1
|
Zhu D, Brückner D, Sosniok M, Skiba M, Feliu N, Gallego M, Liu Y, Schulz F, Falkenberg G, Parak WJ, Sanchez-Cano C. Size-dependent penetration depth of colloidal nanoparticles into cell spheroids. Adv Drug Deliv Rev 2025; 222:115593. [PMID: 40339992 DOI: 10.1016/j.addr.2025.115593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
The penetration of nanoparticle (NP)-based drugs into tissue is essential for their use as nanomedicines. Systematic studies about how different NP properties, such as size, influence NP penetration are helpful for the development of NP-based drugs. An overview of how NPs of different sizes may penetrate three-dimensional cell spheroids is given. In particular different techniques for experimental analysis are compared, including mass spectrometry, flow cytometry, optical fluorescence microscopy, X-ray fluorescence microscopy, and transmission electron microscopy. An experimental data set is supplemented exclusively made for this review, in which the results of different techniques are visualized. Limitations of the analysis techniques for different types of NPs, including carbon-based materials, are discussed.
Collapse
Affiliation(s)
- Dingcheng Zhu
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany; Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Zhejiang Key Laboratory of Organosilicon Material Technology, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121 Zhejiang, China
| | - Dennis Brückner
- Deutsches Elektronen-Synchrotron DESY, Photon Science, 22607 Hamburg, Germany
| | - Martin Sosniok
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany; Zentrum für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Marvin Skiba
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Neus Feliu
- Zentrum für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Marta Gallego
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE) Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain
| | - Yang Liu
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Florian Schulz
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany
| | - Gerald Falkenberg
- Deutsches Elektronen-Synchrotron DESY, Photon Science, 22607 Hamburg, Germany.
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures, University of Hamburg 22761 Hamburg, Germany.
| | - Carlos Sanchez-Cano
- Donostia International Physics Center, 20018 Donostia-San Sebastian, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, 20018 Donostia-San Sebastian, Spain.
| |
Collapse
|
2
|
Cave J, Christiono A, Schiavone C, Pownall HJ, Cristini V, Staquicini DI, Pasqualini R, Arap W, Brinker CJ, Campen M, Wang Z, Van Nguyen H, Noureddine A, Dogra P. Rational Design of Safer Inorganic Nanoparticles via Mechanistic Modeling-Informed Machine Learning. ACS NANO 2025; 19:21538-21555. [PMID: 40460056 PMCID: PMC12177941 DOI: 10.1021/acsnano.5c03590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 06/11/2025]
Abstract
The safety of inorganic nanoparticles (NPs) remains a critical challenge for their clinical translation. To address this, we developed a machine learning (ML) framework that predicts NP toxicity both in vitro and in vivo, leveraging physicochemical properties and experimental conditions. A curated in vitro cytotoxicity dataset was used to train and validate binary classification models, with top-performing models undergoing explainability analysis to identify key determinants of toxicity and establish structure-toxicity relationships. External testing with diverse inorganic NPs validated the predictive accuracy of the framework for in vitro settings. To enable organ-specific toxicity predictions in vivo, we integrated a physiologically based pharmacokinetic (PBPK) model into the ML pipeline to quantify NP exposure across organs. Retraining the ML models with PBPK-derived exposure metrics yielded robust predictions of organ-specific nanotoxicity, further validating the framework. This PBPK-informed ML approach can thus serve as a potential alternative approach to streamline NP safety assessment, enabling the rational design of safer NPs and expediting their clinical translation.
Collapse
Affiliation(s)
- Joseph Cave
- Mathematics
in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas77030, United States
- Physiology,
Biophysics, and Systems Biology Program, Graduate School of Medical
Sciences, Weill Cornell Medicine, New York, New York10065, United States
| | - Anne Christiono
- Department
of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States
| | - Carmine Schiavone
- Mathematics
in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas77030, United States
- Department
of Chemical, Materials, and Industrial Production Engineering, University of Naples Federico II, Naples80138, Italy
| | - Henry J. Pownall
- Department
of Medicine, Houston Methodist, Houston, Texas77030, United States
- Department
of Medicine, Weill Cornell Medicine, New York, New York10065, United States
| | - Vittorio Cristini
- Mathematics
in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas77030, United States
- Physiology,
Biophysics, and Systems Biology Program, Graduate School of Medical
Sciences, Weill Cornell Medicine, New York, New York10065, United States
- Neal
Cancer
Center, Houston Methodist Research Institute, Houston, Texas77030, United States
- Department
of Imaging Physics, University of Texas
M.D. Anderson Cancer Center, Houston, Texas77030, United States
| | - Daniela I. Staquicini
- Rutgers
Cancer Institute, Newark, New Jersey08901, United States
- Division
of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey08901, United States
| | - Renata Pasqualini
- Rutgers
Cancer Institute, Newark, New Jersey08901, United States
- Division
of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey08901, United States
| | - Wadih Arap
- Rutgers
Cancer Institute, Newark, New Jersey08901, United States
- Division
of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey08901, United States
| | - C. Jeffrey Brinker
- Department
of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico87106, United States
| | - Matthew Campen
- College
of Pharmacy, University of New Mexico, Albuquerque, New Mexico87106, United States
| | - Zhihui Wang
- Mathematics
in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas77030, United States
- Neal
Cancer
Center, Houston Methodist Research Institute, Houston, Texas77030, United States
- Department
of Physiology and Biophysics, Weill Cornell
Medical College, New York, New York10065, United States
| | - Hien Van Nguyen
- Department
of Electrical and Computer Engineering, University of Houston, Houston, Texas77204, United States
| | - Achraf Noureddine
- Department
of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico87106, United States
| | - Prashant Dogra
- Mathematics
in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas77030, United States
- Department
of Physiology and Biophysics, Weill Cornell
Medical College, New York, New York10065, United States
| |
Collapse
|
3
|
Wang Y, Tang X, Luo S, Zhang Z, Cao Y. Intratracheal instillation of graphene oxide with different diameters suppressed toll-like receptor 3-mediated lipid droplet biogenesis in lungs and livers of mice. Toxicol Res (Camb) 2025; 14:tfaf069. [PMID: 40384842 PMCID: PMC12085196 DOI: 10.1093/toxres/tfaf069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/23/2025] [Accepted: 05/02/2025] [Indexed: 06/04/2025] Open
Abstract
Recent advances have established lipid droplets as dynamic innate immune hubs coordinating cellular metabolism and defense mechanisms. While previous studies primarily focused on nanomaterials (NMs) altering lipid metabolism to influence lipid droplet dynamics, this study pioneers the investigation of NM-induced immune modulation via Toll-like receptor (TLR) pathways as a novel regulatory axis for lipid droplets. Building on our prior findings that graphene oxide (GO) impaired TLR3-mediated lipid signaling, we systematically explored the role of GO's diameter in modulating this process. Mice were subjected to daily intratracheal instillation of three GO variants (50-200 nm, <500 nm or > 500 nm) at 1 mg/kg for 7 days. Although no significant change in body weight or organ coefficient was observed, all GO exposure suppressed lipid staining in mouse lungs and livers, correlating with altered co-localization of TLR3 and perilipin 2 (PLIN2), critical regulators of lipid droplet biogenesis. Down-regulation of TLR3 signaling components, namely interferon induced protein with tetratricopeptide repeats 1 (IFIT1), radical S-adenosyl methionine domain containing 2 (RSAD2), and PLIN2, occurred in a diameter-dependent manner, with GO 50-200 nm showing the most pronounced effects, likely attributable to the smallest hydrodynamic size and polydispersity index in suspension. This work provides evidence that NM geometry governs TLR-mediated lipid droplet regulation, bridging the knowledge gap between nanotoxicology and immunometabolic cross-talking, a paradigm distinct from conventional lipid metabolism-focused nanotoxicological studies.
Collapse
Affiliation(s)
- Yijin Wang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Changsheng Road No. 28, Zhengxiang District, Hengyang city, Hunan Province, Hengyang 421001, China
| | - Xiaomin Tang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Changsheng Road No. 28, Zhengxiang District, Hengyang city, Hunan Province, Hengyang 421001, China
| | - Sihuan Luo
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Changsheng Road No. 28, Zhengxiang District, Hengyang city, Hunan Province, Hengyang 421001, China
| | - Zhaohui Zhang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Changsheng Road No. 28, Zhengxiang District, Hengyang city, Hunan Province, Hengyang 421001, China
| | - Yi Cao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Changsheng Road No. 28, Zhengxiang District, Hengyang city, Hunan Province, Hengyang 421001, China
| |
Collapse
|
4
|
Venturini J, Chakraborty A, Baysal MA, Tsimberidou AM. Developments in nanotechnology approaches for the treatment of solid tumors. Exp Hematol Oncol 2025; 14:76. [PMID: 40390104 PMCID: PMC12090476 DOI: 10.1186/s40164-025-00656-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 05/21/2025] Open
Abstract
Nanotechnology has revolutionized cancer therapy by introducing advanced drug delivery systems that enhance therapeutic efficacy while reducing adverse effects. By leveraging various nanoparticle platforms-including liposomes, polymeric nanoparticles, and inorganic nanoparticles-researchers have improved drug solubility, stability, and bioavailability. Additionally, new nanodevices are being engineered to respond to specific physiological conditions like temperature and pH variations, enabling controlled drug release and optimizing therapeutic outcomes. Beyond drug delivery, nanotechnology plays a crucial role in the theranostic field due to the functionalization of specific materials that combine tumor detection and targeted treatment features. This review analyzes the clinical impact of nanotechnology, spanning from early-phase trials to pivotal phase 3 studies that have obtained regulatory approval, while also offering a critical perspective on the preclinical domain and its translational potential for future human applications. Despite significant progress, greater attention must be placed on key challenges, such as biocompatibility barriers and the lack of regulatory standardization, to ensure the successful translation of nanomedicine into routine clinical practice.
Collapse
Affiliation(s)
- Jacopo Venturini
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
- Current Affiliation: Department of Medical Oncology, Careggi University Hospital, Florence, Italy
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Saladino GM, Brodin B, Ciobanu M, Kilic NI, Toprak MS, Hertz HM. Design and Biodistribution of PEGylated Core-Shell X-ray Fluorescent Nanoparticle Contrast Agents. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26338-26347. [PMID: 40265284 PMCID: PMC12067382 DOI: 10.1021/acsami.5c01902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
Nanoparticle (NP) uptake by macrophages and their accumulation in undesired organs such as the liver and spleen constitute a major barrier to the effective delivery of NPs to targeted tissues for bioimaging and therapeutics. Surface functionalization with polyethylene glycol (PEG) has been demonstrated to be a promising strategy to limit NP sequestration, although its longitudinal stability under physiological conditions and impact on the NP biodistribution have not been investigated with an in vivo quantitative approach. X-ray fluorescence (XRF) imaging has been employed to noninvasively map the in vivo biodistribution of purposely designed molybdenum-based contrast agents, leading to submillimeter resolution, elemental specificity, and high penetration depth. In the present work, we design a stepwise layering approach for NP synthesis to investigate the role of chemisorbed and physisorbed PEG on silica-coated molybdenum-based contrast agents in affecting their in vivo biodistribution, using whole-body XRF imaging. Comparative quantitative in vivo studies indicated that physisorbed PEG (1.5 kDa) did not substantially affect the biodistribution, while the chemisorption route with mPEG-Si (6-9 PEG units) led to significant macroscopic variations in the biodistribution, leading to a reduction in NP uptake by the liver. Furthermore, the results highlighted the major role of the spleen in compensating for the limited sequestration by the liver, microscopically validated with a multiscale imaging approach with fluorophore doping of the silica shell. These findings demonstrated the promising role of XRF imaging for the rapid assessment of surface-functionalized contrast agents with whole-body in vivo quantitative pharmacokinetic studies, establishing the groundwork for developing strategies to identify and bypass undesired NP uptake.
