartment through a tailored polymerization strategy. We demonstrate that the oil layer can protect the encapsulated material by preventing exposure to the acidic environment of the stomach during the digestive process. Unlike aqueous core capsules, the core is composed of hydrogel, which provides high stability even under high osmolarity conditions in the stomach. By fine-tuning the shell's composition, we can develop capsules that release selectively in response to the gut's pH conditions. We demonstrate the system's efficacy by preserving the anti-inflammatory activities of 5-aminosalicylic acid (5-ASA) and Lys-Pro-Val (KPV) under stomach conditions and maintaining their therapeutic effects on colonic epithelial cell migration and proliferation.
Collapse
Affiliation(s)
- Hye-Seon Jeong
- School of Chemical Engineering, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Yoon Choi
- Division of Cosmetic Science and Technology, Daegu Haany University, 1 Haanydaero, Gyeongsan, Gyeongbuk 38610, Republic of Korea
| | - Do-Wan Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Jae-Young Je
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Sei-Jung Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Chang-Hyung Choi
- School of Chemical Engineering, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| |
Collapse
|
23
|
Cornell H, Sose AT, Ilic S, Chinnabattigalla S, Lidman NE, Oldmixon CM, Yang X, Deshmukh SA, Morris AJ. Photoactivated Multivariate Metal-Organic Frameworks for On-Demand Drug Release: The Role of Host-Guest Interactions. J Am Chem Soc 2025; 147:7423-7432. [PMID: 39992360 PMCID: PMC11887053 DOI: 10.1021/jacs.4c15222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/25/2025]
Abstract
The development of smart drug delivery vehicles capable of controlled release upon application of an external stimulus is of paramount interest for the next generation of personalized medicine. Herein, we report a series of six multivariate (MTV) MOFs capable of visible light-activated drug delivery. The drug loading capacity and release rates were systematically tuned through variation of the linker ratio between 4,4'-azobenzene dicarboxylic acid (H2ABDA) and 4,4'-(diazene-1,2-diyl)bis(3,5-difluorobenzoic acid) (H2ABDA(3,5-F)). The drug loading capacity, dictated by host-guest interactions, was thoroughly explored via a combined experimental and computational approach using two model drug or drug-like molecules, 5-fluorouracil (5-FU) and Nile Red. Notably, the loading capacity for 5-FU follows a "Goldilocks" profile with a maximum loading at 33% H2ABDA(3,5-F) content. Computational results confirm the existence of a cooperative ligand environment that promotes strong, preferential binding at the tetrahedral/octahedral pore window formed between two H2ABDA and one H2ABDA(3,5-F). Thus, the MTV approach enhanced capacity over the native 100% H2ABDA(3,5-F) and 0% H2ABDA(3,5-F) MOFs. In addition to increased loading, the rate of cargo release upon green light excitation also increased as the percentage of H2ABDA(3,5-F) in the MOF was raised, reaching a maximum release rate of 0.9 ± 0.1% of total cargo per minute for the MOF containing 100% H2ABDA(3,5-F) MOF. The results highlight the promise of MTV MOF design for optimizing drug delivery vehicles with relevant payloads and patient-dictated dosing.
Collapse
Affiliation(s)
- Hannah
D. Cornell
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Abhishek T. Sose
- Department
of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Stefan Ilic
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | | | - Naomei E. Lidman
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Colleen M. Oldmixon
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Xiaozhou Yang
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Sanket A. Deshmukh
- Department
of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Amanda J. Morris
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
24
|
Liu Z, Koseki Y, Suzuki R, Dao ATN, Kasai H. Sustained Drug Release from Dual-Responsive Hydrogels for Local Cancer Chemo-Photothermal Therapy. Macromol Biosci 2025; 25:e2400413. [PMID: 39565793 PMCID: PMC11904390 DOI: 10.1002/mabi.202400413] [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: 08/23/2024] [Revised: 11/05/2024] [Indexed: 11/22/2024]
Abstract
As an exceptional carrier for localized drug delivery to tumors, hydrogels can achieve prolonged drug release through careful design and adjustments, effectively targeting cancer cells and minimizing side effects. This study investigates a novel dual-responsive hydrogel system designed for the delivery of nanomedicines, focusing on drug release and the local antitumor efficacy of SN-38-cholesterol nanoparticles (SN-38-chol NPs) and polydopamine NPs (PDA NPs)/poly(n-isopropylacrylamide) (pNIPAM) hydrogels. By combining the thermosensitive properties of pNIPAM with the near-infrared (NIR) responsiveness of PDA NPs, the hydrogel aims to enhance on-demand drug release. SN-38-chol NPs, known for their stability and small size, are incorporated into the hydrogel to improve drug release dynamics. The investigation reveals a drug release cycle of over three weeks, maintaining sensitivity to both temperature and NIR light for controlled drug release. In vivo studies demonstrate the high tumor growth inhibition performance of the system after photothermal treatment induced by 808 nm NIR light. These results suggest that the drug-carrying hydrogel system holds promise for diverse applications in chemical and physical therapies, including the treatment of malignant wounds, post-surgery wound healing, and direct tumor treatment. This study establishes the potential of SN-38-chol NPs and PDA NPs/pNIPAM hydrogels as effective platforms for chemo-phototherapy.
Collapse
Affiliation(s)
- Zhixiang Liu
- Institute of Multidisciplinary Research for Advanced MaterialsTohoku University2‐1‐1 Katahira, AobaSendaiMiyagi980–8577Japan
| | - Yoshitaka Koseki
- Institute of Multidisciplinary Research for Advanced MaterialsTohoku University2‐1‐1 Katahira, AobaSendaiMiyagi980–8577Japan
| | - Ryuju Suzuki
- National Institute of TechnologySendai College4‐16‐1 Ayashi, AobaSendaiMiyagi989–3128Japan
| | - Anh Thi Ngoc Dao
- Graduate School of Integrated Science and TechnologyNagasaki University1–14 BunkyoNagasakiNagasaki852–8521Japan
| | - Hitoshi Kasai
- Institute of Multidisciplinary Research for Advanced MaterialsTohoku University2‐1‐1 Katahira, AobaSendaiMiyagi980–8577Japan
| |
Collapse
|
25
|
Deng D, Zhang R, Wu J, Wang L, Zhang Q, Zhang J, Tan Y, Chen K, Li Y. A kidney protection nanoparticle based on Alpinia oxyphylla fructus polysaccharide by modulating macrophage polarization. Int J Biol Macromol 2025; 292:139367. [PMID: 39743078 DOI: 10.1016/j.ijbiomac.2024.139367] [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: 10/08/2024] [Revised: 12/15/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
The use of natural polysaccharides from traditional Chinese medicine as carrier materials has great potentiality in drug delivery. Nootkatone (NKT) demonstrated good pharmacological activity in treating kidney injury, but its solubility and bioavailability are not very good which may affect the effectiveness of its therapeutic effect. Alpinia oxyphylla fructus polysaccharide (AOP), as a plant polysaccharide, has multiple pharmacological activities and may help to provide synergy for NKT. Therefore, AOP nanoparticles loaded with NKT (AOP-NKT NPs) were prepared for the prevention of acute kidney injury in this study. The sizes of AOP-NKT NPs are 291.60 ± 3.73 nm, and the Zeta potential values are 35.2 ± 0.65 mV. The nanoparticles exhibited excellent stability in pH, NaCl solution, temperature, and storage. The nanoparticles also improved the solubility and oral bioavailability of NKT. In biocompatibility experiments, AOP-NKT NPs showed lower macrophage toxicity than NKT, and the nanoparticles had good blood compatibility and in vivo biosafety. In vivo, prophylactic administration of this nanoparticle could enhance the ability of NKT in promoting macrophage M2 polarization, reducing renal inflammation and thus improve renal function and repair renal damage. In conclusion, the present study may provide the possibility for AOP as a nano delivery vehicle for renal injury protective drugs.
