1
|
Jadhav K, Jhilta A, Singh R, Negi S, Singhal N, Mishra A, Singh AK, Verma RK. Targeted Nasal Route Delivery of Cationic Anti-TB Drug-Loaded Nano-embedded Microparticles for Mycobacterial Elimination in the CNS. Mol Pharm 2025. [PMID: 40340385 DOI: 10.1021/acs.molpharmaceut.5c00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Central nervous system tuberculosis (CNS-TB) is a severe and insidious form of extrapulmonary tuberculosis (TB) associated with a high mortality rate, often leading to fatal outcomes or debilitating neurological impairments. The therapeutic regimen for CNS-TB follows an approach similar to that of pulmonary TB but faces significant challenges in effectively reaching the cerebrospinal fluid and achieving therapeutic drug levels in the brain and surrounding fluids. A major obstacle in CNS-TB treatment is the difficulty in permeating the blood-brain barrier (BBB). The nasal route of drug delivery offers a promising approach for targeting anti-TB drugs directly to the infection sites, enabling higher drug concentrations while bypassing the BBB. The present study focused on the development of cationic poly(lactic-co-glycolic) acid (PLGA) nanoparticles (CS-PLGA NPs) loaded with anti-TB drugs (ATDs), namely, isoniazid (INH) and rifampicin (RIF). These CS-PLGA NPs were then processed into dynamic microsized nanoembedded microparticles (NEMs) using spray drying. The ATD-NEMs formulation demonstrated significantly enhanced permeation across RPMI 2650 nasal septum monolayers compared with free ATDs. Intranasal delivery of the NEM formulation to TB-infected mice over a four-week period resulted in a substantial reduction in colony-forming units (CFUs) (1.53 ± 0.50 log10 CFU/gram) compared to the untreated group (4.45 ± 0.67 log10 CFU/gram). Furthermore, the NEM formulation showed improved recovery in histopathological analysis, consistent with CFU reduction. Preclinical data support the feasibility of intranasally administering the NEMs formulation, demonstrating high therapeutic efficacy and the potential to address brain inflammation in the murine CNS-TB model.
Collapse
Affiliation(s)
- Krishna Jadhav
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India
| | - Agrim Jhilta
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India
| | - Raghuraj Singh
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India
| | - Swarnima Negi
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India
| | - Nitin Singhal
- National Agri-Food and Bio-manufacturing Institute (NABI), Sector 81, Sahibzada Ajit Singh Nagar 140308, Punjab, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur 342011, Rajasthan, India
| | - Amit Kumar Singh
- Experimental Animal Facility, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra 282004, India
| | - Rahul Kumar Verma
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India
| |
Collapse
|
2
|
Bazargani A, Hejazi M, Fernandez M, Cordeiro A, Tsala Ebode J, Lewinski N, da Rocha S, Golshahi L. PEGylated solid lipid nanoparticles for the intranasal delivery of combination antiretroviral therapy composed of Atazanavir and Elvitegravir to treat NeuroAIDS. Int J Pharm 2025; 670:125166. [PMID: 39761706 DOI: 10.1016/j.ijpharm.2025.125166] [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: 10/14/2024] [Revised: 12/19/2024] [Accepted: 01/01/2025] [Indexed: 01/14/2025]
Abstract
Intranasal drug administration offers a promising strategy for delivering combination antiretroviral therapy (cART) directly to the central nervous system to treat NeuroAIDS, leveraging the nose-to-brain route to bypass the blood-brain barrier. However, challenges such as enzymatic degradation in the nasal mucosa, low permeability, and mucociliary clearance within the nasal cavity must first be addressed to make this route feasible. To overcome these barriers, this study developed solid lipid nanoparticles (SLNs) with varying PEGylation levels (0 %, 5 %, 10 %, and 15 % w/w of PEGylated lipid), co-encapsulated with Elvitegravir (EVG) and Atazanavir (ATZ) as an integrase and protease inhibitor, respectively. Pre-formulation studies confirmed the compatibility of the drugs with the excipients. Characterization showed that PEGylation reduces SLN size by approximately up to 12 % while maintaining monodispersity and a high encapsulation efficiency of over 99 % for both EVG and ATZ in their amorphous forms. Incubation of the formulations in artificial nasal mucus revealed that increased PEGylation consistently reduces nanoparticle aggregation and mean aggregate size, suggesting improved SLN stability in the mucus. Importantly, higher PEGylation levels significantly enhanced model drug permeability across the nasal mucus barrier by up to 10-fold. Lastly, cellular uptake studies using the RPMI 2650 nasal epithelial cell line indicated that PEGylation does not reduce nanoparticle uptake rates. These findings highlight the potential of PEGylated SLNs as an effective vehicle for enhancing the intranasal delivery of cART to treat NeuroAIDS. However, further in vivo studies are needed to confirm the brain targeting potential of this formulation.