Collapse
Affiliation(s)
- Giovanni M. Saladino
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, SE 10691, Sweden
- Department
of Radiology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Bertha Brodin
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, SE 10691, Sweden
| | - Mihai Ciobanu
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, SE 10691, Sweden
| | - Nuzhet I. Kilic
- Department
of Fiber and Polymer Technology, School of Engineering Sciences in
Chemistry, Biotechnology and Health, KTH
Royal Institute of Technology, Stockholm, SE 100 44, Sweden
| | - Muhammet S. Toprak
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, SE 10691, Sweden
| | - Hans M. Hertz
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm, SE 10691, Sweden
| |
Collapse
|
6
|
Harting H, Herrmann T, Ehlert N, Meißner J, Angrisani N, Reifenrath J. Comparison of accumulation and distribution of PEGylated and CD-47-functionalized magnetic nanoporous silica nanoparticles in an in vivo mouse model of implant infection. PLoS One 2025; 20:e0321888. [PMID: 40315195 PMCID: PMC12047780 DOI: 10.1371/journal.pone.0321888] [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: 10/07/2024] [Accepted: 03/12/2025] [Indexed: 05/04/2025] Open
Abstract
INTRODUCTION Drug targeting using nanoparticles is a much-researched topic. Rapid interactions of nanoparticles with the host's immune system and clearance from the circulation is a major problem resulting in non-satisfying accumulation rates in the desired region. The aim of the presented study was to compare organ distribution and implant accumulation of magnetic nanoporous silica nanoparticles (MNPSNP) functionalized with either Polyethylenglycol (PEG) or CD-47 in vivo in a mouse model of implant infection. METHODS Synthesis and functionalization of the magnetic core-shell nanoparticles is described. In the in vivo study, 32 mice were included and received an in staphylococcus aureus solution preincubated magnetic implant subcutaneously on the left and a nonmagnetic implant on the right hind leg. MNPSNP accumulation in the inner organs as well as on and around the implants was analyzed in dependence on the functionalization. RESULTS MNPSNP were successfully functionalized with PEG or CD-47. In vivo, unexpectedly both nanoparticle variants accumulated mainly in liver and spleen. In the tissue, surrounding the implants higher nanoparticle accumulation was seen in areas with more severe signs of inflammation Nanoparticles were detectable on both implant materials, but accumulation rate was very low. CONCLUSION Although various literature describes higher accumulation rates for nanoparticles functionalized with CD-47 in target areas and a reduced accumulation in liver and spleen, this could not be shown within this study. Possible instability or rapid agglomeration of the particles are conceivable reasons. Higher accumulation rates in areas with more severe signs of inflammation indicate that inflammatory cells might be essential for the delivery of nanoparticles into inflamed regions.
Collapse
Affiliation(s)
- Heidi Harting
- Hannover Medical School, Department of Orthopaedic Surgery, DIAKOVERE Annastift, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Timo Herrmann
- Institute for Inorganic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Nina Ehlert
- Institute for Inorganic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Jessica Meißner
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hanover, Foundation, Hannover, Germany
| | - Nina Angrisani
- Hannover Medical School, Department of Orthopaedic Surgery, DIAKOVERE Annastift, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Janin Reifenrath
- Hannover Medical School, Department of Orthopaedic Surgery, DIAKOVERE Annastift, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
7
|
Le Meur M, Pignatelli J, Blasi P, Palomo V. Nanoparticles targeting the central circadian clock: Potential applications for neurological disorders. Adv Drug Deliv Rev 2025; 220:115561. [PMID: 40120723 DOI: 10.1016/j.addr.2025.115561] [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: 11/20/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Circadian rhythms and their involvement with various human diseases, including neurological disorders, have become an intense area of research for the development of new pharmacological treatments. The location of the circadian clock machinery in the central nervous system makes it challenging to reach molecular targets at therapeutic concentrations. In addition, a timely administration of the therapeutic agents is necessary to efficiently modulate the circadian clock. Thus, the use of nanoparticles in circadian clock dysfunctions may accelerate their clinical translation by addressing these two key challenges: enhancing brain penetration and/or enabling their formulation in chronodelivery systems. This review describes the implications of the circadian clock in neurological pathologies, reviews potential molecular targets and their modulators and suggests how the use of nanoparticle-based formulations could improve their clinical success. Finally, the potential integration of nanoparticles into chronopharmaceutical drug delivery systems will be described.
Collapse
Affiliation(s)
- Marion Le Meur
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain; Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum - Università di Bologna, 40127 Bologna, Italy
| | - Jaime Pignatelli
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Paolo Blasi
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum - Università di Bologna, 40127 Bologna, Italy.
| | - Valle Palomo
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain; Unidad de Nanobiotecnología asociada al Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain.
| |
Collapse
|
8
|
Rodella G, Préat V, Gallez B, Malfanti A. Design Strategies for Hyaluronic Acid-based Drug Delivery Systems in Cancer Immunotherapy. J Control Release 2025; 383:113784. [PMID: 40294800 DOI: 10.1016/j.jconrel.2025.113784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Despite its robust therapeutic potential, cancer immunotherapy has provided little progress towards improved survival rates for patients bearing immunologically refractory tumors. The implementation of advanced drug delivery systems represents a powerful means of improving cancer immunotherapy by relieving immunosuppression and promoting immune response; however, the overall impact of these systems on immunotherapy currently remains modest. Hyaluronic acid represents a widely used polymer in drug delivery; meanwhile, recent studies linking hyaluronic acid to the immune system make this polymer an attractive component in the design of next-generation cancer immunotherapies. Herein, we review our current understanding of the immunological properties of hyaluronic acid and discuss them in the context of bioactive functions and immune-related interactions with receptors, immune, and cancer cells. We analyze the potential of hyaluronic acid as a component in advanced drug delivery systems, highlighting strategies for the design of more effective vaccines and cancer chemo-immunotherapies. Finally, we discuss critical considerations to facilitate design and clinical translation to overcome existing challenges and maximize therapeutic potential.
Collapse
Affiliation(s)
- Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Bernard Gallez
- UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; Departement of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo, 5, 35131 Padova, Italy.
| |
Collapse
|
9
|
Randhawa S, Saini TC, Bathla M, Bhardwaj R, Dhiman R, Acharya A. Nanomaterials in targeting amyloid-β oligomers: current advances and future directions for Alzheimer's disease diagnosis and therapy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:561-580. [PMID: 40297247 PMCID: PMC12035877 DOI: 10.3762/bjnano.16.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
The amyloid cascade hypothesis posits that amyloid-β oligomers (AβOs) are the most neurotoxic species in Alzheimer's disease (AD). These oligomers, characterized by their high β-sheet content, have been shown to significantly disrupt cell membranes, induce local inflammation, and impair autophagy processes, which collectively contribute to neuronal loss. As such, targeting AβOs specifically, rather than solely focusing on amyloid-β fibrils (AβFs), may offer a more effective therapeutic approach for AD. Recent advances in detection and diagnosis have emphasized the importance of accurately identifying AβOs in patient samples, enhancing the potential for timely intervention. In recent years, nanomaterials (NMs) have emerged as promising agents for addressing AβOs regarding their multivalent interactions, which can more effectively detect and inhibit AβO formation. This review provides an in-depth analysis of various nanochaperones developed to target AβOs, detailing their mechanisms of action and therapeutic potential via focusing on two main strategies, namely, disruption of AβOs through direct interaction and the inhibition of AβO nucleation by binding to intermediates of the oligomerization process. Evidence from in vivo studies indicate that NMs hold promise for ameliorating AD symptoms. Additionally, the review explores the different interaction mechanisms through which nanoparticles exhibit their inhibitory effects on AβOs, providing insights into their potential for clinical application. This comprehensive overview highlights the current advancements in NM-based therapies for AD and outlines future research directions aimed at optimizing these innovative treatments.
Collapse
Affiliation(s)
- Shiwani Randhawa
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Trilok Chand Saini
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Manik Bathla
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rahul Bhardwaj
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rubina Dhiman
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
| | - Amitabha Acharya
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
10
|
Wang J, Xin Y, Chen D, Zhang N, Xue Y, Liu X, Li X, Gao W, Hu Z, Sun T, Liu K, Tian W, Xu B, Lu Y. Ultra-Stable Gold Nanoparticles with Tunable Surface Characteristics. Angew Chem Int Ed Engl 2025:e202507954. [PMID: 40231741 DOI: 10.1002/anie.202507954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/16/2025]
Abstract
Gold nanoparticles (Au NPs), as a class of functional nanomaterials, have attracted considerable interest for biomedical applications owing to their unique chemical and physical properties. However, colloidal solutions of Au NPs are thermodynamically unstable because of their high surface energy, resulting in poor stability and biocompatibility in physiological environments. Herein, we present a novel strategy for coating Au NPs using in situ polymerization to form a three-dimensional (3D) network polymer shell around each particle. This approach enables the creation of an ultra-stable core-shell structure that effectively improves biocompatibility and stability, even in complex biological environments. The surface characteristics of the polymer shell can also be precisely tailored by carefully selecting the monomers to meet biomedical application requirements. These properties enable prolonged circulation within the bloodstream and enhanced tumor targeting in mice. This strategy offers an ultra-stable, aqueous-based, and biocompatible polymer shell for Au NPs, paving the way for the surface modification of gold nanomaterials in biomedical applications.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Yanbao Xin
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, P.R. China
| | - Dazhi Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Ningning Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Yao Xue
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Xinze Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Xiang Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Wenbin Gao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, P.R. China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130061, P.R. China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Tianmeng Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, P.R. China
- International Center of Future Science, Jilin University, Changchun, 130061, P.R. China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130061, P.R. China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, P.R. China
| | - Kun Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Wenjing Tian
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Bin Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Qianjin Street No. 2699, Changchun, 130012, P.R. China
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, California, 90095, USA
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, P.R. China
- Changping Laboratory, Beijing, 100871, P.R. China
| |
Collapse
|
11
|
Sutcliffe R, Doherty CPA, Morgan HP, Dunne NJ, McCarthy HO. Strategies for the design of biomimetic cell-penetrating peptides using AI-driven in silico tools for drug delivery. BIOMATERIALS ADVANCES 2025; 169:214153. [PMID: 39705787 DOI: 10.1016/j.bioadv.2024.214153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/08/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Cell-penetrating peptides (CPP) have gained rapid attention over the last 25 years; this is attributed to their versatility, customisation, and 'Trojan horse' delivery that evades the immune system. However, the current CPP rational design process is limited, as it requires several rounds of peptide synthesis, prediction and wet-lab validation, which is expensive, time-consuming and requires extensive knowledge in peptide chemistry. Artificial intelligence (AI) has emerged as a promising alternative which can augment the design process, for example by determining physiochemical characteristics, secondary structure, solvent accessibility, disorder and flexibility, as well as predicting in vivo behaviour such as toxicity and peptidase degradation. Other more recent tools utilise supervised machine learning (ML) to predict the penetrative ability of an amino acid sequence. The use of AI in the CPP design process has the potential to reduce development costs and increase the chances of success with respect to delivery. This review provides a survey of in silico tools and AI platforms which can be utilised in the design process, and the key features that should be taken into consideration when designing next generation CPPs.
Collapse
Affiliation(s)
- Rebecca Sutcliffe
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland
| | - Ciaran P A Doherty
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; Antigenesis Biologics, Crossgar, Northern Ireland, United Kingdom of Great Britain and Northern Ireland
| | - Hugh P Morgan
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; Antigenesis Biologics, Crossgar, Northern Ireland, United Kingdom of Great Britain and Northern Ireland
| | - Nicholas J Dunne
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
12
|
Tabrizi E, Li B. From innovation to application: safety concerns in nanomaterial implant coatings. Nanomedicine (Lond) 2025:1-4. [PMID: 40103528 DOI: 10.1080/17435889.2025.2480045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025] Open
Affiliation(s)
- Eileen Tabrizi
- Department of Orthopedics, School of Medicine, West Virginia University, Morgantown, WV, USA
- School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Bingyun Li
- Department of Orthopedics, School of Medicine, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
13
|
Benyettou F, Das G, Boitet M, Varghese S, Khair M, Das AK, Matouk Z, Prakasam T, Bazin P, Sharma SK, Thomas S, He Y, Straubinger R, Garai B, Jagannathan R, Gándara F, El-Roz M, Trabolsi A. Freezing-Activated Covalent Organic Frameworks for Precise Fluorescence Cryo-Imaging of Cancer Tissue. J Am Chem Soc 2025; 147:8188-8204. [PMID: 40013936 PMCID: PMC11912341 DOI: 10.1021/jacs.4c13848] [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: 10/03/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025]
Abstract
Cryosurgery represents a transformative approach in the treatment of resistant tumors, utilizing extreme cold to selectively ablate malignant tissue. However, the clinical success of this technique is constrained by the limited ability of current imaging techniques to differentiate effectively between cancerous and healthy tissues with high spatial resolution. To overcome this challenge, we present a nanoscale Covalent Organic Framework, nTG-DFP-COF, specifically designed to enhance fluorescence-guided cryo-imaging. This framework exhibits a unique temperature-dependent luminescence, that results in enhanced fluorescence emission under cryogenic conditions, enabling precise tissue differentiation during surgical procedures. Engineered for biocompatibility and water dispersibility, nTG-DFP-COF demonstrates minimal cytotoxicity and exceptional specificity toward cancer cells. Comprehensive in vitro, in vivo, and ex vivo evaluations confirm its structural stability and functional efficacy under cryogenic conditions. This innovation not only enhances the precision and safety of cryosurgical procedures but also advances the integration of diagnostic and therapeutic functionalities into a unified platform. By substantially improving tumor targeting accuracy, the use of nTG-DFP-COF will reduce the need for repeat surgeries, facilitate faster recovery, and minimize healthcare costs, thus setting a new standard in oncologic imaging and intervention.