Collapse
Affiliation(s)
- Dezhi Deng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Rong Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Jiaoxia Wu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Lisha Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Qiaoling Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Junqing Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Yinfeng Tan
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China.
| | - Kai Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China.
| | - Yonghui Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
26
|
Jindal A, Mainuddin, Kumar A, Ratnesh RK, Singh J. Nanotechnology Driven Lipid and Metalloid Based Formulations Targeting Blood-Brain Barrier (3B) for Brain Tumor. Indian J Microbiol 2025; 65:92-119. [PMID: 40371021 PMCID: PMC12069182 DOI: 10.1007/s12088-024-01330-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/08/2024] [Indexed: 05/16/2025] Open
Abstract
The evolution of nanotechnology-driven lipid and metalloid-based nanoformulations has garnered significant attention for developing effective drug delivery systems with position/time precision and efficacy. This study focuses on challenges of blood-brain barrier (BBB) and their pivotal role in drug targeting in chronic diseases such as brain tumors (BTs). These formulations encapsulate therapeutic agents within lipidic matrices, enhancing drug solubility, bioavailability, and targeted delivery. The diverse lipid materials used in these nanoformulations highlight their biocompatibility and versatility, covering a wide range of drugs. Emphasis is placed on metal nanoparticles, liposomes, ethosomes, quantum dots, carbon nanotubes, nanorobots, and micelles. The analysis explores their drug loading, stability, release characteristics, and bioavailability modulation. It also delves into the enhanced-permeability and retention (EPR) effect, crucial for passive targeting of tumors. Recent nanocarrier systems enable better penetration of therapeutic compounds through the BBB, addressing treatment failures in invasive BTs.This review highlights the latest nanotechnology developments and potential therapeutic approaches, serving as a valuable resource for researchers, clinicians, and pharmaceutical scientists.
Collapse
Affiliation(s)
- Amulya Jindal
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh 250005 India
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204 India
| | - Mainuddin
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh 250005 India
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Sector 125, Noida, Uttar Pradesh 201301 India
| | - Anoop Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh 250005 India
| | - Ratneshwar Kumar Ratnesh
- Department of Electronics and Communication Engineering, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh 250005 India
| | - Jay Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005 India
| |
Collapse
|
27
|
Roszkowski S, Durczyńska Z, Szablewska S. Targeted nanodelivery systems for personalized cancer therapy. Rep Pract Oncol Radiother 2025; 29:776-788. [PMID: 40104662 PMCID: PMC11912883 DOI: 10.5603/rpor.103524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 11/12/2024] [Indexed: 03/20/2025] Open
Abstract
Conventional cancer therapies such as chemotherapy face challenges such as poor tumor targeting, systemic toxicity, and drug resistance. Nanotechnology offers solutions through advanced drug delivery systems that preferentially accumulate in tumors while avoiding healthy tissues. Recent innovations have enabled the optimization of engineered nanocarriers for extended circulation and tumor localization via both passive and active targeting mechanisms. Passive accumulation exploits the leaky vasculature of tumors, whereas active strategies use ligands to selectively bind cancer cell receptors. Multifunctional nanoparticles also allow the combination of imaging, multiple therapeutic modalities and on-demand drug release within a single platform. Overall, precisely tailored nanotherapeutics that leverage unique pathophysiological traits of malignancies provide opportunities to overcome the limitations of traditional treatment regimens. This emerging field promises more effective and personalized nanomedicine approaches to detect and treat cancer. The key aspects highlighted in this review include the biological barriers associated with nanoparticles, rational design principles to optimize nanocarrier pharmacokinetics and tumor uptake, passive and active targeting strategies, multifunctionality, and reversal of multidrug resistance.
Collapse
Affiliation(s)
- Szymon Roszkowski
- Division of Biochemistry and Biogerontology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Zofia Durczyńska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Sylwia Szablewska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| |
Collapse
|
28
|
Marzaman AF, Mahfufah U, Fauziah N, Ulum Ar Rahman F, Hidayati N, Hasyim R, Setiawati D, Choiri S, Nuzulia NA, Madani AF, Mir M, Permana AD, Mansjur KQ. Doxycycline-Loaded pH-Sensitive Microparticles as a Potential Site-Specific Drug Delivery System against Periodontitis. ACS OMEGA 2025; 10:5668-5685. [PMID: 39989785 PMCID: PMC11840606 DOI: 10.1021/acsomega.4c08967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025]
Abstract
A significant obstacle to the healing process of periodontitis is the development of bacterial biofilms within the periodontal pockets. The efficacy of bacterial biofilm therapy is often hindered by the inadequate penetration of antibacterial agents and the nonspecific targeting of bacteria. This study proposes a novel strategy involving the utilization of pH-sensitive microparticles (MPs) of doxycycline (DOX) to enhance biofilm penetration and enable targeted delivery of DOX to infection sites associated with periodontitis. The MPs were developed using a double-emulsion technique with poly(d,l-lactide-co-glycolide) and chitosan in a 1:1 ratio. The morphology of DOX-MP exhibits a spherical form with a particle size of 3.54 ± 0.32 μm and a PDI of 0.221 ± 0.02. The DOX-MP also had great encapsulation efficiency (69.43% ± 5.32) and drug loading efficiency (14.81% ± 1.32) with regulated drug release kinetics and accelerated release rates under low-pH conditions. The antimicrobial activity was evaluated against Escherichia coli and Staphylococcus aureus, and the results indicated the absence of any viable bacterial colonies after 18 h at twice the minimum inhibitory concentration value. Hydrogel-based MPs deliver DOX to the periodontal pocket infection site for ease of use. In situ hydrogels used Pluronic F127 and F68 as the main polymer composition and hydroxypropyl methylcellulose as the adhesion polymer. This formulation exhibited a liquid state at room temperature (25 °C) but went through gelation at 36 °C. The formulation also had good mucoadhesive characteristics (42.65 ± 3.53 dyn/cm2) and good drug permeation at acidic pH in Mueller-Hinton Broth media with the addition of E. coli and S. aureus bacteria. Ex vivo antibacterial activity significantly reduced the microbial count, biofilm quantity, and metabolic activity, confirming the desired antibacterial effect. Hence, the utilization of free drugs and DOX-MPs did not exhibit a notable dissimilarity, showing that integrating the drug into the matrix was not hindering its antibacterial efficacy.