Collapse
Affiliation(s)
- Arya Bazargani
- College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA; School of Pharmacy, Virginia Commonwealth University, 410 N 12th St, Richmond, VA 23298, USA.
| | - Mohammad Hejazi
- College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA.
| | - Matthew Fernandez
- School of Pharmacy, Virginia Commonwealth University, 410 N 12th St, Richmond, VA 23298, USA.
| | - Arthur Cordeiro
- School of Pharmacy, Virginia Commonwealth University, 410 N 12th St, Richmond, VA 23298, USA.
| | - Johanna Tsala Ebode
- College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA.
| | - Nastassja Lewinski
- College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA.
| | - Sandro da Rocha
- School of Pharmacy, Virginia Commonwealth University, 410 N 12th St, Richmond, VA 23298, USA.
| | - Laleh Golshahi
- College of Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23284, USA.
| |
Collapse
|
3
|
Jadhav K, Jhilta A, Singh R, Sharma S, Negi S, Ahirwar K, Shukla R, Singh AK, Verma RK. Trans-nasal brain delivery of anti-TB drugs by methyl-β-cyclodextrin microparticles show efficient mycobacterial clearance from central nervous system. J Control Release 2025; 378:671-686. [PMID: 39689814 DOI: 10.1016/j.jconrel.2024.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
Central nervous system tuberculosis (CNS-TB) is the most severe extra-pulmonary manifestation of tuberculosis (TB), facing significant challenges due to the limited penetration of anti-TB drugs (ATDs) across the blood-brain barrier (BBB) and their insufficient concentrations at the site of infection. This study aimed to enhance the efficacy of ATDs by encapsulating them in methyl-β-cyclodextrin (M-β-CD) microparticles (ATD-MP) using spray drying, intended for intranasal delivery to manage CNS-TB. M-β-CD microparticles loaded with isoniazid (INH) and rifampicin (RIF) exhibited spherical shapes with slightly deflated surfaces and particle sizes of 6.24 ± 0.77 μm and 5.97 ± 0.50 μm, respectively. M-β-CD improved the permeation of ATDs through RPMI-2650 cell monolayers while reducing drug cytotoxicity. Pharmacokinetic and biodistribution analysis demonstrated that intranasal administration of ATD-MP significantly enhanced the trans-nasal brain delivery of ATDs and their distribution in the brain, achieving the minimum inhibitory concentration. In a murine model of CNS-TB, intranasal insufflation of ATD-MP for four weeks led to a significant reduction (∼0.78 Log10 CFU) in mycobacterial burden in the brain compared to the untreated group (∼3.60 Log10 CFU). These preclinical results underscore the potential of intranasal administration of M-β-CD microparticles as an effective therapeutic strategy for combating brain inflammation in CNS-TB.