Collapse
Affiliation(s)
- Farah Benyettou
- Chemistry
Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Gobinda Das
- Chemistry
Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Maylis Boitet
- Core
Technology Platforms, New York University
Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Sabu Varghese
- Core
Technology Platforms, New York University
Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Mostafa Khair
- Core
Technology Platforms, New York University
Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Akshaya Kumar Das
- Chemistry
Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Zineb Matouk
- Technology
Innovative Institute, Abu Dhabi 9639, United Arab
Emirates
| | - Thirumurugan Prakasam
- Chemistry
Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Philippe Bazin
- Normandie
Univ, ENSICAEN, UNICAEN, CNRS, LCS, Caen 14000, France
| | - Sudhir Kumar Sharma
- Engineering
Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Sneha Thomas
- Core
Technology Platforms, New York University
Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Yao He
- Core
Technology Platforms, New York University
Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Rainer Straubinger
- Core
Technology Platforms, New York University
Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Bikash Garai
- Chemistry
Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Ramesh Jagannathan
- Engineering
Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Felipe Gándara
- Instituto
de Ciencia de Materiales de Madrid-CSIC, C. Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain
| | - Mohamad El-Roz
- Normandie
Univ, ENSICAEN, UNICAEN, CNRS, LCS, Caen 14000, France
| | - Ali Trabolsi
- Chemistry
Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| |
Collapse
|
14
|
Chang MR, Matnurov EM, Wu C, Arakelyan J, Choe HJ, Kushnarev V, Yap JY, Soo XX, Chow MJ, Berger W, Ang WH, Babak MV. Leveraging Immunogenic Cell Death to Enhance the Immune Response against Malignant Pleural Mesothelioma Tumors. J Am Chem Soc 2025; 147:7908-7920. [PMID: 39992709 PMCID: PMC11887451 DOI: 10.1021/jacs.4c17966] [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: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025]
Abstract
Although various metal-based compounds have exhibited excellent immunogenic cell death (ICD)-inducing properties both in vitro and in vivo, the majority of these compounds have been discovered serendipitously. In this work, we have successfully synthesized and characterized 35 cyclometalated Au(III) complexes containing dithiocarbamate ligands, with 25 of these complexes being previously unreported. Their ability to induce phagocytosis in vitro against immunologically "cold" malignant pleural mesothelioma (MPM) cells was strongly dependent on the cyclometalated scaffold and the overall lipophilicity of the complexes. We elucidated the role of cell death mechanisms in the observed ICD effects and identified correlations between the ability of the complexes to induce necrotic cell death and ICD, both in vitro and in vivo. Complex 2G, with its high phagocytosis rates and low necrosis rates, was recognized as a bona fide ICD inducer, demonstrating a remarkably long-lasting immune response in vaccinated mice. In contrast, complex 1C, characterized by high phagocytosis rates and high necrosis rates, failed to elicit a sustained immune response upon following vaccination; however, it triggered selective activation of calreticulin in tumors upon direct in vivo administration. Overall, this study offers a framework for predicting ICD effects in vivo for structurally similar Au(III) complexes, with the potential for extension to other series of metal complexes.
Collapse
Affiliation(s)
- Meng Rui Chang
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Egor M. Matnurov
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Chengnan Wu
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Jemma Arakelyan
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Ho-Jung Choe
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Vladimir Kushnarev
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| | - Jian Yu Yap
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate
School - Integrated Science and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Xiu Xuan Soo
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Mun Juinn Chow
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
| | - Walter Berger
- Center for
Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, Vienna 1090, Austria
| | - Wee Han Ang
- Department
of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117543, Singapore
- NUS Graduate
School - Integrated Science and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Maria V. Babak
- Drug Discovery
Lab, Department of Chemistry, City University
of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, People’s Republic
of China
| |
Collapse
|
15
|
Deng H, Chen J, Wang H, Liu R, Zhang Y, Chang H, Tung CH, Zhang W. Hijacking the hyaluronan assisted iron endocytosis to promote the ferroptosis in anticancer photodynamic therapy. Carbohydr Polym 2025; 351:123123. [PMID: 39779030 DOI: 10.1016/j.carbpol.2024.123123] [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: 10/09/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
Photodynamic therapy (PDT) eradicates tumor cells by the light-stimulated reactive oxygen species, which also induces lipid peroxidation (LPO) and subsequently ferroptosis, an iron-depended cell death. Ferroptosis has a tremendous therapeutic potential in cancer treatment, however, the ferroptosis efficiency is largely limited by the available iron in cells. Through hijacking the CD44-mediated iron endocytosis of hyaluronan (HA), here PDT with enhanced ferroptosis was realized by a HA@Ce6 nanogel self-assembled from HA, a photosensitizer Chlorin e6 (Ce6) and Fe3+ as cross-linkers. Taking advantages of HA's natural affinity towards CD44, HA@Ce6 enabled a targeted Ce6 delivery in CD44-overexpressed breast cancer cells and meanwhile enhanced iron uptake to "fuel" ferroptosis together with the light-stimulated LPO. Further, HA@Ce6 demonstrated an excellent anticancer PDT efficacy and ferroptosis induction in the murine 4 T1 xenograft model. This HA@Ce6 successfully exploited the role of HA in iron transport to sensitize ferroptosis, providing a potent strategy to facilitate the anticancer PDT.
Collapse
Affiliation(s)
- Hong Deng
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Jiayu Chen
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Huimin Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Runmeng Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Yiyi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Hui Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Weiqi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China.
| |
Collapse
|
16
|
Fusco L, Gazzi A, Giro L, Schefer RB, D'Almeida SM, Cagliani R, Zoccheddu M, Uyar R, Besbinar Ö, Çelik D, Yilmazer A, Mitrano DM, Orecchioni M, Delogu LG. Nanoplastics: Immune Impact, Detection, and Internalization after Human Blood Exposure by Single-Cell Mass Cytometry. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413413. [PMID: 39449193 PMCID: PMC11938024 DOI: 10.1002/adma.202413413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/07/2024] [Indexed: 10/26/2024]
Abstract
The increasing exposure to nanoplastics (NPs) raises significant concerns for human health, primarily due to their potential bioaccumulative properties. While NPs have recently been detected in human blood, their interactions with specific immune cell subtypes and their impact on immune regulation remain unclear. In this proof-of-concept study, model palladium-doped polystyrene NPs (PS-Pd NPs) are utilized to enable single-cell mass cytometry (CyTOF) detection. The size-dependent impact of carboxylate polystyrene NPs (50-200 nm) is investigated across 15 primary immune cell subpopulations using CyTOF. By taking advantage of Pd-doping for detecting PS-Pd NPs, this work evaluates their impact on human immune-cells at the single-cell level following blood exposure. This work traces PS-Pd NPs in 37 primary immune-cell subpopulations from human blood, quantifying the palladium atom count per cell by CyTOF while simultaneously assessing the impact of PS-Pd NPs on cell viability, functionality, and uptake. These results demonstrate that NPs can interact with, interfere with, and translocate into several immune cell subpopulations after exposure. In vivo distribution experiments in mice further confirmed their accumulation in immune cells within the liver, blood, and spleen, particularly in monocytes, macrophages, and dendritic cells. These findings provide valuable insights into the impact of NPs on human health.
Collapse
Affiliation(s)
- Laura Fusco
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
| | - Arianna Gazzi
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
| | - Linda Giro
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
- Immunology Center of GeorgiaAugusta UniversityAugusta30912United States
| | - Roman B. Schefer
- Department of Environmental Systems ScienceETH ZürichZürich8092Switzerland
| | | | - Roberta Cagliani
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
| | - Martina Zoccheddu
- Immunology Center of GeorgiaAugusta UniversityAugusta30912United States
- Georgia Cancer Center Integrated Genomics coreAugusta UniversityAugusta30912United States
| | - Recep Uyar
- Stem Cell InstituteAnkara UniversityAnkara06100Turkey
| | - Ömur Besbinar
- Stem Cell InstituteAnkara UniversityAnkara06100Turkey
| | | | - Acelya Yilmazer
- Stem Cell InstituteAnkara UniversityAnkara06100Turkey
- Department of Biomedical EngineeringFaculty of EngineeringAnkara UniversityAnkara06100Turkey
| | - Denise M. Mitrano
- Department of Environmental Systems ScienceETH ZürichZürich8092Switzerland
| | - Marco Orecchioni
- Immunology Center of GeorgiaAugusta UniversityAugusta30912United States
- Department of Pharmacology & ToxicologyAugusta UniversityAugusta30912United States
| | - Lucia Gemma Delogu
- ImmuneNano‐labDepartment of Biomedical SciencesUniversity of PaduaPadua35131Italy
- Department of Biological SciencesKhalifa University of Science & TechnologyAbu Dhabi127788UAE
| |
Collapse
|
17
|
Ma M, Wang J, Guo K, Zhong W, Cheng Y, Lin L, Zhao Y. A Self-Reinforced "Microglia Energy Modulator" for Synergistic Amyloid-β Clearance in Alzheimer's Disease Model. Angew Chem Int Ed Engl 2025; 64:e202420547. [PMID: 39714451 DOI: 10.1002/anie.202420547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Microglial phagocytosis is a highly energy-consuming process that plays critical roles in clearing neurotoxic amyloid-β (Aβ) in Alzheimer's disease (AD). However, microglial metabolism is defective overall in AD, thereby undermining microglial phagocytic functions. Herein, we repurpose the existing antineoplastic drug lonidamine (LND) conjugated with hollow mesoporous Prussian blue (HMPB) as a "microglial energy modulator" (termed LND@HMPB-T7) for safe and synergistic Aβ clearance. The modified blood-brain barrier penetrating heptapeptide (T7) enables efficient transport of LND@HMPB-T7 to the AD brain. LND in LND@HMPB-T7 could fuel Aβ phagocytosis by stimulating microglial adenosine triphosphate (ATP) production, whereas HMPB with catalase and superoxide dismutase-mimicking activities substantially alleviates the mitochondrial side effects commonly associated with LND and thus further enhances ATP production. The synergism of LND and nanozyme affords a high microglial Aβ clearance efficacy without triggering mitochondrial dysfunction. In vivo experiments ascertain that LND@HMPB-T7 could synergistically promote phagocytic clearance of Aβ, relieve neuroinflammation and ameliorate cognitive function in AD mice. These findings indicate that LND@HMPB-T7 holds tremendous clinical potential as a repurposed drug for AD treatment.
Collapse
Affiliation(s)
- Mengmeng Ma
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang, Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Jing Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Kaiming Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenbin Zhong
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang, Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Yu Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang, Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang, Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| |
Collapse
|
18
|
Morsy HM, Zaky MY, Yassin NYS, Khalifa AYZ. Nanoparticle-based flavonoid therapeutics: Pioneering biomedical applications in antioxidants, cancer treatment, cardiovascular health, neuroprotection, and cosmeceuticals. Int J Pharm 2025; 670:125135. [PMID: 39732216 DOI: 10.1016/j.ijpharm.2024.125135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Flavonoids, a type of natural polyphenolic molecule, have garnered significant research interest due to their ubiquitous nature and diverse biological activities, including antioxidant, anti-inflammatory, and anticancer effects, making them appealing to various scientific disciplines. In this regard, the use of a flavonoid nanoparticle delivery system is to overcome low bioavailability, bioactivity, poor aqueous solubility, systemic absorption, and intensive metabolism. Therefore, this review summarizes the classification of nanoparticles (liposomes, polymeric, and solid lipid nanoparticles) and the advantages of using nanoparticle-flavonoid formulations to boost flavonoid bioavailability. Moreover, this review illustrated the pioneering biomedical applications of nanoparticle-based flavonoid therapeutics, as well as safety and toxicity considerations of using a flavonoid nanoparticle delivery system.
Collapse
Affiliation(s)
- Hadeer M Morsy
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Mohamed Y Zaky
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt.
| | - Nour Y S Yassin
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Ashraf Y Z Khalifa
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia.
| |
Collapse
|
19
|
Li H, Li C, Fu C, Wang Y, Liang T, Wu H, Wu C, Wang C, Sun T, Liu S. Innovative nanoparticle-based approaches for modulating neutrophil extracellular traps in diseases: from mechanisms to therapeutics. J Nanobiotechnology 2025; 23:88. [PMID: 39915767 PMCID: PMC11800495 DOI: 10.1186/s12951-025-03195-3] [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: 10/30/2024] [Accepted: 02/02/2025] [Indexed: 02/11/2025] Open
Abstract
Neutrophil extracellular traps (NETs) participate in both host defense and the pathogenesis of various diseases, such as infections, thrombosis, and tumors. While they help capture and eliminate pathogens, NETs' excessive or dysregulated formation can lead to tissue damage and disease progression. Therapeutic strategies targeting NET modulation have shown potential, but challenges remain, particularly in achieving precise drug delivery and maintaining drug stability. Nanoparticle (NP)-based drug delivery systems offer innovative solutions for overcoming the limitations of conventional therapies. This review explores the biological mechanisms of NET formation, their interactions with NPs, and the therapeutic applications of NP-based drug delivery systems for modulating NETs. We discuss how NPs can be designed to either promote or inhibit NET formation and provide a comprehensive analysis of their potential in treating NET-related diseases. Additionally, we address the current challenges and future prospects for NP-based therapies in NET research, aiming to bridge the gap between nanotechnology and NET modulation for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Haisong Li
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
- Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Can Li
- Department of Hematology, The Second Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Cong Fu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yizhuo Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tingting Liang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Haitao Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chenxi Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chang Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, Jilin, China.