Collapse
Affiliation(s)
| | - Ulfah Mahfufah
- Faculty
of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia
| | - Nurul Fauziah
- Faculty
of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia
| | - Fadhlil Ulum Ar Rahman
- Department
of Oral Maxillofacial Radiology, Faculty of Dentistry, Hasanuddin University, Makassar 90245, Indonesia
| | - Nasyrah Hidayati
- Department
of Orthodontic, Faculty of Dentistry, Hasanuddin
University, Makassar 90245, Indonesia
| | - Rafikah Hasyim
- Department
of Oral Biology, Faculty of Dentistry, Hasanuddin
University, Makassar 90245, Indonesia
| | - Dian Setiawati
- Department
of Periodontology, Faculty of Dentistry, Hasanuddin University, Makassar 90245, Indonesia
| | - Syaiful Choiri
- Faculty
of
Mathematics and Natural Sciences, Sebelas
Maret University, Surakarta 57126, Indonesia
| | - Nur Aisyah Nuzulia
- Faculty
of Mathematics and Natural Sciences, Institute
Pertanian Bogor, Bogor 16680, Indonesia
| | | | - Maria Mir
- Department
of Pharmacy, Iqra University, Islamabad Campus 44000, Pakistan
| | - Andi Dian Permana
- Faculty
of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia
| | - Karima Qurnia Mansjur
- Department
of Orthodontic, Faculty of Dentistry, Hasanuddin
University, Makassar 90245, Indonesia
| |
Collapse
|
29
|
Rahma MN, Suhandi C, Mohammed AFA, El-Rayyes A, Elamin KM, Sulastri E, Wathoni N. The Role and Advancement of Liposomes for Oral Diseases Therapy. Int J Nanomedicine 2025; 20:1865-1880. [PMID: 39975418 PMCID: PMC11837752 DOI: 10.2147/ijn.s492353] [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: 08/20/2024] [Accepted: 11/25/2024] [Indexed: 02/21/2025] Open
Abstract
As many as 48.0% of the global population suffers from disabilities caused by oral conditions. These conditions encompass dental caries, periodontal diseases, oral cancers, and other pathologies affecting the hard and soft tissues of the oral and maxillofacial regions. Topical drug treatments in the oral cavity are often ineffective due to the short contact time, which prevents the drug from reaching optimal concentrations necessary for therapeutic effect. Conventional liposomes have several limitations, including low stability, challenges in long-term storage, and rapid clearance by the reticuloendothelial system (RES). These factors significantly reduce their effectiveness in maintaining sustained drug delivery and achieving desired therapeutic outcomes. To overcome these challenges, advanced drug delivery systems have been developed. Among these systems, liposomes have been extensively explored as nanocarriers in targeted drug delivery systems, particularly in mucosal drug delivery, due to their biocompatibility and degradability, making them promising agents for the treatment of oral diseases. To address these issues, extensive research has been conducted to modify the surface of liposomes, optimizing their efficacy, and understanding their mechanisms of action. This review article discusses the role and recent advancements of liposomes in the treatment of oral diseases, highlighting their potential to revolutionize oral health care through improved drug delivery and therapeutic outcomes.
Collapse
Affiliation(s)
- Maya Nurul Rahma
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Ahmed F A Mohammed
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Ali El-Rayyes
- Center for Scientific Research and Entrepreneurship, Northern Border University, Arar, 73213, Saudi Arabia
| | - Khaled M Elamin
- Graduated School of Pharmaceutical Science, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Evi Sulastri
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Tadulako University, Palu, Central Sulawesi, 94118, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| |
Collapse
|
30
|
Alkatheeri A, Salih S, Kamil N, Alnuaimi S, Abuzar M, Abdelrahman SS. Nano-Radiopharmaceuticals in Colon Cancer: Current Applications, Challenges, and Future Directions. Pharmaceuticals (Basel) 2025; 18:257. [PMID: 40006069 PMCID: PMC11859487 DOI: 10.3390/ph18020257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Colon cancer remains a significant global health challenge; however, the treatment outcome for colon patients can be improved through early detection and effective treatment. Nano-radiopharmaceuticals, combining nanotechnology with radiopharmaceuticals, are emerging as a revolutionary approach in both colon cancer diagnostic imaging and therapy, playing a significant role in the management of colon cancer patients. This review examines the use of nano-radiopharmaceuticals in the diagnosis and treatment of colon cancer, highlighting current applications, challenges, and future directions. Nanocarriers of radionuclides have shown potential in improving cancer treatment, including liposomes, microparticles, nanoparticles, micelles, dendrimers, and hydrogels, which are approved by the FDA. These nanocarriers can deliver targeted drugs into malignant cells without affecting normal cells, reducing side effects. Antibody-guided systemic radionuclide-targeted therapy has shown potential for treating cancer. Novel cancer nanomedicines, like Hensify and 32P BioSilicon, are under clinical development for targeted radiation delivery in percutaneous intratumoral injections. Although using nano-radiopharmaceuticals is a superior technique for diagnosing and treating colon cancer, there are limitations and challenges, such as the unintentional accumulation of nanoparticles in healthy tissues, which leads to toxicity due to biodistribution issues, as well as high manufacturing costs that limit their availability for patients. However, the future direction is moving toward providing more precise radiopharmaceuticals, which is crucial for enhancing the diagnosis and treatment of colon cancer and reducing production costs.
Collapse
Affiliation(s)
- Ajnas Alkatheeri
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates;
| | - Suliman Salih
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates;
- National Cancer Institute, University of Gezira, Wad Madani 2667, Sudan
| | - Noon Kamil
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | - Sara Alnuaimi
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | - Memona Abuzar
- Department of Pharmacy, Fatima College of Health Sciences, Abu Dhabi 3798, United Arab Emirates; (N.K.); (S.A.); (M.A.)
| | | |
Collapse
|
31
|
Elter JK, Sedlák F, Palušák T, Bernardová N, Lobaz V, Tihlaříková E, Neděla V, Šácha P, Hrubý M. Solid Lipid Nanoparticles Coated with Glucosylated poly(2-oxazoline)s: A Supramolecular Toolbox Approach. Biomacromolecules 2025; 26:861-882. [PMID: 39779305 PMCID: PMC11815865 DOI: 10.1021/acs.biomac.4c01052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025]
Abstract
Multifunctional polymers are interesting substances for the formulation of drug molecules that cannot be administered in their pure form due to their pharmacokinetic profiles or side effects. Polymer-drug formulations can enhance pharmacological properties or create tissue specificity by encapsulating the drug into nanocontainers, or stabilizing nanoparticles for drug transport. We present the synthesis of multifunctional poly(2-ethyl-2-oxazoline-co-2-glyco-2-oxazoline)s containing two reactive end groups, and an additional hydrophobic anchor at one end of the molecule. These polymers were successfully used to stabilize (solid) lipid nanoparticles ((S)LNP) consisting of tetradecan-1-ol and cholesterol with their hydrophobic anchor. While the pure polymers interacted with GLUT1-expressing cell lines mainly based on their physicochemical properties, especially via interactions of the hydrophobic anchor with membranous compartments of the cells, LNP-cell interactions hinted toward an influence of the glucosylation on particle-cell interactions. The presented LNP are therefore promising systems for the delivery of drugs into GLUT1-expressing cell lines.
Collapse
Affiliation(s)
- Johanna K. Elter
- Institute
of Macromolecular Chemistry, CAS, Heyrovského nám. 2, Praha 6 162 06, Czech Republic
| | - František Sedlák
- Institute
of Biochemistry and Experimental Oncology, First Faculty of Medicine, U Nemocnice 5, Praha 2 128 53, Czech Republic
| | - Tomáš Palušák
- Institute
of Biochemistry and Experimental Oncology, First Faculty of Medicine, U Nemocnice 5, Praha 2 128 53, Czech Republic
| | - Nicol Bernardová
- Institute
of Biochemistry and Experimental Oncology, First Faculty of Medicine, U Nemocnice 5, Praha 2 128 53, Czech Republic
| | - Volodymyr Lobaz
- Institute
of Macromolecular Chemistry, CAS, Heyrovského nám. 2, Praha 6 162 06, Czech Republic
| | - Eva Tihlaříková
- Institute
of Scientific Instruments, CAS, Královopolská 147, Brno 612 00, Czech Republic
| | - Vilém Neděla
- Institute
of Scientific Instruments, CAS, Královopolská 147, Brno 612 00, Czech Republic
| | - Pavel Šácha
- Institute
of Organic Chemistry and Biochemistry, CAS, Flemingovo nám. 2, Praha 6 166 10, Czech Republic
| | - Martin Hrubý
- Institute
of Macromolecular Chemistry, CAS, Heyrovského nám. 2, Praha 6 162 06, Czech Republic
| |
Collapse
|
32
|
Song X, Ji M, Shu X, Zou L. Drug delivery systems loaded with plant-derived natural products for dental caries prevention and treatment. J Mater Chem B 2025; 13:1920-1934. [PMID: 39791142 DOI: 10.1039/d4tb01924e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Dental caries, driven by dysbiosis in oral flora and acid accumulation, pose a significant threat to oral health. Traditional methods of managing dental biofilms using broad-spectrum antimicrobials and fluoride face limitations such as microbial resistance. Natural products, with their antimicrobial properties, present a promising solution for managing dental caries, yet their clinical application faces significant challenges, including low bioavailability, variable efficacy, and patient resistance due to sensory properties. Advanced drug delivery systems (DDSs) are emerging to address these limitations by enhancing the delivery and effectiveness of natural products. These systems, such as nanoparticles and micelles, aim to enhance drug solubility, stability, and targeted release, leading to increased therapeutic efficacy and decreased side effects. Furthermore, innovative approaches like pH-responsive nanoparticles offer controlled release triggered by the acidic environment of carious lesions. Despite these technological advancements, further validation is necessary for the clinical application of these DDSs.