Collapse
Affiliation(s)
- Krishna Jadhav
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology (INST), Sector-81, Mohali, Punjab 140306, India
| | - Agrim Jhilta
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology (INST), Sector-81, Mohali, Punjab 140306, India
| | - Raghuraj Singh
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology (INST), Sector-81, Mohali, Punjab 140306, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Shweta Sharma
- Experimental Animal Facility, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra 282004, India
| | - Swarnima Negi
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology (INST), Sector-81, Mohali, Punjab 140306, India
| | - Kailash Ahirwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Lucknow, UP 226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Lucknow, UP 226002, India
| | - Amit Kumar Singh
- Experimental Animal Facility, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra 282004, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| | - Rahul Kumar Verma
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology (INST), Sector-81, Mohali, Punjab 140306, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
4
|
Hua T, Li S, Han B. Nanomedicines for intranasal delivery: understanding the nano-bio interactions at the nasal mucus-mucosal barrier. Expert Opin Drug Deliv 2024; 21:553-572. [PMID: 38720439 DOI: 10.1080/17425247.2024.2339335] [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: 01/04/2024] [Accepted: 04/02/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Intranasal administration is an effective drug delivery routes in modern pharmaceutics. However, unlike other in vivo biological barriers, the nasal mucosal barrier is characterized by high turnover and selective permeability, hindering the diffusion of both particulate drug delivery systems and drug molecules. The in vivo fate of administrated nanomedicines is often significantly affected by nano-biointeractions. AREAS COVERED The biological barriers that nanomedicines encounter when administered intranasally are introduced, with a discussion on the factors influencing the interaction between nanomedicines and the mucus layer/mucosal barriers. General design strategies for nanomedicines administered via the nasal route are further proposed. Furthermore, the most common methods to investigate the characteristics and the interactions of nanomedicines when in presence of the mucus layer/mucosal barrier are briefly summarized. EXPERT OPINION Detailed investigation of nanomedicine-mucus/mucosal interactions and exploration of their mechanisms provide solutions for designing better intranasal nanomedicines. Designing and applying nanomedicines with mucus interaction properties or non-mucosal interactions should be customized according to the therapeutic need, considering the target of the drug, i.e. brain, lung or nose. Then how to improve the precise targeting efficiency of nanomedicines becomes a difficult task for further research.
Collapse
Affiliation(s)
- Tangsiyuan Hua
- School of Pharmacy, Changzhou Univesity, Changzhou, PR China
| | - Shuling Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, PR China
| | - Bing Han
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| |
Collapse
|
5
|
Maaz A, Blagbrough IS, De Bank PA. A Cell-Based Nasal Model for Screening the Deposition, Biocompatibility, and Transport of Aerosolized PLGA Nanoparticles. Mol Pharm 2024; 21:1108-1124. [PMID: 38333983 PMCID: PMC10915796 DOI: 10.1021/acs.molpharmaceut.3c00639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/07/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024]
Abstract
The olfactory region of the nasal cavity directly links the brain to the external environment, presenting a potential direct route to the central nervous system (CNS). However, targeting drugs to the olfactory region is challenging and relies on a combination of drug formulation, delivery device, and administration technique to navigate human nasal anatomy. In addition, in vitro and in vivo models utilized to evaluate the performance of nasal formulations do not accurately reflect deposition and uptake in the human nasal cavity. The current study describes the development of a respirable poly(lactic-co-glycolic acid) nanoparticle (PLGA NP) formulation, delivered via a pressurized metered dose inhaler (pMDI), and a cell-containing three-dimensional (3D) human nasal cast model for deposition assessment of nasal formulations in the olfactory region. Fluorescent PLGA NPs (193 ± 3 nm by dynamic light scattering) were successfully formulated in an HFA134a-based pMDI and were collected intact following aerosolization. RPMI 2650 cells, widely employed as a nasal epithelial model, were grown at the air-liquid interface (ALI) for 14 days to develop a suitable barrier function prior to exposure to the aerosolized PLGA NPs in a glass deposition apparatus. Direct aerosol exposure was shown to have little effect on cell viability. Compared to an aqueous NP suspension, the transport rate of the aerosolized NPs across the RPMI 2650 barrier was higher at all time points indicating the potential advantages of delivery via aerosolization and the importance of employing ALI cellular models for testing respirable formulations. The PLGA NPs were then aerosolized into a 3D-printed human nasal cavity model with an insert of ALI RPMI 2650 cells positioned in the olfactory region. Cells remained highly viable, and there was significant deposition of the fluorescent NPs on the ALI cultures. This study is a proof of concept that pMDI delivery of NPs is a viable means of targeting the olfactory region for nose-to-brain drug delivery (NTBDD). The cell-based model allows not only maintenance under ALI culture conditions but also sampling from the basal chamber compartment; hence, this model could be adapted to assess drug deposition, uptake, and transport kinetics in parallel under real-life settings.