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Shuhan Liu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| |
Collapse
|
20
|
Razack SA, Kim YE, Kang HW. κ-carrageenan - Gelatin hydrogel embedding carvacrol loaded gold nanobipyramids for treating prostate cancer via fractionated photothermal-chemotherapy. Int J Biol Macromol 2025; 291:138974. [PMID: 39710028 DOI: 10.1016/j.ijbiomac.2024.138974] [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: 09/30/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Conventional treatment of prostate cancer need more specificity, and higher efficiency. The present work is the first attempt to utilize hydrogel-loaded carvacrol-based chemotherapy with fractionated photothermal therapy (F-PTT) using a 635 nm laser for its treatment. Gold nanobipyramids (AuNBPs) were used as drug carrier and photosensitizer. A marine-derived hydrogel (AuNBP-HG) was fabricated and physicochemically characterized. A 635 nm NIR light was used for the fractionated PTT in three cycles of treatment on different days at varying power densities for 5 min. The efficiency of single and dual treatments was experimented in vitro and in vivo. The results showed that the dual therapy imparted a better effect than monotherapy. The AuNBP-CVL synthesized was nearly 50 nm and efficiently loaded within AuNBP-HG, displaying thermal responsiveness, a good sol-gel transition, a controlled drug release rate, and high stability. In vitro evaluation demonstrated that the F-PTT evinced stability during temperature rise without damaging healthy surrounding tissue. In vivo study revealed that a stable temperature rise up to 60 °C by the 635 nm NIR light due to photosensitization of AuNBP and sustained CVL release from AuNBP-HG into the tumor microenvironment exhibited successful thermal ablation of the cancer cells. Histological analysis confirmed that the uniform suspension of AuNBP within cancer tissues might have enhanced tumor cell lysis during the dual therapy. Western blot analysis exhibited that the cellular death could result from the upregulation of pJNK and p53 protein. In conclusion, the current study illustrated that dual therapy could be a feasible alternative to non-target specific and invasive traditional therapies against prostate cancer.
Collapse
Affiliation(s)
- Sirajunnisa Abdul Razack
- Center for Marine Integrated Bionics Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
| | - Yeong Eun Kim
- Biomedical Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Hyun Wook Kang
- Center for Marine Integrated Bionics Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
21
|
Kośnik W, Sikorska H, Kiciak A, Ciach T. Biodistribution of Polyaldehydedextran Nanoparticle-Encapsulated Epirubicin in Ovarian Tumor-Bearing Mice via Optical Imaging. Int J Mol Sci 2025; 26:970. [PMID: 39940738 PMCID: PMC11817601 DOI: 10.3390/ijms26030970] [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: 11/19/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
This study investigates the biodistribution of polysaccharide-based nanoparticles loaded with epirubicin (POLEPI) compared to epirubicin hydrochloride (EPI) in naïve female nude mice following a single intravenous dose. The inherent fluorescence of epirubicin was tracked using Newton 7 animal imager and Varioskan. Initial whole-animal optical imaging failed to reliably detect epirubicin distribution, necessitating ex vivo imaging of key tissues harvested at intervals between 10 min and 48 h post-injection. Optimal imaging conditions were established using a 5 s exposure time with excitation (Ex)/emission (Em) at 480 nm/550 nm. The biodistribution of POLEPI was further evaluated in both naïve mice and immunocompromised mice bearing patient-derived ovarian tumors. Unlike epirubicin, POLEPI exhibited notable tissue distribution within 3 h post-injection. By 48 h, fluorescence signals were undetectable in both models, although non-tumored animals exhibited persistent signals. In both models, the liver was the primary organ for POLEPI accumulation, with lower levels in tumored mice. Interestingly, brain fluorescence was higher in POLEPI-treated mice compared to those receiving epirubicin. Neither POLEPI nor epirubicin accumulated in the spleen or bone marrow. In tumors, POLEPI fluorescence peaked at 24 h, with levels 2.1 times higher than in the epirubicin-treated group over a 48 h period. Furthermore, POLEPI uptake in tumors exceeded that in healthy ovaries, with the most significant tumor-to-healthy-ovary ratio observed between 6 and 24 h post-injection. These findings demonstrate that POLEPI, a novel polyaldehydedextran nanoparticle formulation, exhibits enhanced accumulation and retention in tumor tissue compared to epirubicin, with preferential distribution to the orthotopic tumor-bearing ovary over healthy ovarian tissue. The inherent fluorescence of epirubicin provided a rapid and cost-effective means of estimating biodistribution, although the limitations of this method-particularly, the inability to differentiate between the parent drug and its metabolites-were acknowledged.
Collapse
Affiliation(s)
| | | | - Adam Kiciak
- NanoGroup S.A., Rakowiecka 36, 02-532 Warsaw, Poland
| | - Tomasz Ciach
- NanoVelos S.A., Rakowiecka 36, 02-532 Warsaw, Poland
- NanoGroup S.A., Rakowiecka 36, 02-532 Warsaw, Poland
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Waryńskiego 1, 00-645 Warsaw, Poland
| |
Collapse
|
22
|
Zohora FT, Pathmanathan R, Chowdhury EH. Application of Strontium Chloride Hexahydrate to Synthesize Strontium-Substituted Carbonate Apatite as a pH-Sensitive, Biologically Safe, and Highly Efficient siRNA Nanocarrier. ACS APPLIED BIO MATERIALS 2025; 8:348-367. [PMID: 39723844 DOI: 10.1021/acsabm.4c01319] [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: 12/28/2024]
Abstract
Naked siRNAs are sensitive to enzymatic degradation, phagocytic entrapment, quick renal excretion, membrane impermeability, endosomal escape, and off-target effects. Designing a safe and efficient nanocarrier for siRNA delivery to the target site without toxicity remains a significant hurdle in gene therapy. CA is a unique derivative of hydroxyapatite and a highly pH-sensitive nanocarrier with strong particle aggregation and a high polydispersity index. Strontium (Sr2+), a group two divalent metal in the periodic table, has been reported for substituting calcium (Ca2+) ions from the apatite lattice and limiting particle growth/aggregation. This study used strontium chloride hexahydrate (SrCl2·6H2O) salt to develop a Sr-substituted CA (Sr-CA) nanocarrier with ∼30 nm size, spherical shape, less aggregation, homodispersity, and a fair anionic charge. Sr-CA demonstrated a large surface area-to-volume ratio, an improved cargo loading efficiency, and enhanced cellular uptake in HEK-293 cells. Moreover, Sr-CA is a pH-responsive nanocarrier responsible for its long physiological stability, efficient endosomal escape, and optimal cargo delivery within cells. These NPs have differential effects on MAPK1, MAP2K4, PIK3Ca, CAMK4, and p53 gene expression in HEK-293 cells without showing any significant cytotoxicity in cell growth properties. Gene silencing by Sr-CA-mediated siRNA delivery against MAPK1, MAP2K4, PIK3Ca, and CAMK4 genes significantly decreased the level of target gene expression and cell survival, demonstrating successful intracellular siRNA delivery in HEK-293 cells. Additionally, biocompatibility testing confirmed the biological safety of the Sr-CA nanocarrier in mice. These findings suggest that Sr-CA nanocarriers are a promising siRNA delivery system, combining high efficiency with pH-sensitive release and excellent biocompatibility, making them a viable option for future therapeutic applications.
Collapse
Affiliation(s)
- Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Rajadurai Pathmanathan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
- Daffodil International University, Daffodil Smart City, Birulia 1216, Bangladesh
- Nanoflex LLC, 31756 Broadwater Avenue, Leesburg, Florida 34748, United States
| |
Collapse
|
23
|
Wahnou H, El Kebbaj R, Liagre B, Sol V, Limami Y, Duval RE. Curcumin-Based Nanoparticles: Advancements and Challenges in Tumor Therapy. Pharmaceutics 2025; 17:114. [PMID: 39861761 PMCID: PMC11768525 DOI: 10.3390/pharmaceutics17010114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Curcumin, a bioactive compound derived from the rhizome of Curcuma longa L., has garnered significant attention for its potent anticancer properties. Despite its promising therapeutic potential, its poor bioavailability, rapid metabolism, and low water solubility hinder curcumin's clinical application. Nanotechnology offers a viable solution to these challenges by enabling the development of curcumin-based nanoparticles (CNPs) that enhance its bioavailability and therapeutic efficacy. This review provides a comprehensive overview of the recent advancements in the design and synthesis of CNPs for cancer therapy. We discuss various NP formulations, including polymeric, lipid-based, and inorganic nanoparticles, highlighting their role in improving curcumin's pharmacokinetic and pharmacodynamic profiles. The mechanisms by which CNPs exert anticancer effects, such as inducing apoptosis, inhibiting cell proliferation, and modulating signaling pathways, are explored in details. Furthermore, we examine the preclinical and clinical studies that have demonstrated the efficacy of CNPs in treating different types of tumors, including breast, colorectal, and pancreatic cancers. Finally, the review addresses the current challenges and future perspectives in the clinical translation of CNPs, emphasizing the need for further research to optimize their design for targeted delivery and to enhance their therapeutic outcomes. By synthesizing the latest research, this review underscores the potential of CNPs as a promising avenue for advancing cancer therapy.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P2693, Maarif, Casablanca 20100, Morocco;
| | - Riad El Kebbaj
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco;
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (B.L.); (V.S.)
| | - Vincent Sol
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (B.L.); (V.S.)
| | - Youness Limami
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco;
| | | |
Collapse
|
24
|
Amaeze O, Isoherranen N, Shum S. The absorption, distribution, metabolism and elimination characteristics of small interfering RNA therapeutics and the opportunity to predict disposition in pregnant women. Drug Metab Dispos 2025; 53:100018. [PMID: 39884813 DOI: 10.1124/dmd.123.001383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/19/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024] Open
Abstract
Small interfering RNA (siRNA) therapeutics represent an emerging class of pharmacotherapy with the potential to address previously hard-to-treat diseases. Currently approved siRNA therapeutics include lipid nanoparticle-encapsulated siRNA and tri-N-acetylated galactosamine-conjugated siRNA. These siRNA therapeutics exhibit distinct pharmacokinetic characteristics and unique absorption, distribution, metabolism, and elimination (ADME) properties. As a new drug modality, limited clinical data are available for siRNA therapeutics in specific populations, including pediatrics, geriatrics, individuals with renal or hepatic impairment, and pregnant women, making dosing challenging. In this Minireview, a mechanistic overview of the ADME properties of the 5 currently approved siRNA therapeutics is presented. A concise overview of the clinical data available for therapeutic siRNAs in special populations, focusing on the potential impact of physiologic changes during pregnancy on siRNA disposition, is provided. The utility of physiologically based pharmacokinetic (PBPK) modeling as a tool to elucidate the characteristics and disposition of siRNA therapeutics in pregnant women is explored. Additionally, opportunities to integrate known physiologic alterations induced by pregnancy into PBPK models that incorporate siRNA ADME mechanisms to predict the effects of pregnancy on siRNA disposition are discussed. Clinical data regarding the use of therapeutic siRNA in special populations remain limited. Data for precise parameterization of maternal-fetal siRNA PBPK models are lacking presently and underscore the need for further research in this area. Addressing this gap in knowledge will not only enhance our understanding of siRNA pharmacokinetics during pregnancy but also advance the possible development of siRNA therapeutics to treat pregnancy-related conditions. SIGNIFICANCE STATEMENT: This Minireview proposes a framework on how small interfering RNA (siRNA) disposition can be predicted in pregnancy based on mechanistic absorption, distribution, metabolism, and elimination (ADME) information using physiologically-based pharmacokinetic (PBPK) modeling. The mechanistic ADME information and available clinical data in special populations of currently Food and Drug Administration-approved siRNA therapeutics are summarized. Additionally, how physiological changes during pregnancy may affect siRNA disposition is reviewed, and the opportunities to use PBPK modeling to predict siRNA disposition in pregnant women is explored.
Collapse
Affiliation(s)
- Ogochukwu Amaeze
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Sara Shum
- ReNAgade Therapeutics Management Inc, Cambridge, Massachusetts.
| |
Collapse
|
25
|
Yuan S, Zhu L, Luo Y, Chen X, Jing H, Wang J, Su X, Liang M, Zhuang Z. Igniting tumour microenvironment in triple-negative breast cancer using a mannose/hyaluronic acid dual-coated Ganoderma polysaccharide-superparamagnetic iron oxide nanocomplex for combinational therapies. J Drug Target 2025; 33:111-126. [PMID: 39470031 DOI: 10.1080/1061186x.2024.2408721] [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: 08/31/2024] [Accepted: 09/21/2024] [Indexed: 10/30/2024]
Abstract
Eliciting tumour microenvironment (TME) activation in triple-negative breast cancer (TNBC) is crucial for effective anti-tumour therapies. The aim of this study is to employ pharmaceutical approaches to precisely deliver Ganoderma polysaccharide (GPS) to tumour sites, thereby enhancing TME activation. We first established a direct link between the accumulation of GPS within tumours and its efficacy in the TME activation. Building upon this insight, we then engineered a mannose/hyaluronic acid dual-coated GPS-loaded superparamagnetic iron oxide nanocomplex (Man/HA/GPS-SPIONs) with a particle size of 33.8 ± 1.6 nm and a zeta potential of -22.4 ± 3.5 mV, capable of precise tumour accumulation through magnet-assisted targeting and internalisation by tumour-associated macrophages (TAMs) and tumour cells, facilitated by dual ligand modification. In vitro, Man/HA/GPS-SPIONs effectively induced M1 polarisation of macrophages (CD86+ cells: 38.6 ± 2.8%), curbed 4T1 cell proliferation (viability: 47.3 ± 2.9%) and heightened Th1 cytokine release. Significantly, in vivo, Man/HA/GPS-SPIONs notably suppressed tumour growth (tumour index: 0.048 ± 0.005), fostered M1 polarisation of TAMs (CD45+F4/80+CD86+ cells: 26.1 ± 7.2%), consequently bolstering intratumoural T cytotoxic cells. This enhancement was intricately tied to the efficient co-delivery of GPS and iron ions to the tumours, made possible by the Man/HA/GPS-SPIONs delivery system. The synergistic effects with paclitaxel (PTX, inhibition rate: 61.2 ± 4.3%) and PD-1 inhibitors (inhibition rate: 69.8 ± 7.6%) underscored the translational potential of this approach. By harnessing a well-conceived iron-based drug delivery strategy, this study amplifies the tumour immune modulatory potential of natural polysaccharides, offering insightful guidance for interventions in the TME and synergistic therapies.