Collapse
Affiliation(s)
- Xiaowen Song
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mengzhen Ji
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xingyue Shu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
33
|
Tenchov R, Hughes KJ, Ganesan M, Iyer KA, Ralhan K, Lotti Diaz LM, Bird RE, Ivanov JM, Zhou QA. Transforming Medicine: Cutting-Edge Applications of Nanoscale Materials in Drug Delivery. ACS NANO 2025; 19:4011-4038. [PMID: 39823199 PMCID: PMC11803921 DOI: 10.1021/acsnano.4c09566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Since their inception in the early 1960s, the development and use of nanoscale materials have progressed tremendously, and their roles in diverse fields ranging from human health to energy and electronics are undeniable. The application of nanotechnology inventions has revolutionized many aspects of everyday life including various medical applications and specifically drug delivery systems, maximizing the therapeutic efficacy of the contained drugs by means of bioavailability enhancement or minimization of adverse effects. In this review, we utilize the CAS Content Collection, a vast repository of scientific information extracted from journal and patent publications, to analyze trends in nanoscience research relevant to drug delivery in an effort to provide a comprehensive and detailed picture of the use of nanotechnology in this field. We examine the publication landscape in the area to provide insights into current knowledge advances and developments. We review the major classes of nanosized drug delivery systems, their delivery routes, and targeted diseases. We outline the most discussed concepts and assess the advantages of various nanocarriers. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding nanosized drug delivery systems, to outline challenges, and to evaluate growth opportunities. The merit of the review stems from the extensive, wide-ranging coverage of the most up-to-date scientific information, allowing unmatched breadth of landscape analysis and in-depth insights.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Kevin J. Hughes
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Kavita A. Iyer
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Leilani M. Lotti Diaz
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert E. Bird
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Julian M. Ivanov
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
34
|
Wang Z, Wang C, Ji Y, Yang M, Li C, Li M, Yang J, Tang H, Luo X, Hao H, Liu Z, Chen K, Chang Y, Yuan H, Feng L, Xing G, Li J. Magnetically driven bionic nanorobots enhance chemotherapeutic efficacy and the tumor immune response via precise targeting. Innovation (N Y) 2025; 6:100777. [PMID: 39991478 PMCID: PMC11846086 DOI: 10.1016/j.xinn.2024.100777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 12/23/2024] [Indexed: 02/25/2025] Open
Abstract
We developed magnetically driven bionic drug-loaded nanorobots (MDNs) to accurately target tumors and deliver chemotherapy agents using a customized three-dimensional (3D) magnetic manipulation platform (MMP) system to precisely control their movement mode. MDNs were based on polyethylene glycol-modified homogeneous ultrasmall iron oxide nanoparticles (7.02 ± 0.18 nm). Doxorubicin (12% ± 2% [w/w]) was encapsulated in MDNs by an imide bond. MDNs could imitate the movement mode of a school of wild herrings (e.g., re-dispersion/arrangement/vortex/directional movement) to adapt to the changing and complex physiological environment through the 3D MMP system. MDNs overcame blood flow resistance and biological barriers using optimized magnetic driving properties according to in vivo imaging (magnetic resonance imaging and fluorescence) and histopathology. The performance of fabricated MDNs was verified through cells and tumor-bearing mouse models. The MDNs showed high efficiency of drug delivery and targeting at the tumor site (>10-fold), lower toxicity than free doxorubicin (5 mg/kg body weight), activated immune response in the tumor site, and significantly lengthened survival for mice. The synergistic interaction between MDNs and the 3D MMP system underscores the immense potential of this drug delivery system, indicating a potential revolution in the field of tumor chemotherapy.
Collapse
Affiliation(s)
- Zhijie Wang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chutian Wang
- School of Mechanical Engineering and Automation, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China
| | - Ying Ji
- Institute of Textiles and Clothing, School of Fashion and Textiles, Research Institute for Intelligent Wearable Systems, The Hong Kong Polytechnic University, Hong Kong SAR 999077, China
| | - Mingxin Yang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chan Li
- School of Mechanical Engineering and Automation, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China
| | - Mengyao Li
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingru Yang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyu Tang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianwei Luo
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haoyang Hao
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhicai Liu
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kui Chen
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanan Chang
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Yuan
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Feng
- School of Mechanical Engineering and Automation, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China
| | - Gengmei Xing
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juan Li
- Key Laboratory for Biomedical Effects of Nanomaterial and Nanosafety, Institute of High Energy Physics, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
35
|
Vardar US, Konings G, Yang J, Sagis LMC, Bitter JH, Nikiforidis CV. Modifying the interfacial dynamics of oleosome (lipid droplet) membrane using curcumin. J Colloid Interface Sci 2025; 678:1077-1086. [PMID: 39341139 DOI: 10.1016/j.jcis.2024.09.181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024]
Abstract
Cells store energy in lipid droplets, known as oleosomes, which have a neutral lipid core surrounded by a dilatable membrane of phospholipids and proteins. Oleosomes can be loaded with therapeutic lipophilic cargos through their permeable membrane and used as carriers. However, the cargo can also adsorb between the phospholipids and affect the membrane properties. In the present work, we investigated the effect of adsorbed curcumin on the mechanical properties of oleosome membranes using dilatational interfacial rheology (LAOD). The oleosome membrane had a weak-stretchable behavior, while the adsorption of curcumin led to stronger in-plane interactions, which were dependent on curcumin concentration and indicated a glassy-like structure. Our findings showed that adsorbed curcumin molecules can enhance the molecular interactions on the oleosome membrane. This behavior suggests that oleosomes membranes can be modulated by loaded cargo. Understanding cargo and membrane interactions can help to design oleosome-based formulations with tailored mechanical properties for applications.
Collapse
Affiliation(s)
- Umay Sevgi Vardar
- Laboratory of Biobased Chemistry and Technology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Gijs Konings
- Laboratory of Biobased Chemistry and Technology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Jack Yang
- Laboratory of Biobased Chemistry and Technology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands; Laboratory of Physics and Physical Chemistry of Foods, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Leonard M C Sagis
- Laboratory of Physics and Physical Chemistry of Foods, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Johannes H Bitter
- Laboratory of Biobased Chemistry and Technology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Constantinos V Nikiforidis
- Laboratory of Biobased Chemistry and Technology, Wageningen University and Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands.