Collapse
Affiliation(s)
- Aida Maaz
- Department
of Life Sciences, Centre for Therapeutic Innovation, and Centre for Bioengineering
& Biomedical Technologies, University
of Bath, Bath BA2 7AY, U.K.
| | - Ian S. Blagbrough
- Department
of Life Sciences, Centre for Therapeutic Innovation, and Centre for Bioengineering
& Biomedical Technologies, University
of Bath, Bath BA2 7AY, U.K.
| | - Paul A. De Bank
- Department
of Life Sciences, Centre for Therapeutic Innovation, and Centre for Bioengineering
& Biomedical Technologies, University
of Bath, Bath BA2 7AY, U.K.
| |
Collapse
|
6
|
Peng G, Cai J, Wang Z, Zhang W, Xu J, Zhang D, Gong D. Facile fabrication of diatomite biosilica-based nasal drug delivery vehicle for enhanced treatment of allergic rhinitis. Colloids Surf B Biointerfaces 2024; 234:113715. [PMID: 38134821 DOI: 10.1016/j.colsurfb.2023.113715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023]
Abstract
Nanostructured silica-based materials have great potential as drug delivery vehicles for precise and personalized medical applications. As natural nanostructured silica, diatomite biosilica (DB) is recognized as a novel carrier to construct oral/parenteral smart drug delivery systems due to high surface area, biocompatibility, and applicability at low cost, yet the related studies on its use in local delivery routes are still scarce. Herein, we proposed a novel strategy to develop multifunctional nasal drug delivery vehicles based on DB, and demonstrated their versatile performance for enhanced treatment of allergic rhinitis (AR). As a proof of concept, the purified DB microparticles were loaded with budesonide as an anti-inflammatory model drug, and further processed via surface modification to graft polydopamine and carboxymethyl chitosan layers. The synthesized microcapsules exhibited remarkable mucin binding capacity and antibacterial activity against Staphylococcus aureus. Besides, toxicity evaluation with human skin fibroblast cells and hemolysis tests indicated their high biocompatibility. Moreover, in vitro drug release results demonstrated pH-responsive release performance of the microcapsules under simulated AR environment (pH 5.0, 35 °C). Hence, this study provides a facile and reliable approach to construct DB-based mucoadhesive nasal drug delivery vehicles, showing great potential for treatment of allergic airway inflammatory diseases.
Collapse
Affiliation(s)
- Guanya Peng
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China
| | - Jun Cai
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China
| | - Zhenhu Wang
- Beijing Institute of Radio Measurement, Beijing 100854, China
| | - Wenqiang Zhang
- College of Engineering, China Agricultural University, Beijing 100083, China
| | - Junjie Xu
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China
| | - Deyuan Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China
| | - De Gong
- School of Mechanical Engineering and Automation, Beihang University, Beijing 100191, China.