Collapse
Affiliation(s)
- Shaofei Yuan
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, P.R. China
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Linjia Zhu
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Yi Luo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaoqiang Chen
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Haibo Jing
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Jiaqi Wang
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Xiangyu Su
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Meizhen Liang
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, P.R. China
| |
Collapse
|
26
|
Klika K, Han J, Busse MS, Soloshonok VA, Javahershenas R, Vanhaecke F, Makarem A. Inductively Coupled Plasma-Mass Spectrometry (ICP-MS): An Emerging Tool in Radiopharmaceutical Science. J Am Chem Soc 2024; 146:30717-30727. [PMID: 39478417 PMCID: PMC11565647 DOI: 10.1021/jacs.4c12254] [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: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/14/2024]
Abstract
Although radioactive experiments are necessary in radiopharmaceutical drug discovery and theranostic cancer research, they are expensive, require special facilities, and face certain restrictions. Thus, finding techniques not involving radioactivity is highly beneficial for minimizing these disadvantages in such research. In this regard, methods using inductively coupled plasma-mass spectrometry (ICP-MS) have emerged as viable alternatives to traditional radioactive approaches. Despite its potential, practical applications of ICP-MS in radiopharmaceutical cancer research have only emerged in recent years. This Perspective focuses on the development and implementation of nonradioactive ICP-MS-based assays in radiopharmaceutical research and aims to inspire future research efforts in this area.
Collapse
Affiliation(s)
- Karel
D. Klika
- Molecular
Structure Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jianlin Han
- College
of Chemical Engineering, Nanjing Forestry
University, 210037 Nanjing, China
| | - Marvin S. Busse
- Institute
of Pharmacy, University of Hamburg, 20146 Hamburg, Germany
| | - Vadim A. Soloshonok
- Department
of Organic Chemistry I, University of the
Basque Country, 20018 San Sebastián, Spain
- IKERBASQUE,
Basque Foundation for Science, 48009 Bilbao, Spain
| | - Ramin Javahershenas
- Department
of Organic Chemistry, Faculty of Chemistry, Urmia University, 57179-44514 Urmia, Iran
| | - Frank Vanhaecke
- Atomic
and Mass Spectrometry − A&MS Research Unit, Department
of Chemistry, Ghent University, 9000 Ghent, Belgium
| | - Ata Makarem
- Institute
of Pharmacy, University of Hamburg, 20146 Hamburg, Germany
| |
Collapse
|
27
|
Zhang LK, Li Y, Zhai L, Tang Y, Jiao Y, Mei Y, Yang R, You R, Yin L, Ni H, Ge J, Guan YQ. Natural Phycocyanin/Paclitaxel Micelle Delivery of Therapeutic P53 to Activate Apoptosis for HER2 or ER Positive Breast Cancer Therapy. ACS Biomater Sci Eng 2024; 10:6995-7004. [PMID: 39390952 DOI: 10.1021/acsbiomaterials.4c00756] [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: 10/12/2024]
Abstract
The P53 gene is commonly mutated in breast cancer, protein based the gene as anticancer drugs could provide efficient and stable advantages by restoring the function of the wild-type P53 protein. In this study, we describe the creation and utilization of a micelle composed by natural phycocyanin and paclitaxel and grafting anti-HER2 (PPH), which effectively packages and transports recombinant P53 protein with anti-ER (PE), resulting in a new entity designated as PE@PPH, to address localization obstacles and modify cellular tropism to the cell membrane or nucleus. The results indicate that PE@PPH has strong antitumor properties, even at low doses of PTX both in vitro and in vivo. These findings suggest that PE@PPH could be an enhancing micelle for delivering therapeutic proteins and promoting protein functional recovery, particularly in addressing the challenges posed by tumor heterogeneity in breast cancer.
Collapse
Affiliation(s)
- Ling-Kun Zhang
- School of Life Science, South China Normal University, Guangzhou 510631, China
- School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Yuan Li
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Limin Zhai
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yunzhi Tang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yuxuan Jiao
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yitong Mei
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Runcai Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Rong You
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - He Ni
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou 510631, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
28
|
Pashirova T, Shaihutdinova Z, Tatarinov D, Titova A, Malanyeva A, Vasileva O, Gabdurakhmanov K, Dudnikov S, Schopfer LM, Lockridge O, Masson P. Pharmacokinetics and fate of free and encapsulated IRD800CW-labelled human BChE intravenously administered in mice. Int J Biol Macromol 2024; 282:137305. [PMID: 39515732 DOI: 10.1016/j.ijbiomac.2024.137305] [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: 08/06/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Human butyrylcholinesterase (BChE) is an efficient bioscavenger of toxicants. Highly purified BChE was labelled with the near infrared fluorescent IRDye800CW. The goal was to determine the pharmacokinetics and fate of enzyme in mice. BChE-IRDye800CW was encapsulated in polyethylene glycol-polypropylene sulfide-based spherical polymersome nanoreactors with the following characteristics: 140 nm diameter, ξ = -6 mV, PDI ≤ 0.2, 1 year stability. Encapsulation did not alter the functional properties of BChE. Free and encapsulated enzyme were injected intravenously to CD-1 mice (single dose of enzyme 1.5 mg/kg and PEG-PPS polymersomes 25 mg/kg) and were analyzed for 8 days using an in vivo imaging system. Results showed that the pharmacokinetic distribution α-phase of encapsulated BChE (t1/2 = 17.6 h) was longer than for free enzyme (t1/2 = 6.6 h). The mean half-time for elimination β-phase was 2-time longer for encapsulated enzyme than for free enzyme (150 vs 72 h). Transient changes in infrared fluorescence in organs showed that BChE is eliminated from liver. However, free and encapsulated enzymes were cleared via different pathways. This first study of pharmacokinetics and fate of BChE encapsulated in polymersomes initiates research of new formulations of bioscavengers aimed at increasing the residence time of enzymes in the blood stream.
Collapse
Affiliation(s)
- Tatiana Pashirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation; Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation.
| | - Zukhra Shaihutdinova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation; Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation
| | - Dmitry Tatarinov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation
| | - Angelina Titova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Albina Malanyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Olga Vasileva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Kamil Gabdurakhmanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Sergei Dudnikov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | | | - Oksana Lockridge
- University of Nebraska Medical Center, Eppley Institute, Omaha, NE, USA
| | - Patrick Masson
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation.
| |
Collapse
|
29
|
Macone A, Cappelletti C, Incocciati A, Piacentini R, Botta S, Boffi A, Bonamore A. Challenges in Exploiting Human H Ferritin Nanoparticles for Drug Delivery: Navigating Physiological Constraints. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2016. [PMID: 39541599 DOI: 10.1002/wnan.2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/14/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Over the past two decades, ferritin has emerged as a promising nanoparticle for drug delivery, catalyzing the development of numerous prototypes capable of encapsulating a wide array of therapeutic agents. These ferritin-based nanoparticles exhibit selectivity for various molecular targets and are distinguished by their potential biocompatibility, unique symmetrical structure, and highly controlled size. The hollow interior of ferritin nanoparticles allows for efficient encapsulation of diverse therapeutic agents, enhancing their delivery and effectiveness. Despite these promising features, the anticipated clinical advancements have yet to be fully realized. As a physiological protein with a central role in both health and disease, ferritin can exert unexpected effects on physiology when employed as a drug delivery system. Many studies have not thoroughly evaluated the pharmacokinetic properties of the ferritin protein shell when administered in vivo, overlooking crucial aspects such as biodistribution, clearance, cellular trafficking, and immune response. Addressing these challenges is crucial for achieving the desired transition from bench to bedside. Biodistribution studies need to account for ferritin's natural accumulation in specific organs (liver, spleen, and kidneys), which may lead to off-target effects. Moreover, the mechanisms of clearance and cellular trafficking must be elucidated to optimize the delivery and reduce potential toxicity of ferritin nanoparticles. Additionally, understanding the immune response elicited by exogenous ferritin is essential to mitigate adverse reactions and enhance therapeutic efficacy. A comprehensive understanding of these physiological constraints, along with innovative solutions, is essential to fully realize the therapeutic potential of ferritin nanoparticles paving the way for their successful clinical translation.
Collapse
Affiliation(s)
- Alberto Macone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Chiara Cappelletti
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessio Incocciati
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Roberta Piacentini
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Sofia Botta
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alberto Boffi
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessandra Bonamore
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
30
|
Wang H, Wang X, Wang L, Wang H, Zhang Y. Exploiting lignin-based nanomaterials for enhanced anticancer therapy: A comprehensive review and future direction. Int J Biol Macromol 2024; 281:136266. [PMID: 39366596 DOI: 10.1016/j.ijbiomac.2024.136266] [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: 05/27/2024] [Revised: 09/18/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Lignin, a renewable and abundant natural polymer, has emerged as a promising candidate for anticancer therapy due to its unique properties and biocompatibility. This review provides a comprehensive overview of recent advancements in the utilization of lignin-based nanomaterials for enhancing anticancer drug delivery and therapeutic outcomes. A detailed examination of the literature reveals several synthesis methods, including nanoprecipitation, microemulsion, and solvent exchange, which produce lignin nanoparticles with improved drug solubility and bioavailability. The anticancer mechanisms of lignin nanoparticles, such as the generation of reactive oxygen species (ROS), induction of apoptosis, and enhanced cellular uptake, are also explored. Lignin nanoparticles loaded with drugs like curcumin, doxorubicin, camptothecin, and resveratrol have demonstrated the ability to improve drug efficacy, selectively target cancer cells, overcome multidrug resistance, and minimize toxicity in both in vitro and in vivo studies. These nanoparticles have shown significant potential in suppressing tumor growth, inducing cell death through apoptotic pathways, and enhancing the synergistic effects of combination therapies, such as chemo-phototherapy. Future research directions include optimizing lignin nanoparticle formulations for clinical applications, refining targeted delivery mechanisms to cancer cells, and conducting thorough biocompatibility and toxicity assessments. Overall, this review highlights the significant progress made in utilizing lignin-based nanomaterials for cancer therapy and outlines promising areas for further exploration in this rapidly evolving field.
Collapse
Affiliation(s)
- Haoyu Wang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China; Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaoyang Wang
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Long Wang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China
| | - Haifan Wang
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yuxing Zhang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China.
| |
Collapse
|
31
|
Subhasri D, Leena MM, Moses JA, Anandharamakrishnan C. Factors affecting the fate of nanoencapsulates post administration. Crit Rev Food Sci Nutr 2024; 64:11949-11973. [PMID: 37599624 DOI: 10.1080/10408398.2023.2245462] [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: 08/22/2023]
Abstract
Nanoencapsulation has found numerous applications in the food and nutraceutical industries. Micro and nanoencapsulated forms of bioactives have proven benefits in terms of stability, release, and performance in the body. However, the encapsulated ingredient is often subjected to a wide range of processing conditions and this is followed by storage, consumption, and transit along the gastrointestinal tract. A strong understanding of the fate of nanoencapsulates in the biological system is mandatory as it provides valuable insights for ingredient selection, formulation, and application. In addition to their efficacy, there is also the need to assess the safety of ingested nanoencapsulates. Given the rising research and commercial focus of this subject, this review provides a strong focus on their interaction factors and mechanisms, highlighting their prospective biological fate. This review also covers various approaches to studying the fate of nanoencapsulates in the body. Also, with emphasis on the overall scope, the need for a new advanced integrated common methodology to evaluate the fate of nanoencapsulates post-administration is discussed.