| |
Collapse
|
36
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
37
|
Liew WJM, Alkaff SA, Leong SY, Yee MZL, Hou HW, Czarny B. Cell Membrane- and Extracellular Vesicle-Coated Chitosan Methacrylate-Tripolyphosphate Nanoparticles for RNA Delivery. Int J Mol Sci 2024; 25:13724. [PMID: 39769486 PMCID: PMC11678704 DOI: 10.3390/ijms252413724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
mRNA-based vaccines against the COVID-19 pandemic have propelled the use of nucleic acids for drug delivery. Conventional lipid-based carriers, such as liposomes and nanolipogels, effectively encapsulate and deliver RNA but are hindered by issues such as premature burst release and immunogenicity. To address these challenges, cell membrane-coated nanoparticles offer a promising alternative. We developed a novel nanoparticle system using chitosan methacrylate-tripolyphosphate (CMATPP), which capitalizes on interactions involving membrane proteins at biointerfaces. Ionic crosslinking between chitosan methacrylate and tripolyphosphate facilitates the formation of nanoparticles amenable to coating with red blood cell (RBC) membranes, extracellular vesicles (EVs), and cell-derived nanovesicles (CDNs). Coating CMATPP nanoparticles with RBC membranes effectively mitigated the initial burst release of encapsulated small interfering RNA (siRNA), sustaining controlled release while preserving membrane proteins. This concept was extended to EVs, where CMATPP nanoparticles and CDNs were incorporated into a microfluidic device and subjected to electroporation to create hybrid CDN-CMATPP nanoparticles. Our findings demonstrate that CMATPP nanoparticles are a robust siRNA delivery system with suppressed burst release and enhanced membrane properties conferred by cell or vesicle membranes. Furthermore, the adaptation of the CDN-CMATPP nanoparticle formation in a microfluidic device suggests its potential for personalized therapies using diverse cell sources and increased throughput via automation. This study underscores the versatility and efficacy of CMATPP nanoparticles in RNA delivery, offering a pathway towards advanced therapeutic strategies that utilize biomimetic principles and microfluidic technologies.
Collapse
Affiliation(s)
- Wen Jie Melvin Liew
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Syed Abdullah Alkaff
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Sheng Yuan Leong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Marin Zhen Lin Yee
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Bertrand Czarny
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| |
Collapse
|
38
|
Bhargava S, Dewangan HK, Deshmukh R. Targeted polymeric primaquine nanoparticles: optimization, evaluation, and in-vivo liver uptake for improved malaria treatment. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:2867-2883. [PMID: 39217616 DOI: 10.1080/09205063.2024.2391225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Primaquine (PQ) is a widely used antimalarial drug, but its high dosage requirements can lead to significant tissue damage and adverse gastrointestinal and hematological effects. Recent studies have shown that nanoformulations can enhance the bioavailability of pharmaceuticals, thereby increasing efficacy, reducing dosing frequency, and minimizing toxicity. In this study, PQ-loaded PLGA nanoparticles (PQ-NPs) were prepared using a modified double emulsion solvent evaporation technique (w/o/w). The PQ-NPs exhibited a mean particle size of 228 ± 2.6 nm, a zeta potential of +27.4 mV, and an encapsulation efficiency of 81.3 ± 3.5%. Scanning electron microscopy (SEM) confirmed their spherical morphology, and the in vitro release profile demonstrated continuous drug release over 72 h. Differential scanning calorimetry (DSC) thermograms indicated that the drug was present in the nanoparticles, with improved physical stability. Fourier-transform infrared spectroscopy (FTIR) analysis showed no interactions between the various substances in the NPs. In vivo studies in Swiss albino mice infected with Plasmodium berghei revealed that the nanoformulated PQ was 20% more effective than the standard oral dose. Biodistribution studies indicated that 80% of the NPs accumulated in the liver, highlighting their potential for targeted drug delivery. This research demonstrates the successful development of a nanomedicine delivery system for antimalarial drugs, offering a promising strategy to enhance treatment efficacy while reducing adverse effects.
Collapse
Affiliation(s)
- Sarvesh Bhargava
- Institute of Pharmaceutical Research (IPR), GLA University, Mathura, Uttar Pradesh, India
- ShriRam College of Pharmacy, National Expressway, Banmore Gwalior, Madhya Pradesh, India
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences (UIPS), Chandigarh University NH-95, Mohali, Punjab, India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research (IPR), GLA University, Mathura, Uttar Pradesh, India
| |
Collapse
|
39
|
Mody DR, Lathiya V, Kolte AP, Biradar V, Langde V. Doxycycline Proniosomal Gel as Local Drug Delivery System in Periodontal Disease: A Vitro Study. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S3227-S3229. [PMID: 39927011 PMCID: PMC11805169 DOI: 10.4103/jpbs.jpbs_726_24] [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: 06/25/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 02/11/2025] Open
Abstract
Background Periodontitis is an inflammatory condition of the oral cavity in which microorganisms play a significant role in the pathogenesis and onset. The host immune system starts as an inflammatory process in response to microbial attack that protects and kills the periodontium. Doxycycline is a semi-synthetic variant of tetracycline that is made from oxytetracycline. Doxycycline inhibits the production of both aerobic and anaerobic microbial protein through its potent activity against both Gram-positive and Gram-negative bacteria. Materials and Methods The preparation method of doxycycline proniosomal gel involved accurately weighing doxycycline and all excipients (12 Batches). Doxycycline was mixed with 1 ml of ethanol, and all the excipients, like surfactant, lecithin, cholesterol, were added in precisely weighed amounts. The characterization of proniosomal gel and determination of drug entrapment efficacy is calculated. Result Doxycycline release from the doxycycline proniosomal gel (Batch F12) was studied using Franz diffusion cell assembly with dialysis membrane for 7 days in phosphate-buffered saline (7.4) indicated sustained release of doxycycline from the formulation. Conclusion Proniosome containing doxycycline may therefore be a locally effective drug delivery method for treating periodontitis. Current in vitro study is mainly focused on preparation of proniosomal gel containing doxycycline as local drug delivery system in periodontal disease.
Collapse
Affiliation(s)
- Dhawal Rajendra Mody
- Associate Professor, Department of Periodontology, Ranjeet Deshmukh Dental College and Research Centre, Nagpur, Maharashtra, India
| | - Vrushali Lathiya
- Associate Professor, Department of Periodontology, Ranjeet Deshmukh Dental College and Research Centre, Nagpur, Maharashtra, India
| | - Abhay P. Kolte
- Dean, Professor and Head, Department of Periodontology, Ranjeet Deshmukh Dental College and Research Centre, Nagpur, Maharashtra, India
| | - Vaishnavi Biradar
- Post Graduate Students, Department of Periodontology, Ranjeet Deshmukh Dental College and Research Centre, Nagpur, Maharashtra, India
| | - Vaishnavi Langde
- Post Graduate Students, Department of Periodontology, Ranjeet Deshmukh Dental College and Research Centre, Nagpur, Maharashtra, India
| |
Collapse
|
40
|
Kamble OS, Chatterjee R, Abishek KG, Chandra J, Alsayari A, Wahab S, Sahebkar A, Kesharwani P, Dandela R. Small molecules targeting mitochondria as an innovative approach to cancer therapy. Cell Signal 2024; 124:111396. [PMID: 39251050 DOI: 10.1016/j.cellsig.2024.111396] [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: 07/16/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Cellular death evasion is a defining characteristic of human malignancies and a significant contributor to therapeutic inefficacy. As a result of oncogenic inhibition of cell death mechanisms, established therapeutic regimens seems to be ineffective. Mitochondria serve as the cellular powerhouses, but they also function as repositories of self-destructive weaponry. Changes in the structure and activities of mitochondria have been consistently documented in cancer cells. In recent years, there has been an increasing focus on using mitochondria as a targeted approach for treating cancer. Considerable attention has been devoted to the development of delivery systems that selectively aim to deliver small molecules called "mitocans" to mitochondria, with the ultimate goal of modulating the physiology of cancer cells. This review summarizes the rationale and mechanism of mitochondrial targeting with small molecules in the treatment of cancer, and their impact on the mitochondria. This paper provides a concise overview of the reasoning and mechanism behind directing treatment towards mitochondria in cancer therapy, with a particular focus on targeting using small molecules. This review also examines diverse small molecule types within each category as potential therapeutic agents for cancer.