| |
Collapse
|
7
|
Khalil A, Barras A, Boukherroub R, Tseng CL, Devos D, Burnouf T, Neuhaus W, Szunerits S. Enhancing paracellular and transcellular permeability using nanotechnological approaches for the treatment of brain and retinal diseases. NANOSCALE HORIZONS 2023; 9:14-43. [PMID: 37853828 DOI: 10.1039/d3nh00306j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Paracellular permeability across epithelial and endothelial cells is, in large part, regulated by apical intercellular junctions also referred to as tight junctions (TJs). These junctions contribute to the spatial definition of different tissue compartments within organisms, separating them from the outside world as well as from inner compartments, with their primary physiological role of maintaining tissue homeostasis. TJs restrict the free, passive diffusion of ions and hydrophilic small molecules through paracellular clefts and are important for appropriate cell polarization and transporter protein localisation, supporting the controlled transcellular diffusion of smaller and larger hydrophilic as well as hydrophobic substances. This traditional diffusion barrier concept of TJs has been challenged lately, owing to a better understanding of the components that are associated with TJs. It is now well-established that mutations in TJ proteins are associated with a range of human diseases and that a change in the membrane fluidity of neighbouring cells can open possibilities for therapeutics to cross intercellular junctions. Nanotechnological approaches, exploiting ultrasound or hyperosmotic agents and permeation enhancers, are the paradigm for achieving enhanced paracellular diffusion. The other widely used transport route of drugs is via transcellular transport, allowing the passage of a variety of pro-drugs and nanoparticle-encapsulated drugs via different mechanisms based on receptors and others. For a long time, there was an expectation that lipidic nanocarriers and polymeric nanostructures could revolutionize the field for the delivery of RNA and protein-based therapeutics across different biological barriers equipped with TJs (e.g., blood-brain barrier (BBB), retina-blood barrier (RBB), corneal TJs, etc.). However, only a limited increase in therapeutic efficiency has been reported for most systems until now. The purpose of this review is to explore the reasons behind the current failures and to examine the emergence of synthetic and cell-derived nanomaterials and nanotechnological approaches as potential game-changers in enhancing drug delivery to target locations both at and across TJs using innovative concepts. Specifically, we will focus on recent advancements in various nanotechnological strategies enabling the bypassing or temporally opening of TJs to the brain and to the retina, and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Asmaa Khalil
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Alexandre Barras
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Ching-Li Tseng
- Taipei Medical University, Graduate Institute of Biomedical Materials and Tissue Engineering (GIBMTE), New Taipei City 235603, Taiwan
- Taipei Medical University, International PhD Program in Biomedical Engineering (IPBME), New Taipei City 235603, Taiwan
| | - David Devos
- University Lille, CHU-Lille, Inserm, U1172, Lille Neuroscience & Cognition, LICEND, Lille, France
| | - Thierry Burnouf
- Taipei Medical University, Graduate Institute of Biomedical Materials and Tissue Engineering (GIBMTE), New Taipei City 235603, Taiwan
- Taipei Medical University, International PhD Program in Biomedical Engineering (IPBME), New Taipei City 235603, Taiwan
| | - Winfried Neuhaus
- AIT - Austrian Institute of Technology GmbH, Center Health and Bioresources, Competence Unit Molecular Diagnostics, 1210 Vienna, Austria
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems, Austria
| | - Sabine Szunerits
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| |
Collapse
|
8
|
Wang Y, Chen L, Wang Y, Wang X, Qian D, Yan J, Sun Z, Cui P, Yu L, Wu J, He Z. Marine biomaterials in biomedical nano/micro-systems. J Nanobiotechnology 2023; 21:408. [PMID: 37926815 PMCID: PMC10626837 DOI: 10.1186/s12951-023-02112-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/15/2023] [Indexed: 11/07/2023] Open
Abstract
Marine resources in unique marine environments provide abundant, cost-effective natural biomaterials with distinct structures, compositions, and biological activities compared to terrestrial species. These marine-derived raw materials, including polysaccharides, natural protein components, fatty acids, and marine minerals, etc., have shown great potential in preparing, stabilizing, or modifying multifunctional nano-/micro-systems and are widely applied in drug delivery, theragnostic, tissue engineering, etc. This review provides a comprehensive summary of the most current marine biomaterial-based nano-/micro-systems developed over the past three years, primarily focusing on therapeutic delivery studies and highlighting their potential to cure a variety of diseases. Specifically, we first provided a detailed introduction to the physicochemical characteristics and biological activities of natural marine biocomponents in their raw state. Furthermore, the assembly processes, potential functionalities of each building block, and a thorough evaluation of the pharmacokinetics and pharmacodynamics of advanced marine biomaterial-based systems and their effects on molecular pathophysiological processes were fully elucidated. Finally, a list of unresolved issues and pivotal challenges of marine-derived biomaterials applications, such as standardized distinction of raw materials, long-term biosafety in vivo, the feasibility of scale-up, etc., was presented. This review is expected to serve as a roadmap for fundamental research and facilitate the rational design of marine biomaterials for diverse emerging applications.