Collapse
Affiliation(s)
- D Subhasri
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology Entrepreneurship and Management - Thanjavur, Ministry of Food Processing Industries, Government of India, Thanjavur, India
| | - M Maria Leena
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology Entrepreneurship and Management - Thanjavur, Ministry of Food Processing Industries, Government of India, Thanjavur, India
- Department of Biotechnology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Tiruchirappalli, India
| | - J A Moses
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology Entrepreneurship and Management - Thanjavur, Ministry of Food Processing Industries, Government of India, Thanjavur, India
| | - C Anandharamakrishnan
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology Entrepreneurship and Management - Thanjavur, Ministry of Food Processing Industries, Government of India, Thanjavur, India
- CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Ministry of Science and Technology, Government of India, Industrial Estate PO, Thiruvananthapuram, INDIA
| |
Collapse
|
32
|
Chang X, Liu J, Li Y, Li W. Pluronic F127-Complexed PEGylated Poly(glutamic acid)-Cisplatin Nanomedicine for Enhanced Glioblastoma Therapy. Macromol Rapid Commun 2024; 45:e2400662. [PMID: 39264576 DOI: 10.1002/marc.202400662] [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] [Indexed: 09/13/2024]
Abstract
Glioblastoma is one of the most aggressive and treatment-resistant forms of primary brain cancer, posing significant challenges in effective therapy. This study aimed to enhance the effectiveness of glioblastoma therapy by developing a unique nanomedicine composed of Pluronic F127-complexed PEGylated poly(glutamic acid)-cisplatin (PLG-PEG/PF127-CDDP). PLG-PEG/PF127-CDDP demonstrated an optimal size of 133.97 ± 12.60 nm, facilitating efficient cell uptake by GL261 glioma cells. In vitro studies showed significant cytotoxicity against glioma cells with a half-maximal (50%) inhibitory concentration (IC50) of 12.61 µg mL-1 at 48 h and a 72.53% ± 1.89% reduction in cell invasion. Furthermore, PLG-PEG/PF127-CDDP prolonged the circulation half-life of cisplatin to 9.75 h in vivo, leading to a more than 50% reduction in tumor size on day 16 post-treatment initiation in a murine model of glioma. The treatment significantly elevated lactate levels in GL261 cells, indicating enhanced metabolic disruption. Therefore, PLG-PEG/PF127-CDDP offers a promising approach for glioblastoma therapy due to its effects on improving drug delivery efficiency, therapeutic outcomes, and safety while minimizing systemic side effects. This work underscores the potential of polymer-based nanomedicines in overcoming the challenges of treating brain tumors, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Xiaoyu Chang
- Department of Neurosurgery, the First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
| | - Jiaxue Liu
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, 5 Jilin Street, Jilin, 132000, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Yunqian Li
- Department of Neurosurgery, the First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
| | - Wenliang Li
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, 5 Jilin Street, Jilin, 132000, P. R. China
| |
Collapse
|
33
|
Benyettou F, Khair M, Prakasam T, Varghese S, Matouk Z, Alkaabi M, Pena-Sánchez P, Boitet M, AbdulHalim R, Sharma SK, Ghemrawi R, Thomas S, Whelan J, Pasricha R, Jagannathan R, Gándara F, Trabolsi A. cRGD-Peptide Modified Covalent Organic Frameworks for Precision Chemotherapy in Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56676-56695. [PMID: 39267454 PMCID: PMC11503616 DOI: 10.1021/acsami.4c10812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024]
Abstract
This study presents the use of nanoscale covalent organic frameworks (nCOFs) conjugated with tumor-targeting peptides for the targeted therapy of triple-negative breast cancer (TNBC). While peptides have previously been used for targeted delivery, their conjugation with COFs represents an innovative approach in this field. In particular, we have developed alkyne-functionalized nCOFs chemically modified with cyclic RGD peptides (Alkyn-nCOF-cRGD). This configuration is designed to specifically target αvβ3 integrins that are overexpressed in TNBC cells. These nCOFs exhibit excellent biocompatibility and are engineered to selectively disintegrate under acidic conditions, allowing for precise and localized drug release in tumor environment. Doxorubicin, a chemotherapeutic agent, has been encapsulated in these nCOFs with high loading efficiency. The therapeutic potential of Alkyn-nCOF-cRGD has been demonstrated in vitro and in vivo models. It shows significantly improved drug uptake and targeted cell death in TNBC, highlighting the efficacy of receptor-mediated endocytosis and pH-controlled drug release. This strategy leverages the unique properties of nCOFs with targeted drug delivery to achieve significant advances in personalized cancer therapy and set a new standard for precision chemotherapeutic delivery.
Collapse
Affiliation(s)
- Farah Benyettou
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Mostafa Khair
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Thirumurugan Prakasam
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Sabu Varghese
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Zineb Matouk
- Technology
Innovative Institute, P.O. Box 9639, Abu Dhabi 9639, United Arab Emirates
| | - Maryam Alkaabi
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Pilar Pena-Sánchez
- Instituto
de Ciencia de Materiales de Madrid-CSIC, C. Sor Juana Inés de La Cruz 3, Madrid 28049, Spain
| | - Maylis Boitet
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Rasha AbdulHalim
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Sudhir Kumar Sharma
- Engineering
Division, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Rose Ghemrawi
- College
of Pharmacy, Al Ain University, P.O. Box 112612, Abu Dhabi 112612, United Arab Emirates
- AAU
Health and Biomedical Research Center, Al
Ain University, P.O. Box 112612, Abu Dhabi 112612, United Arab Emirates
| | - Sneha Thomas
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Jamie Whelan
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Renu Pasricha
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Ramesh Jagannathan
- Engineering
Division, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Felipe Gándara
- Instituto
de Ciencia de Materiales de Madrid-CSIC, C. Sor Juana Inés de La Cruz 3, Madrid 28049, Spain
| | - Ali Trabolsi
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| |
Collapse
|
34
|
Vasileva O, Zaborova O, Shmykov B, Ivanov R, Reshetnikov V. Composition of lipid nanoparticles for targeted delivery: application to mRNA therapeutics. Front Pharmacol 2024; 15:1466337. [PMID: 39508050 PMCID: PMC11537937 DOI: 10.3389/fphar.2024.1466337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/10/2024] [Indexed: 11/08/2024] Open
Abstract
Today, lipid nanoparticles (LNPs) are some of the main delivery systems for mRNA-based therapeutics. The scope of LNP applications in terms of RNA is not limited to antiviral vaccines but encompasses anticancer drugs and therapeutics for genetic (including rare) diseases. Such widespread use implies high customizability of targeted delivery of LNPs to specific organs and tissues. This review addresses vector-free options for targeted delivery of LNPs, namely the influence of lipid composition of these nanoparticles on their biodistribution. In the review, experimental studies are examined that are focused on the biodistribution of mRNA or of the encoded protein after mRNA administration via LNPs in mammals. We also performed a comprehensive analysis of individual lipids' functional groups that ensure biodistribution to desired organs. These data will allow us to outline prospects for further optimization of lipid compositions of nanoparticles for targeted delivery of mRNA therapeutics.
Collapse
Affiliation(s)
- Olga Vasileva
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Olga Zaborova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
- Chemistry Department, Moscow State University, Moscow, Russia
| | - Bogdan Shmykov
- Chemistry Department, Moscow State University, Moscow, Russia
| | - Roman Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Vasiliy Reshetnikov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
35
|
Selc M, Macova R, Babelova A. Novel Strategies Enhancing Bioavailability and Therapeutical Potential of Silibinin for Treatment of Liver Disorders. Drug Des Devel Ther 2024; 18:4629-4659. [PMID: 39444787 PMCID: PMC11498047 DOI: 10.2147/dddt.s483140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Silibinin, a bioactive component found in milk thistle extract (Silybum marianum), is known to have significant therapeutic potential in the treatment of various liver diseases. It is considered a key element of silymarin, which is traditionally used to support liver function. The main mechanisms of action of silibinin are attributed to its antioxidant properties protecting liver cells from damage caused by free radicals. Experimental studies conducted in vitro and in vivo have confirmed its ability to inhibit inflammatory and fibrotic processes, as well as promote the regeneration of damaged liver tissue. Therefore, silibinin represents a promising tool for the treatment of liver diseases. Since the silibinin molecule is insoluble in water and has poor bioavailability in vivo, new perspectives on solving this problem are being sought. The two most promising approaches are the water-soluble derivative silibinin-C-2',3-dihydrogen succinate, disodium salt, and the silibinin-phosphatidylcholine complex. Both drugs are currently under evaluation in liver disease clinical trials. Nevertheless, the mechanism underlying silibinin biological activity is still elusive and its more detailed understanding would undoubtedly increase its potential in the development of effective therapeutic strategies against liver diseases. This review is focused on the therapeutic potential of silibinin and its derivates, approaches to increase the bioavailability and the benefits in the treatment of liver diseases that have been achieved so far. The review discusses the relevant in vitro and in vivo studies that investigated the protective effects of silibinin in various forms of liver damage.
Collapse
Affiliation(s)
- Michal Selc
- Centre for Advanced Material Application, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Radka Macova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Genetics, Faculty of Natural Sciences, Comenius University Bratislava, Bratislava, Slovakia
| | - Andrea Babelova
- Centre for Advanced Material Application, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
36
|
Yang T, Zhang N, Liu Y, Yang R, Wei Z, Liu F, Song D, Wang L, Wei J, Li Y, Shen D, Liang G. Nanoplatelets modified with RVG for targeted delivery of miR-375 and temozolomide to enhance gliomas therapy. J Nanobiotechnology 2024; 22:623. [PMID: 39402578 PMCID: PMC11476726 DOI: 10.1186/s12951-024-02895-6] [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: 06/10/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Gliomas are one of the most frequent primary brain tumors and pose a serious threat to people's lives and health. Platelets, a crucial component of blood, have been applied as drug delivery carriers for disease diagnosis and treatment. In this study, we designed engineered nanoplatelets for targeted delivery of therapeutic miR-375 and temozolomide (TMZ, a first-line glioma treatment agent) to enhance glioma therapy. Nanoplatelets were prepared through mild ultrasound, TMZ and miR-375 were co-loaded through ultrasound and electrostatic interactions, respectively, to combine chemotherapy with gene therapy against glioma. To improve the blood brain barrier (BBB) crossing efficiency and glioma targeting ability, the nanoplatelets were modified with central nervous system-specific rabies viral glycoprotein peptide (RVG) through thiol-maleimide click reaction. The RVG modified nanoplatelets co-loaded TMZ and miR-375 (NR/TMZ/miR-375) not only inherited the good stability and remarkable biocompatibility of platelets, but also promoted the cellular uptake and penetration of glioma tissues, and effectively induced cell apoptosis to enhance the therapeutic effect of drugs. In vivo studies showed that NR/TMZ/miR-375 significantly increased the circulation time of TMZ, and exhibited superior combined antitumor effects. In summary, this multifunctional 'natural' nanodrug delivery system provides a potent, scalable, and safety approach for platelet-based combined cancer chemotherapy and gene therapy.
Collapse
Affiliation(s)
- Tingting Yang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
- Zhumadian Cental Hospital, Zhumadian, 463000, China
| | - Nan Zhang
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Yuanyuan Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Ruyue Yang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Zhaoyi Wei
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Futai Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Dan Song
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Longwei Wang
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Jiangyan Wei
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Yuanpei Li
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Deliang Shen
- Key Laboratory of Cardiac Injury and Repair of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Gaofeng Liang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China.
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China.
| |
Collapse
|
37
|
Mi K, Chou WC, Chen Q, Yuan L, Kamineni VN, Kuchimanchi Y, He C, Monteiro-Riviere NA, Riviere JE, Lin Z. Predicting tissue distribution and tumor delivery of nanoparticles in mice using machine learning models. J Control Release 2024; 374:219-229. [PMID: 39146980 PMCID: PMC11886896 DOI: 10.1016/j.jconrel.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/24/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
Nanoparticles (NPs) can be designed for targeted delivery in cancer nanomedicine, but the challenge is a low delivery efficiency (DE) to the tumor site. Understanding the impact of NPs' physicochemical properties on target tissue distribution and tumor DE can help improve the design of nanomedicines. Multiple machine learning and artificial intelligence models, including linear regression, support vector machine, random forest, gradient boosting, and deep neural networks (DNN), were trained and validated to predict tissue distribution and tumor delivery based on NPs' physicochemical properties and tumor therapeutic strategies with the dataset from Nano-Tumor Database. Compared to other machine learning models, the DNN model had superior predictions of DE to tumors and major tissues. The determination coefficients (R2) for the test datasets were 0.41, 0.42, 0.45, 0.79, 0.87, and 0.83 for DE in tumor, heart, liver, spleen, lung, and kidney, respectively. All the R2 and root mean squared error (RMSE) results of the test datasets were similar to the 5-fold cross validation results. Feature importance analysis showed that the core material of NPs played an important role in output predictions among all physicochemical properties. Furthermore, multiple NP formulations with greater DE to the tumor were determined by the DNN model. To facilitate model applications, the final model was converted to a web dashboard. This model could serve as a high-throughput pre-screening tool to support the design of new and efficient nanomedicines with greater tumor DE and serve as an alternative tool to reduce, refine, and partially replace animal experimentation in cancer nanomedicine research.