Collapse
Affiliation(s)
- Omkar S Kamble
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Rana Chatterjee
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - K G Abishek
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India.
| |
Collapse
|
41
|
Patel S, Salaman SD, Kapoor DU, Yadav R, Sharma S. Latest developments in biomaterial interfaces and drug delivery: challenges, innovations, and future outlook. Z NATURFORSCH C 2024:znc-2024-0208. [PMID: 39566511 DOI: 10.1515/znc-2024-0208] [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: 11/03/2024] [Indexed: 11/22/2024]
Abstract
An ideal drug carrier system should demonstrate optimal payload and release characteristics, thereby ensuring prolonged therapeutic index while minimizing adverse effects. The field of drug delivery has undergone significant advancements, particularly within the last two decades, owing to the revolutionary impact of biomaterials. The use of biomaterials presents significant due to their biocompatibility and biodegradability, which must be addressed in order to achieve effective drug delivery. The properties of the biomaterial and its interface are primarily influenced by their physicochemical attributes, physiological barriers, cellular trafficking, and immunomodulatory effects. By attuning these barriers, regulating the physicochemical properties, and masking the immune system's response, the bio interface can be effectively modulated, leading to the development of innovative supramolecular structures with enhanced effectiveness. With a comprehensive understanding of these technologies, there is a growing demand for repurposing existing drugs for new therapeutic indications within this space. This review aims to provide a substantial body of evidence showcasing the productiveness of biomaterials and their interface in drug delivery, as well as methods for mitigating and modulating barriers and physicochemical properties along with an examination of future prospects in this field.
Collapse
Affiliation(s)
- Saraswati Patel
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602105, Tamil Nadu, India
| | - Samsi D Salaman
- Apollo College of Pharmacy, Mevaloorkuppam, Kanchipuram, 602105, Tamil Nadu, India
| | - Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Sardar Baug, Station Road, 394601 Bardoli, Gujarat, India
| | - Richa Yadav
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O., Rajasthan, 304022, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O., Rajasthan, 304022, India
| |
Collapse
|
42
|
Rejinold NS, Jin GW, Choy JH. Insight into Preventing Global Dengue Spread: Nanoengineered Niclosamide for Viral Infections. NANO LETTERS 2024; 24:14541-14551. [PMID: 39194045 PMCID: PMC11583367 DOI: 10.1021/acs.nanolett.4c02845] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 08/29/2024]
Abstract
Millions of cases of dengue virus (DENV) infection yearly from Aedes mosquitoes stress the need for effective antivirals. No current drug effectively combats dengue efficiently. Transient immunity and severe risks highlight the need for broad-spectrum antivirals targeting all serotypes of DENV. Niclosamide, an antiparasitic, shows promising antiviral activity against the dengue virus, but enhancing its bioavailability is challenging. To overcome this issue and enable niclosamide to address the global dengue problem, nanoengineered niclosamides can be the solution. Not only does it address cost issues but also with its broad-spectrum antiviral effects nanoengineered niclosamide offers hope in addressing the current health crisis associated with DENV and will play a crucial role in combating other arboviruses as well.
Collapse
Affiliation(s)
- N. Sanoj Rejinold
- Intelligent
Nanohybrid Materials Laboratory (INML), College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Institute
of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic
of Korea
| | - Geun-woo Jin
- R&D
Center, Hyundai Bioscience Co. LTD., Seoul 03759, Republic
of Korea
| | - Jin-Ho Choy
- Intelligent
Nanohybrid Materials Laboratory (INML), College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Division
of Natural Sciences, The National Academy
of Sciences, Seoul 06579, Republic of Korea
- Tokyo
Tech World Research Hub Initiative (WRHI), Institute of Innovative
Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| |
Collapse
|
43
|
Strzelecka K, Kasiński A, Biela T, Bocho-Janiszewska A, Laskowska A, Szeleszczuk Ł, Gawlak M, Sobczak M, Oledzka E. Future-Oriented Nanosystems Composed of Polyamidoamine Dendrimer and Biodegradable Polymers as an Anticancer Drug Carrier for Potential Targeted Treatment. Pharmaceutics 2024; 16:1482. [PMID: 39598604 PMCID: PMC11597463 DOI: 10.3390/pharmaceutics16111482] [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: 10/25/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Camptothecin (CPT) is a well-known chemical compound recognized for its significant anticancer properties. However, its clinical application remains limited due to challenges related to CPT's high hydrophobicity and the instability of its active form. To address these difficulties, our research focused on the development of four novel nanoparticulate systems intended for either oral or intravenous administration. Methods: These nanosystems were based on a poly(amidoamine) (PAMAM) dendrimer/CPT complex, which had been coated with biodegradable homo- and copolymers, designed with appropriate physicochemical properties and chain microstructures. Results: The resulting nanomaterials, with diameters ranging from 110 to 406 nm and dispersity values between 0.10 and 0.67, exhibited a positive surface charge and were synthesized using biodegradable poly(L-lactide) (PLLA), poly(L-lactide-co-ε-caprolactone) (PLACL), and poly(glycolide-co-ε-caprolactone) (PGACL). Biological assessments, including cell viability and hemolysis tests, indicated that all polymers demonstrated less than 5% hemolysis, confirming their hemocompatibility for potential intravenous use. Furthermore, fibroblasts exposed to these matrices showed concentration-dependent viability. The entrapment efficiency (EE) of CPT reached up to 27%, with drug loading (DL) values as high as 17%. The in vitro drug release studies lasted over 400 h with the use of phosphate buffer solutions at two different pH levels, demonstrating that time-dependent processes allowed for a gradual and controlled release of CPT from the developed nanosystems. The release kinetics of the active compound at pH 7.4 ± 0.05 and 6.5 ± 0.05 followed near-first-order or first-order models, with diffusion and Fickian/non-Fickian transport mechanisms. Importantly, the nanoparticulate systems enabled the stabilization of the pharmacologically active form of CPT, while providing protection against hydrolysis, even in physiological environments. Conclusions: In our opinion, these results underscore the promising future of biodegradable nanosystems as effective drug delivery systems (DDSs) for targeted cancer treatment, offering stability and efficacy over short, medium, and long-term applications.
Collapse
Affiliation(s)
- Katarzyna Strzelecka
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland; (K.S.); (A.K.); (M.S.)
| | - Adam Kasiński
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland; (K.S.); (A.K.); (M.S.)
| | - Tadeusz Biela
- Department of Polymer Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112 Str., 90-363 Lodz, Poland;
| | - Anita Bocho-Janiszewska
- Faculty of Applied Chemistry, Casimir Pulaski Radom University, Chrobrego 27 Str., 26-600 Radom, Poland;
| | - Anna Laskowska
- Department of Pharmaceutical Microbiology, Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b Str., 02-097 Warsaw, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland;
| | - Maciej Gawlak
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b Str., 02-097 Warsaw, Poland;
| | - Marcin Sobczak
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland; (K.S.); (A.K.); (M.S.)
| | - Ewa Oledzka
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland; (K.S.); (A.K.); (M.S.)
| |
Collapse
|
44
|
Sakhaii P, Bohorc B, Olpp T, Mohnicke M, Rieke-Zapp J, Dhal PK. Radio frequency gradient enhanced diffusion-edited semi-solid state NMR spectroscopy for detailed structural characterization of chemically modified hyaluronic acid hydrogels. Sci Rep 2024; 14:28612. [PMID: 39562623 PMCID: PMC11577061 DOI: 10.1038/s41598-024-78731-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Applications of functionalized hyaluronic acid (HA) hydrogels for numerous biomedical applications requires their detailed structural characterization. Since these materials are prepared by multistep chemical modifications in the solid phase and not amenable to characterization by standard analytical tools, we employed high-resolution solid-state NMR spectroscopy to gain detailed insights into the structures of the functionalized HA hydrogels. Divinyl sulfone crosslinked HA hydrogels were converted into maleimide-functionalized hydrogels, which were subjected to chemoselective thiol-maleimide reaction using L-cysteine as the protein mimetic thiol reagent. To overcome challenges associated with obtaining high-resolution NMR spectra of crosslinked hydrogels (such as line broadening and overlapping of signals of the hydrogel with those of residual reagents and solvents used during multi-step reaction processes on insoluble polymer matrices), we devised a radio frequency mediated diffusion-edited semi solid-state NMR technique. This technique enabled us to record NMR spectra of hydrogels exclusively by effectively suppressing signals associated with low molecular weight impurities. Thus, it became possible to perform in-depth characterization of these chemically modified HA hydrogels including quantification of reaction outcome for each reaction step.