Collapse
Affiliation(s)
- Yanan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Long Chen
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 55000, Guizhou, China
| | - Yuanzheng Wang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 55000, Guizhou, China.
| | - Xinyuan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Deyao Qian
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Jiahui Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Zeyu Sun
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 55000, Guizhou, China
| | - Pengfei Cui
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266100, China.
| | - Liangmin Yu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Jun Wu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, 999077, China.
| | - Zhiyu He
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China.
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China.
| |
Collapse
|
9
|
Shi S, Ren H, Xie Y, Yu M, Chen Y, Yang L. Engineering advanced nanomedicines against central nervous system diseases. MATERIALS TODAY 2023; 69:355-392. [DOI: 10.1016/j.mattod.2023.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Botti G, Bianchi A, Dalpiaz A, Tedeschi P, Albanese V, Sorrenti M, Catenacci L, Bonferoni MC, Beggiato S, Pavan B. Dimeric ferulic acid conjugate as a prodrug for brain targeting after nasal administration of loaded solid lipid microparticles. Expert Opin Drug Deliv 2023; 20:1657-1679. [PMID: 38014509 DOI: 10.1080/17425247.2023.2286369] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023]
Abstract
OBJECTIVE Ferulic acid (Fer) displays antioxidant/anti-inflammatory properties useful against neurodegenerative diseases. To increase Fer uptake and its central nervous system residence time, a dimeric prodrug, optimizing the Fer loading on nasally administrable solid lipid microparticles (SLMs), was developed. METHODS The prodrug was synthesized as Fer dimeric conjugate methylated on the carboxylic moiety. Prodrug antioxidant/anti-inflammatory properties and ability to release Fer in physiologic environments were evaluated. Tristearin or stearic acid SLMs were obtained by hot emulsion technique. In vivo pharmacokinetics were quantified by HPLC. RESULTS The prodrug was able to release Fer in physiologic environments (whole blood and brain homogenates) and induce in vitro antioxidant/anti-inflammatory effects. Its half-life in rats was 18.0 ± 1.9 min. Stearic acid SLMs, exhibiting the highest prodrug loading and dissolution rate, were selected for nasal administration to rats (1 mg/kg dose), allowing to obtain high prodrug bioavailability and prolonged residence in the cerebrospinal fluid, showing AUC (Area Under Concentration) values (108.5 ± 3.9 μg∙mL-1∙min) up to 30 times over those of Fer free drug, after its intravenous/nasal administration (3.3 ± 0.3/5.16 ± 0.20 μg∙mL-1∙min, respectively) at the same dose. Chitosan presence further improved the prodrug brain uptake. CONCLUSIONS Nasal administration of prodrug-loaded SLMs can be proposed as a noninvasive approach for neurodegenerative disease therapy.