Collapse
Affiliation(s)
- Kun Mi
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Wei-Chun Chou
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA; Department of Environmental Sciences, College of Natural & Agricultural Sciences, University of California, Riverside, CA 92521, USA
| | - Qiran Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Long Yuan
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Venkata N Kamineni
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Yashas Kuchimanchi
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Chunla He
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA
| | - Nancy A Monteiro-Riviere
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS 66506, USA; Center for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, NC 27606, USA
| | - Jim E Riviere
- Center for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, NC 27606, USA; 1Data Consortium, Kansas State University, Olathe, KS 66061, USA
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
38
|
Andreani T, Cheng R, Elbadri K, Ferro C, Menezes T, Dos Santos MR, Pereira CM, Santos HA. Natural compounds-based nanomedicines for cancer treatment: Future directions and challenges. Drug Deliv Transl Res 2024; 14:2845-2916. [PMID: 39003425 PMCID: PMC11385056 DOI: 10.1007/s13346-024-01649-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2024] [Indexed: 07/15/2024]
Abstract
Several efforts have been extensively accomplished for the amelioration of the cancer treatments using different types of new drugs and less invasives therapies in comparison with the traditional therapeutic modalities, which are widely associated with numerous drawbacks, such as drug resistance, non-selectivity and high costs, restraining their clinical response. The application of natural compounds for the prevention and treatment of different cancer cells has attracted significant attention from the pharmaceuticals and scientific communities over the past decades. Although the use of nanotechnology in cancer therapy is still in the preliminary stages, the application of nanotherapeutics has demonstrated to decrease the various limitations related to the use of natural compounds, such as physical/chemical instability, poor aqueous solubility, and low bioavailability. Despite the nanotechnology has emerged as a promise to improve the bioavailability of the natural compounds, there are still limited clinical trials performed for their application with various challenges required for the pre-clinical and clinical trials, such as production at an industrial level, assurance of nanotherapeutics long-term stability, physiological barriers and safety and regulatory issues. This review highlights the most recent advances in the nanocarriers for natural compounds secreted from plants, bacteria, fungi, and marine organisms, as well as their role on cell signaling pathways for anticancer treatments. Additionally, the clinical status and the main challenges regarding the natural compounds loaded in nanocarriers for clinical applications were also discussed.
Collapse
Affiliation(s)
- Tatiana Andreani
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
- GreenUPorto-Sustainable Agrifood Production Research Centre & Inov4Agro, Department of Biology, Faculty of Sciences of University of Porto, Rua Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Ruoyu Cheng
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute Groningen (PRECISION), University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Khalil Elbadri
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Claudio Ferro
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Research Institute for Medicines, iMed.Ulisboa, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Thacilla Menezes
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Mayara R Dos Santos
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Carlos M Pereira
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland.
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute Groningen (PRECISION), University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
39
|
Ren M, Abdullah SW, Pei C, Guo H, Sun S. Use of virus-like particles and nanoparticle-based vaccines for combating picornavirus infections. Vet Res 2024; 55:128. [PMID: 39350170 PMCID: PMC11443892 DOI: 10.1186/s13567-024-01383-x] [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: 04/17/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
Picornaviridae are non-enveloped ssRNA viruses that cause diseases such as poliomyelitis, hand-foot-and-mouth disease (HFMD), hepatitis A, encephalitis, myocarditis, and foot-and-mouth disease (FMD). Virus-like particles (VLPs) vaccines mainly comprise particles formed through the self-assembly of viral capsid proteins (for enveloped viruses, envelope proteins are also an option). They do not contain the viral genome. On the other hand, the nanoparticles vaccine (NPs) is mainly composed of self-assembling biological proteins or nanomaterials, with viral antigens displayed on the surface. The presentation of viral antigens on these particles in a repetitive array can elicit a strong immune response in animals. VLPs and NPs can be powerful platforms for multivalent antigen presentation. This review summarises the development of virus-like particle vaccines (VLPs) and nanoparticle vaccines (NPs) against picornaviruses. By detailing the progress made in the fight against various picornaviruses such as poliovirus (PV), foot-and-mouth disease virus (FMDV), enterovirus (EV), Senecavirus A (SVA), and encephalomyocarditis virus (EMCV), we in turn highlight the significant strides made in vaccine technology. These advancements include diverse construction methods, expression systems, elicited immune responses, and the use of various adjuvants. We see promising prospects for the continued development and optimisation of VLPs and NPs vaccines. Future research should focus on enhancing these vaccines' immunogenicity, stability, and delivery methods. Moreover, expanding our understanding of the interplay between these vaccines and the immune system will be crucial. We hope these insights will inspire and guide fellow researchers in the ongoing quest to combat picornavirus infections more effectively.
Collapse
Affiliation(s)
- Mei Ren
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gembloux Agro-Biotech, University of Liege, Gembloux, Belgium
| | - Sahibzada Waheed Abdullah
- Livestock and dairy development department peshawar, Government of Khyber Pakhtunkhwa, Peshawar, Pakistan
| | - Chenchen Pei
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
| |
Collapse
|
40
|
Desai DA, Schmidt S, Cristofoletti R. A quantitative systems pharmacology (QSP) platform for preclinical to clinical translation of in-vivo CRISPR-Cas therapy. Front Pharmacol 2024; 15:1454785. [PMID: 39372210 PMCID: PMC11449743 DOI: 10.3389/fphar.2024.1454785] [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] [Accepted: 08/30/2024] [Indexed: 10/08/2024] Open
Abstract
Background: In-vivo CRISPR Cas genome editing is a complex therapy involving lipid nanoparticle (LNP), messenger RNA (mRNA), and single guide RNA (sgRNA). This novel modality requires prior modeling to predict dose-exposure-response relationships due to limited information on sgRNA and mRNA biodistribution. This work presents a QSP model to characterize, predict, and translate the Pharmacokinetics/Pharmacodynamics (PK/PD) of CRISPR therapies from preclinical species (mouse, non-human primate (NHP)) to humans using two case studies: transthyretin amyloidosis and LDL-cholesterol reduction. Methods: PK/PD data were sourced from literature. The QSP model incorporates mechanisms post-IV injection: 1) LNP binding to opsonins in liver vasculature; 2) Phagocytosis into the Mononuclear Phagocytotic System (MPS); 3) LNP internalization via endocytosis and LDL receptor-mediated endocytosis in the liver; 4) Cellular internalization and transgene product release; 5) mRNA and sgRNA disposition via exocytosis and clathrin-mediated endocytosis; 6) Renal elimination of LNP and sgRNA; 7) Exonuclease degradation of sgRNA and mRNA; 8) mRNA translation into Cas9 and RNP complex formation for gene editing. Monte-Carlo simulations were performed for 1000 subjects and showed a reduction in serum TTR. Results: The rate of internalization in interstitial layer was 0.039 1/h in NHP and 0.007 1/h in humans. The rate of exocytosis was 6.84 1/h in mouse, 2690 1/h in NHP, and 775 1/h in humans. Pharmacodynamics were modeled using an indirect response model, estimating first-order degradation rate (0.493 1/d) and TTR reduction parameters in NHP. Discussion: The QSP model effectively characterized biodistribution and dose-exposure relationships, aiding the development of these novel therapies. The utility of platform QSP model can be paramount in facilitating the discovery and development of these novel agents.
Collapse
Affiliation(s)
- Devam A. Desai
- Center of Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, FL, United States
| | | | - Rodrigo Cristofoletti
- Center of Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, FL, United States
| |
Collapse
|
41
|
Parrot M, Cave J, Pelaez MJ, Ghandehari H, Dogra P, Yellepeddi V. A Minimal PBPK Model Describes the Differential Disposition of Silica Nanoparticles In Vivo. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.18.24313941. [PMID: 39371117 PMCID: PMC11451661 DOI: 10.1101/2024.09.18.24313941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Nanoparticles (NPs) have emerged as promising candidates for drug delivery due to their tunable physical and chemical properties. Among these, silica nanoparticles (SiNPs) are particularly valued for their biocompatibility and adaptability in applications like drug delivery and medical imaging. However, predicting SiNP biodistribution and clearance remains a significant challenge. To address this, we developed a minimal physiologically-based pharmacokinetic (mPBPK) model to simulate the systemic disposition of SiNPs, calibrated using in vivo PK data from mice. The model assesses how variations in surface charge, size, porosity, and geometry influence SiNP biodistribution across key organs, including the kidneys, lungs, liver, and spleen. A global sensitivity analysis identified the most influential parameters, with the unbound fraction and elimination rate constants for the kidneys and MPS emerging as critical determinants of SiNP clearance. Non-compartmental analysis (NCA) further revealed that aminated SiNPs exhibit high accumulation in the liver, spleen, and kidneys, while mesoporous SiNPs primarily accumulate in the lungs. Rod-shaped SiNPs showed faster clearance compared to spherical NPs. The mPBPK model was extrapolated to predict SiNP behavior in humans, yielding strong predictive accuracy with Pearson correlation coefficients of 0.98 for mice and 0.92 for humans. This model provides a robust framework for predicting the pharmacokinetics of diverse SiNPs, offering valuable insights for optimizing NP-based drug delivery systems and guiding the translation of these therapies from preclinical models to human applications.
Collapse
|
42
|
Su S, Shen X, Shi X, Li X, Chen J, Yang W, Sun M, Tang YD, Wang H, Wang S, Cai X, Lu Y, An T, Yang Y, Meng F. Cell-penetrating peptides TAT and 8R functionalize P22 virus-like particles to enhance tissue distribution and retention in vivo. Front Vet Sci 2024; 11:1460973. [PMID: 39290505 PMCID: PMC11405305 DOI: 10.3389/fvets.2024.1460973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Virus-like particles (VLPs) are used as nanocontainers for targeted drug, protein, and vaccine delivery. The phage P22 VLP is an ideal macromolecule delivery vehicle, as it has a large exterior surface area, which facilitates multivalent genetic and chemical modifications for cell recognition and penetration. Arginine-rich cell-penetrating peptides (CPPs) can increase cargo transport efficiency in vivo. However, studies on the tissue distribution and retention of P22 VLPs mediated by TAT and 8R are lacking. This study aimed to analyze the TAT and 8R effects on the P22 VLPs transport efficiency and tissue distribution both in vitro and in vivo. We used a prokaryotic system to prepare P22 VLP self-assembled particles and expressed TAT-or 8R-conjugated mCherry on the VLP capsid protein as model cargoes and revealed that the level of P22 VLP-mCherry penetrating the cell membrane was low. However, both TAT and 8R significantly promoted the cellular uptake efficiency of P22 VLPs in vitro, as well as enhanced the tissue accumulation and retention of P22 VLPs in vivo. At 24 h postinjection, TAT enhanced the tissue distribution and retention in the lung, whereas 8R could be better accumulation in brain. Thus, TAT was superior in terms of cellular uptake and tissue accumulation in the P22 VLPs delivery system. Understanding CPP biocompatibility and tissue retention will expand their potential applications in macromolecular cargo delivery.
Collapse
Affiliation(s)
- Shibo Su
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuegang Shen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xinqi Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jin Chen
- Institute of Veterinary Immunology and Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Wei Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mingxia Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yan-Dong Tang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Haiwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shujie Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuehui Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Research Center for Veterinary Biopharmaceutical Technology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yu Lu
- Institute of Veterinary Immunology and Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Tongqing An
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yongbo Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Research Center for Veterinary Biopharmaceutical Technology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Fandan Meng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
43
|
Kirman CR, Kent B, Bigelow J, Canady RA, Chen Q, Chou WC, Li D, Lin Z, Kumar V, Paini A, Poulin P, Sweeney LM, Hays SM. Physiologically based pharmacokinetic modeling of metal nanoparticles for risk assessment of inhalation exposures: a state-of-the-science expert panel review. Nanotoxicology 2024; 18:527-541. [PMID: 39311096 DOI: 10.1080/17435390.2024.2401430] [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: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/29/2024] [Indexed: 09/29/2024]
Abstract
A critical review of the current state-of-the-science for the physiologically based pharmacokinetic (PBPK) modeling of metal nanoparticles and their application to human health risk assessment for inhalation exposures was conducted. A systematic literature search was used to identify four model groups (defined as a primary publication along with multiple supplementary publications) subject to review. Using a recent guideline document from the Organization for Economic Cooperation and Development (OECD) for PBPK model evaluation, these model groups were critically peer-reviewed by an independent panel of experts to identify those to be considered for modeling and simulation application. Based upon the expert panel input, model confidence scores for the four model groups ranged from 30 to 41 (out of a maximum score of 50). The three highest-scoring model groups were then applied to compare predictions to a different metal nanoparticle (i.e. not specifically used to parameterize the original models) using a recently published data set for tissue burdens in rats, as well as predicting human tissue burdens expected for corresponding occupational exposures. Overall, the rat models performed reasonably well in predicting the lung but tended to overestimate systemic tissue burdens. Data needs for improving the state-of-the-science, including quantitative particle characterization in tissues, nanoparticle-corona data, long-term exposure data, interspecies extrapolation methods, and human biomonitoring/toxicokinetic data are discussed.