Collapse
Affiliation(s)
- Peyman Sakhaii
- Global CMC Development, Global R&D, Sanofi, Industrial Park Hoechst, D-65926, Frankfurt/Main, Germany.
| | - Bojan Bohorc
- Global CMC Development, Global R&D, Sanofi, Industrial Park Hoechst, D-65926, Frankfurt/Main, Germany
| | - Thomas Olpp
- Global CMC Development, Global R&D, Sanofi, Industrial Park Hoechst, D-65926, Frankfurt/Main, Germany
| | - Mandy Mohnicke
- Global CMC Development, Global R&D, Sanofi, Industrial Park Hoechst, D-65926, Frankfurt/Main, Germany
| | - Joerg Rieke-Zapp
- Manufacturing Science and Analytical Technology, Sanofi, Industrial Park Hoechst, D- 65926, Frankfurt/Main, Germany
| | - Pradeep K Dhal
- Global CMC Development, Global R&D, Sanofi, 350 Water Street, MA 02141, Cambridge, USA.
| |
Collapse
|
45
|
Sobornova VV, Mulloyarova VV, Belov KV, Dyshin AA, Tolstoy PM, Kiselev MG, Khodov IA. Structural and sorption characteristics of an aerogel composite material loaded with flufenamic acid: insights from MAS NMR and high-pressure NOESY studies. Phys Chem Chem Phys 2024; 26:27301-27313. [PMID: 39439409 DOI: 10.1039/d4cp03217a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
The structural and sorption characteristics of a composite material consisting of a silica aerogel loaded with flufenamic acid were investigated using a variety of nuclear magnetic resonance techniques. The composite structure was analyzed using magic angle spinning nuclear magnetic resonance (MAS NMR) spectroscopy, which revealed significant interactions between the aerogel matrix and the FFA molecules. Solid-state 29Si NMR provided insights into the aerogel's stability, while 1H and 13C NMR confirmed the presence of FFA in the matrix, with signals from FFA molecules observed alongside tetraethoxysilane (TEOS) groups. Ethanol-induced desorption of FFA led to narrowed spectral lines, suggesting the breaking of intermolecular hydrogen bonds. 19F MAS NMR spectra indicated changes in FFA local environments upon loading into AG pores. Evaluation of CO2 sorption characteristics using 13C NMR demonstrated a slower sorption rate for AG + FFA than that for pure AG, attributed to decreased pore volume. Furthermore, nuclear Overhauser effect spectroscopy (NOESY) was employed to explore the conformational behavior of FFA within the aerogel matrix. The results indicated a shift in conformer populations, particularly those related to the rotation of one cyclic fragment relative to the other. These findings provide insights into the structural and sorption characteristics of the AG + FFA composite, which are valuable for developing novel drug solid forms.
Collapse
Affiliation(s)
- Valentina V Sobornova
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, Akademicheskaja st., 1, 153045, Ivanovo, Russia.
| | - Valeriya V Mulloyarova
- Institute of Chemistry, Saint Petersburg State University, 198504, Petergof, Universitetskii prospect, 26, Saint Petersburg, Russia
| | - Konstantin V Belov
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, Akademicheskaja st., 1, 153045, Ivanovo, Russia.
| | - Alexey A Dyshin
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, Akademicheskaja st., 1, 153045, Ivanovo, Russia.
| | - Peter M Tolstoy
- Institute of Chemistry, Saint Petersburg State University, 198504, Petergof, Universitetskii prospect, 26, Saint Petersburg, Russia
| | - Mikhail G Kiselev
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, Akademicheskaja st., 1, 153045, Ivanovo, Russia.
| | - Ilya A Khodov
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, Akademicheskaja st., 1, 153045, Ivanovo, Russia.
| |
Collapse
|
46
|
Garaizar A, Díaz-Oviedo D, Zablowsky N, Rissanen S, Köbberling J, Sun J, Steiger C, Steigemann P, Mann FA, Meier K. Toward understanding lipid reorganization in RNA lipid nanoparticles in acidic environments. Proc Natl Acad Sci U S A 2024; 121:e2404555121. [PMID: 39475644 PMCID: PMC11551392 DOI: 10.1073/pnas.2404555121] [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: 03/05/2024] [Accepted: 10/02/2024] [Indexed: 11/13/2024] Open
Abstract
The use of lipid nanoparticles (LNPs) for therapeutic RNA delivery has gained significant interest, particularly highlighted by recent milestones such as the approval of Onpattro and two mRNA-based SARS-CoV-2 vaccines. However, despite substantial advancements in this field, our understanding of the structure and internal organization of RNA-LNPs -and their relationship to efficacy, both in vitro and in vivo- remains limited. In this study, we present a coarse-grained molecular dynamics (MD) approach that allows for the simulations of full-size LNPs. By analyzing MD-derived structural characteristics in conjunction with cellular experiments, we investigate the effect of critical parameters, such as pH and composition, on LNP structure and potency. Additionally, we examine the mobility and chemical environment within LNPs at a molecular level. Our findings highlight the significant impact that LNP composition and internal molecular mobility can have on key stages of LNP-based intracellular RNA delivery.
Collapse
Affiliation(s)
- Adiran Garaizar
- Drug Discovery Sciences, Bayer Pharmaceuticals, Wuppertal42113, Germany
- Computational Life Science, Bayer Crop Science, Monheim am Rhein40789, Germany
| | - David Díaz-Oviedo
- Drug Discovery Sciences, Bayer Pharmaceuticals, Wuppertal42113, Germany
| | - Nina Zablowsky
- Lead Discovery, Nuvisan Innovation Campus Berlin, Berlin13353, Germany
| | - Sami Rissanen
- Chemical and Pharmaceutical Development, Bayer Pharmaceuticals, Turku20210, Finland
| | | | - Jiawei Sun
- Chemical and Pharmaceutical Development, Bayer Pharmaceuticals, Berlin13353, Germany
| | - Christoph Steiger
- Chemical and Pharmaceutical Development, Bayer Pharmaceuticals, Berlin13353, Germany
| | | | - Florian A. Mann
- Chemical and Pharmaceutical Development, Bayer Pharmaceuticals, Berlin13353, Germany
| | - Katharina Meier
- Drug Discovery Sciences, Bayer Pharmaceuticals, Wuppertal42113, Germany
| |
Collapse
|
47
|
Dutta D, Pajaniradje S, Nair AS, Chandramohan S, Bhat SA, Manikandan E, Rajagopalan R. An in-vitro study of active targeting & anti-cancer effect of folic acid conjugated chitosan encapsulated indole curcumin analogue nanoparticles. Int J Biol Macromol 2024; 282:136990. [PMID: 39505180 DOI: 10.1016/j.ijbiomac.2024.136990] [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: 06/09/2024] [Revised: 10/18/2024] [Accepted: 10/26/2024] [Indexed: 11/08/2024]
Abstract
Natural compounds like Curcumin with anti-cancer, anti-inflammatory and anti-bacterial properties are good target for drug development but its poor aqueous solubility, bioavailability, and low retention properties makes it a poor drug candidate in clinical settings. Here in this study, we have used an indole curcumin analogue (ICA) that has better bioavailability and enhanced permeability and retention (EPR) effect than curcumin. To make an active targeting drug we have designed folic acid conjugated chitosan-based nanoparticles encapsulating Indole curcumin analogue (CS-FA-ICA-np). The physical characteristics of CS-FA-ICA-np were evaluated by DLS, SEM, FTIR, XPS, XRD and TGA. Anti-cancer activity was analyzed using MTT, Fluorescence staining, Flow cytometry, comet assay, DNA fragmentation assay, wound healing, gelatin zymography, chick chorioallantoic membrane (CAM) assay and hemolysis assay. The size of CS-FA-ICA-nps were found to be 111 nm, and spherical in shape as observed in SEM. In-vitro assays show that CS-FA-ICA np has IC50 of 90 μg/mL in MDA-MB-231, increases ROS concentration, arrests cell cycle in G2-M phase, reduces matrix metalloproteinase-9 (MMP-9) activity and initiates apoptosis in cancer cells. Our results indicate that encapsulation of ICA increases its anti-cancer effect, drug stability, enhanced drug delivery to cancer microenvironment.