Collapse
Affiliation(s)
| | - B Kent
- Waterborne, Leesburg, Virginia, USA
| | | | - R A Canady
- NeutralScience, Camano Island, Washington, USA
| | - Q Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA
| | - W C Chou
- Department of Environmental Sciences, College of Natural & Agricultural Sciences, University of California, Riverside, California, USA
| | - D Li
- School of Public Health, University of Nevada, Reno, Nevada, USA
| | - Z Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA
| | - V Kumar
- Environmental Engineering Laboratory, Department d'Enginyeria Quimica, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
- IISPV, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, Reus, Spain
| | - A Paini
- esqLABS GmbH, Saterland, Germany
| | - P Poulin
- Patrick Poulin Inc, Québec City, Canada
- School of Public Health, University of Montréal, Montréal, Canada
| | | | - S M Hays
- SciPinion, Bozeman, Montana, USA
| |
Collapse
|
44
|
Longobardi G, Moore TL, Conte C, Ungaro F, Satchi‐Fainaro R, Quaglia F. Polyester nanoparticles delivering chemotherapeutics: Learning from the past and looking to the future to enhance their clinical impact in tumor therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1990. [PMID: 39217459 PMCID: PMC11670051 DOI: 10.1002/wnan.1990] [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: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Polymeric nanoparticles (NPs), specifically those comprised of biodegradable and biocompatible polyesters, have been heralded as a game-changing drug delivery platform. In fact, poly(α-hydroxy acids) such as polylactide (PLA), poly(lactide-co-glycolide) (PLGA), and poly(ε-caprolactone) (PCL) have been heavily researched in the past three decades as the material basis of polymeric NPs for drug delivery applications. As materials, these polymers have found success in resorbable sutures, biodegradable implants, and even monolithic, biodegradable platforms for sustained release of therapeutics (e.g., proteins and small molecules) and diagnostics. Few fields have gained more attention in drug delivery through polymeric NPs than cancer therapy. However, the clinical translational of polymeric nanomedicines for treating solid tumors has not been congruent with the fervor or funding in this particular field of research. Here, we attempt to provide a comprehensive snapshot of polyester NPs in the context of chemotherapeutic delivery. This includes a preliminary exploration of the polymeric nanomedicine in the cancer research space. We examine the various processes for producing polyester NPs, including methods for surface-functionalization, and related challenges. After a detailed overview of the multiple factors involved with the delivery of NPs to solid tumors, the crosstalk between particle design and interactions with biological systems is discussed. Finally, we report state-of-the-art approaches toward effective delivery of NPs to tumors, aiming at identifying new research areas and re-evaluating the reasons why some research avenues have underdelivered. We hope our effort will contribute to a better understanding of the gap to fill and delineate the future research work needed to bring polyester-based NPs closer to clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Thomas Lee Moore
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Claudia Conte
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Francesca Ungaro
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- Sagol School of NeurosciencesTel Aviv UniversityTel AvivIsrael
| | - Fabiana Quaglia
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
45
|
Choi W, Kohane DS. Hybrid Nanoparticle-Hydrogel Systems for Drug Delivery Depots and Other Biomedical Applications. ACS NANO 2024; 18:22780-22792. [PMID: 39140388 PMCID: PMC11926886 DOI: 10.1021/acsnano.4c06888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Hydrogel-based depots typically tend to remain where injected and have excellent biocompatibility but are relatively poor at controlling drug release. Nanoparticles (NPs) typically have the opposite properties. The smaller the NPs are, the more likely they are to leave the site of injection. Their biocompatibility is variable depending on the material but can be poor. However, NPs can be good at controlling drug release. In these and other properties, combining NPs and hydrogels can leverage their advantages and negate their disadvantages. This review highlights the rationale for hybrid NP-hydrogel systems in drug delivery, the basic methods of producing them, and examples where combining the two systems addressed specific problems.
Collapse
Affiliation(s)
- Wonmin Choi
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
46
|
Palomino-Cano C, Moreno E, Irache JM, Espuelas S. Targeting and activation of macrophages in leishmaniasis. A focus on iron oxide nanoparticles. Front Immunol 2024; 15:1437430. [PMID: 39211053 PMCID: PMC11357945 DOI: 10.3389/fimmu.2024.1437430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages play a pivotal role as host cells for Leishmania parasites, displaying a notable functional adaptability ranging from the proinflammatory, leishmanicidal M1 phenotype to the anti-inflammatory, parasite-permissive M2 phenotype. While macrophages can potentially eradicate amastigotes through appropriate activation, Leishmania employs diverse strategies to thwart this activation and redirect macrophages toward an M2 phenotype, facilitating its survival and replication. Additionally, a competition for iron between the two entities exits, as iron is vital for both and is also implicated in macrophage defensive oxidative mechanisms and modulation of their phenotype. This review explores the intricate interplay between macrophages, Leishmania, and iron. We focus the attention on the potential of iron oxide nanoparticles (IONPs) as a sort of immunotherapy to treat some leishmaniasis forms by reprogramming Leishmania-permissive M2 macrophages into antimicrobial M1 macrophages. Through the specific targeting of iron in macrophages, the use of IONPs emerges as a promising strategy to finely tune the parasite-host interaction, endowing macrophages with an augmented antimicrobial arsenal capable of efficiently eliminating these intrusive microbes.
Collapse
Affiliation(s)
- Carmen Palomino-Cano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Esther Moreno
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Juan M. Irache
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| | - Socorro Espuelas
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| |
Collapse
|
47
|
Littrell CA, Takacs GP, Sankara CS, Sherman A, Rubach KA, Garcia JS, Bell CA, Lnu T, Harrison JK, Zhang F. Systemically targeting monocytic myloid-derrived suppressor cells using dendrimers and their cell-level biodistribution kinetics. J Control Release 2024; 374:181-193. [PMID: 39103055 DOI: 10.1016/j.jconrel.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
The focus of nanoparticles in vivo trafficking has been mostly on their tissue-level biodistribution and clearance. Recent progress in the nanomedicine field suggests that the targeting of nanoparticles to immune cells can be used to modulate the immune response and enhance therapeutic delivery to the diseased tissue. In the presence of tumor lesions, monocytic-myeloid-derived suppressor cells (M-MDSCs) expand significantly in the bone marrow, egress into peripheral blood, and traffic to the solid tumor, where they help maintain an immuno-suppressive tumor microenvironment. In this study, we investigated the interaction between PAMAM dendrimers and M-MDSCs in two murine models of glioblastoma, by examining the cell-level biodistribution kinetics of the systemically injected dendrimers. We found that M-MDSCs in the tumor and lymphoid organs can efficiently endocytose hydroxyl dendrimers. Interestingly, the trafficking of M-MDSCs from the bone marrow to the tumor contributed to the deposition of hydroxyl dendrimers in the tumor. M-MDSCs showed different capacities of endocytosing dendrimers of different functionalities in vivo. This differential uptake was mediated by the unique serum proteins associated with each dendrimer surface functionality. The results of this study set up the framework for developing dendrimer-based immunotherapy to target M-MDSCs for cancer treatment.
Collapse
Affiliation(s)
- Chad A Littrell
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Gregory P Takacs
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Chenikkayala Siva Sankara
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Alexandra Sherman
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Kai A Rubach
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Julia S Garcia
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Coral A Bell
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Tejashwini Lnu
- Department of Chemical Engineering, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Jeffrey K Harrison
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Fan Zhang
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States; Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States; Department of Chemical Engineering, University of Florida College of Pharmacy, Gainesville, FL, United States.
| |
Collapse
|
48
|
Rojekar S, Gholap AD, Togre N, Bhoj P, Haeck C, Hatvate N, Singh N, Vitore J, Dhoble S, Kashid S, Patravale V. Current status of mannose receptor-targeted drug delivery for improved anti-HIV therapy. J Control Release 2024; 372:494-521. [PMID: 38849091 DOI: 10.1016/j.jconrel.2024.06.002] [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: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
In the pursuit of achieving better therapeutic outcomes in the treatment of HIV, innovative drug delivery strategies have been extensively explored. Mannose receptors, which are primarily found on macrophages and dendritic cells, offer promising targets for drug delivery due to their involvement in HIV pathogenesis. This review article comprehensively evaluates recent drug delivery system advancements targeting the mannose receptor. We have systematically described recent developments in creating and utilizing drug delivery platforms, including nanoparticles, liposomes, micelles, noisomes, dendrimers, and other nanocarrier systems targeted at the mannose receptor. These strategies aim to enhance drug delivery specificity, bioavailability, and therapeutic efficacy while decreasing off-target effects and systemic toxicity. Furthermore, the article delves into how mannose receptors and HIV interact, highlighting the potential for exploiting this interaction to enhance drug delivery to infected cells. The review covers essential topics, such as the rational design of nanocarriers for mannose receptor recognition, the impact of physicochemical properties on drug delivery performance, and how targeted delivery affects the pharmacokinetics and pharmacodynamics of anti-HIV agents. The challenges of these novel strategies, including immunogenicity, stability, and scalability, and future research directions in this rapidly growing area are discussed. The knowledge synthesis presented in this review underscores the potential of mannose receptor-based targeted drug delivery as a promising avenue for advancing HIV treatment. By leveraging the unique properties of mannose receptors, researchers can design drug delivery systems that cater to individual needs, overcome existing limitations, and create more effective and patient-friendly treatments in the ongoing fight against HIV/AIDS.
Collapse
Affiliation(s)
- Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Namdev Togre
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Priyanka Bhoj
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Clement Haeck
- Population Council, , Center for Biomedical Research, 1230 York Avenue, New York, NY 10065, USA
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India
| | - Jyotsna Vitore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Sagar Dhoble
- Department of Pharmacology and Toxicology, R. K. Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Snehal Kashid
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
49
|
Zong Q, He C, Long B, Huang Q, Chen Y, Li Y, Dang Y, Cai C. Targeted Delivery of Nanoparticles to Blood Vessels for the Treatment of Atherosclerosis. Biomedicines 2024; 12:1504. [PMID: 39062077 PMCID: PMC11275173 DOI: 10.3390/biomedicines12071504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis is a common form of cardiovascular disease, which is one of the most prevalent causes of death worldwide, particularly among older individuals. Surgery is the mainstay of treatment for severe stenotic lesions, though the rate of restenosis remains relatively high. Current medication therapy for atherosclerosis has limited efficacy in reversing the formation of atherosclerotic plaques. The search for new drug treatment options is imminent. Some potent medications have shown surprising therapeutic benefits in inhibiting inflammation and endothelial proliferation in plaques. Unfortunately, their use is restricted due to notable dose-dependent systemic side effects or degradation. Nevertheless, with advances in nanotechnology, an increasing number of nano-related medical applications are emerging, such as nano-drug delivery, nano-imaging, nanorobots, and so forth, which allow for restrictions on the use of novel atherosclerotic drugs to be lifted. This paper reviews new perspectives on the targeted delivery of nanoparticles to blood vessels for the treatment of atherosclerosis in both systemic and local drug delivery. In systemic drug delivery, nanoparticles inhibit drug degradation and reduce systemic toxicity through passive and active pathways. To further enhance the precise release of drugs, the localized delivery of nanoparticles can also be accomplished through blood vessel wall injection or using endovascular interventional devices coated with nanoparticles. Overall, nanotechnology holds boundless potential for the diagnosis and treatment of atherosclerotic diseases in the future.
Collapse
Affiliation(s)
- Qiushuo Zong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chengyi He
- Department of Vascular Surgery, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Binbin Long
- Department of General Surgery, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan 442099, China;
| | - Qingyun Huang
- Department of Cardiothoracic Surgery, The First Hospital of Putian Affiliated to Fujian Medical University, Putian 351106, China;
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| |
Collapse
|
50
|
Zhou Y, Zhou XX, Jiang H, Liu W, Chen F, Gardea-Torresdey JL, Yan B. In Vitro Toxicity and Modeling Reveal Nanoplastic Effects on Marine Bivalves. ACS NANO 2024; 18:17228-17239. [PMID: 38877988 DOI: 10.1021/acsnano.4c04607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Nanoplastics (NPs) represent a growing concern for global environmental health, particularly in marine ecosystems where they predominantly accumulate. The impact of NPs on marine benthic organisms, such as bivalves, raises critical questions regarding ecological integrity and food safety. Traditional methods for assessing NP toxicity are often limited by their time-intensive nature and ethical considerations. Herein, we explore the toxicological effects of NPs on the marine bivalve Ruditapes philippinarum, employing a combination of in vitro cellular assays and advanced modeling techniques. Results indicate a range of adverse effects at the organismal level, including growth inhibition (69.5-108%), oxidative stress, lipid peroxidation, and DNA damage in bivalves, following exposure to NPs at concentrations in the range of 1.6 × 109-1.6 × 1011 particles/mL (p/mL). Interestingly, the growth inhibition predicted by models (54.7-104%), based on in vitro cellular proliferation assays, shows strong agreement with the in vivo outcomes of NP exposure. Furthermore, we establish a clear correlation between cytotoxicity observed in vitro and the toxicological responses at the organismal level. Taken together, this work suggests that the integration of computational modeling with in vitro toxicity assays can predict the detrimental effects of NPs on bivalves, offering insightful references for assessing the environmental risk assessment of NPs in marine benthic ecosystems.
Collapse
Affiliation(s)
- Yanfei Zhou
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Xiao-Xia Zhou
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Hao Jiang
- CAS Key Laboratory of Aquatic Botany and Watershed Ecology, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Wenzhi Liu
- CAS Key Laboratory of Aquatic Botany and Watershed Ecology, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Fengyuan Chen
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Jorge L Gardea-Torresdey
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Bing Yan
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|