Collapse
Affiliation(s)
- Dipranil Dutta
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - Sankar Pajaniradje
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - Anjali Suresh Nair
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - Sathyapriya Chandramohan
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - Suhail Ahmad Bhat
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - E Manikandan
- Centre for Nano Sciences and Technology, Madanjeet School of Green Energy Technologies, Pondicherry University, Puducherry 605014, India
| | - Rukkumani Rajagopalan
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India.
| |
Collapse
|
48
|
Völlmecke K, Kramer M, Horky C, Dückmann O, Mulac D, Langer K, Kuckling D. Self-immolative polydisulfides and their use as nanoparticles for drug delivery systems. RSC Adv 2024; 14:35568-35577. [PMID: 39512642 PMCID: PMC11541933 DOI: 10.1039/d4ra07228f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024] Open
Abstract
Over the last few decades, nanotechnology has established to be a promising field in medicine. A remaining dominant challenge in today's pharmacotherapy is the limited selectivity of active pharmaceutical ingredients and associated undesirable side effects. Controlled drug release can be promoted by smart drug delivery systems, which release embedded API primarily depending on specific stimuli. Consequently, also the microenvironment of tumor tissue can be used advantageously. Dithiothreitol (DTT) based self-immolative polydisulfides were synthesized that preferentially respond to pathologically increased glutathione (GSH) concentrations, as found in solid tumors. The synthesis with different degrees of polymerisation was investigated as well as the synthesis of a copolymer consisting of dithiothreitol and butanedithiol (BDT). Toxicity tests were carried out on pure polymers and their degradation products. The ability to degrade was examined at pathological and physiological glutathione concentrations in order to test the suitability of the polymer as a matrix for nanoparticulate carrier systems. In addition, the processability of one polymer into nanoparticles was investigated as well as the degradation behaviour with glutathione.
Collapse
Affiliation(s)
| | - Maurice Kramer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Münster Corrensstr. 48 D-48149 Münster Germany
| | - Corinna Horky
- Institute of Pharmaceutical Technology and Biopharmacy, University of Münster Corrensstr. 48 D-48149 Münster Germany
| | - Oliver Dückmann
- Paderborn University Warburger Straße 100 33098 Paderborn Germany
| | - Dennis Mulac
- Institute of Pharmaceutical Technology and Biopharmacy, University of Münster Corrensstr. 48 D-48149 Münster Germany
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Münster Corrensstr. 48 D-48149 Münster Germany
| | - Dirk Kuckling
- Paderborn University Warburger Straße 100 33098 Paderborn Germany
| |
Collapse
|
49
|
Hari Priya VM, Ganapathy A A, Veeran MG, Raphael M S, Kumaran A. Nanotechnology-based drug delivery platforms for erectile dysfunction: addressing efficacy, safety, and bioavailability concerns. Pharm Dev Technol 2024; 29:996-1015. [PMID: 39392251 DOI: 10.1080/10837450.2024.2414379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
Erectile dysfunction (ED), is a common and multidimensional sexual disorder, which comprises changes among any of the processes of the erectile response such as organic, relational, and psychological. However, both endocrine and nonendocrine causes of ED produce substantial health implications including depression and anxiety due to poor sexual performance, eventually affecting man's life eminence. Marginally invasive interventions following ED consist of lifestyle modifications, oral drugs, injections, vacuum erection devices, etc. Nevertheless, these conventional treatment regimens follow certain drawbacks such as efficacy and safety issues, and navigate to the development of novel therapeutic approaches such as nanomedicine for ED management. Nanotechnology-centred drug delivery platforms are being explored to minimize these limitations with better in vitro and in vivo effectiveness. Moreover, nanomedicine and nanocarrier-linked approaches are rapidly developing science in the nanoscale range, which contributes to site-specific delivery in a controlled manner and has generated considerable interest prominent to their potential to enhance bioavailability, decrease side effects, and avoidance of first-pass metabolism. This review provides an overview of recent discoveries regarding various nanocarriers and nano-delivery methods, along with current trends in the clinical aspects of ED. Additionally, strategies for clinical translation have been incorporated.
Collapse
Affiliation(s)
- Vijayakumari Mahadevan Hari Priya
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anand Ganapathy A
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Midhu George Veeran
- Corporate Research and Development Centre (CRDC), HLL Lifecare Ltd, Akkulam, Thiruvananthapuram, India
| | - Shyni Raphael M
- Department of Chemistry, Government College for Women, Thiruvananthapuram, India
| | - Alaganandam Kumaran
- Agroprocessing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
50
|
Vigo M, Haro-Martínez E, Ruiz E, Fumadó-Navarro J, Placci M, Muro S. New Cellular Models to Support Preclinical Studies on ICAM-1-Targeted Drug Delivery. J Drug Deliv Sci Technol 2024; 101:106170. [PMID: 39669707 PMCID: PMC11633371 DOI: 10.1016/j.jddst.2024.106170] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Intercellular adhesion molecule 1 (ICAM-1) is a cell-surface protein actively explored for targeted drug delivery. Anti-ICAM-1 nanocarriers (NCs) target ICAM-1-positive sites after intravenous injection in animal models, but quantitative mechanistic examination of cellular-level transport in vivo is not possible. Prior studies in human cell cultures indicated efficient uptake of these formulations via cell adhesion molecule-(CAM)-mediated endocytosis. However, ICAM-1 sequence differs among species; thus, whether anti-ICAM-1 NCs induce similar behavior in animal cells, key for intracellular drug delivery, is unknown. To begin bridging this gap, we first qualitatively verified intracellular transport of anti-ICAM-1 NCs in vivo and then developed new cellular models expressing ICAM-1 from mouse, dog, pig, and monkey, species relevant to pharmaceutical translation and veterinary medicine. ICAM-1 expression was verified by flow cytometry and confocal microscopy. These cells showed specific targeting compared to IgG NCs or cells treated with anti-ICAM-1 blocker. Anti-ICAM-1 NCs entered cells in a time- and temperature-dependent manner, with kinetics and pathway compatible with CAM-mediated endocytosis. All parameters tested were strikingly similar to those from human cells expressing ICAM-1 endogenously. Therefore, this new cellular platform represents a valuable tool that can be used in parallel to support in vivo studies on ICAM-1-targeted NCs during pharmaceutical translation.
Collapse
Affiliation(s)
- Marco Vigo
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, 08028, Spain
- Biomedicine Doctorate Program, University of Barcelona, 08007, Spain
| | - Elena Haro-Martínez
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, 08028, Spain
| | - Eloy Ruiz
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, 08028, Spain
| | - Josep Fumadó-Navarro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, 08028, Spain
| | - Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, 08028, Spain
- Biotechnology Doctorate Program, University of Barcelona, 080007, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, 08028, Spain
- Institution of Catalonia for Research and Advanced Studies (ICREA), Barcelona, 08010, Spain
- Institute for Bioscience and Biotechnology Research and Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
| |
Collapse
|