Collapse
Affiliation(s)
- Giada Botti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Anna Bianchi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandro Dalpiaz
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Paola Tedeschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Valentina Albanese
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| | - Milena Sorrenti
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Laura Catenacci
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | | | - Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara and LTTA Center, Ferrara, Italy
| | - Barbara Pavan
- Department of Neuroscience and Rehabilitation-Section of Physiology, University of Ferrara, Ferrara, Italy
- Center for Translational Neurophysiology of Speech and Communication (CTNSC), Italian Institute of Technology (IIT), Ferrara, Italy
| |
Collapse
|
11
|
Puri V, Kaur VP, Singh A, Singh C. Recent advances on drug delivery applications of mucopenetrative/mucoadhesive particles: A review. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Surendranath M, M R R, Parameswaran R. Recent advances in functionally modified polymers for mucoadhesive drug delivery. J Mater Chem B 2022; 10:5913-5924. [PMID: 35880449 DOI: 10.1039/d2tb00856d] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Novel methods for the delivery of drugs other than the conventional method of oral administration have been a thrust area of research for a few decades. Mucoadhesive delivery of drugs opened up a new domain where rapid and patient-friendly delivery of drugs can be achieved. Delivery of drugs through the mucosal sites such as buccal, nasal, ocular, sublingual, rectal and vaginal facilitates bypassing the first-pass metabolism and the drug reaches the systemic circulation directly. This helps to increase the bioavailability of the drug. The study of the chemical characteristics of polymers with mucoadhesive properties and how the molecules or the pharmaceuticals are transported across the mucosa is very much needed for the advancement of research in this field. And at the same time, it is very pertinent to know about the anatomy and the physiology of the mucosal tissue and its variation in different regions of the body. In this review, we try to present a comprehensive understanding of relevant topics of mucoadhesion giving more emphasis on the mechanism of transport of drugs across mucosa, and different possible functional modifications of polymers to enhance the property of mucoadhesion.
Collapse
Affiliation(s)
- Medha Surendranath
- Division of Polymeric Medical Devices, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India.
| | - Rekha M R
- Division of Biosurface Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Ramesh Parameswaran
- Division of Polymeric Medical Devices, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
13
|
Trousil J, Dal NJK, Fenaroli F, Schlachet I, Kubíčková P, Janoušková O, Pavlova E, Škorič M, Trejbalová K, Pavliš O, Sosnik A. Antibiotic-Loaded Amphiphilic Chitosan Nanoparticles Target Macrophages and Kill an Intracellular Pathogen. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201853. [PMID: 35691939 DOI: 10.1002/smll.202201853] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/19/2022] [Indexed: 06/15/2023]
Abstract
In this work, levofloxacin (LVX), a third-generation fluoroquinolone antibiotic, is encapsulated within amphiphilic polymeric nanoparticles of a chitosan-g-poly(methyl methacrylate) produced by self-assembly and physically stabilized by ionotropic crosslinking with sodium tripolyphosphate. Non-crosslinked nanoparticles display a size of 29 nm and a zeta-potential of +36 mV, while the crosslinked counterparts display 45 nm and +24 mV, respectively. The cell compatibility, uptake, and intracellular trafficking are characterized in the murine alveolar macrophage cell line MH-S and the human bronchial epithelial cell line BEAS-2B in vitro. Internalization events are detected after 10 min and the uptake is inhibited by several endocytosis inhibitors, indicating the involvement of complex endocytic pathways. In addition, the nanoparticles are detected in the lysosomal compartment. Then, the antibacterial efficacy of LVX-loaded nanoformulations (50% w/w drug content) is assessed in MH-S and BEAS-2B cells infected with Staphylococcus aureus and the bacterial burden is decreased by 49% and 46%, respectively. In contrast, free LVX leads to a decrease of 8% and 5%, respectively, in the same infected cell lines. Finally, intravenous injection to a zebrafish larval model shows that the nanoparticles accumulate in macrophages and endothelium and demonstrate the promise of these amphiphilic nanoparticles to target intracellular infections.
Collapse
Affiliation(s)
- Jiří Trousil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, 162 00, Czech Republic
| | | | | | - Inbar Schlachet
- Laboratory of Pharmaceutical Nanomaterials Science, Faculty of Materials Science and Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Pavla Kubíčková
- Military Health Institute, Military Medical Agency, Prague, 160 00, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, 162 00, Czech Republic
- Department of Biology, Faculty of Science, University of J. E. Purkyně, Ústí nad Labem, 400 96, Czech Republic
| | - Ewa Pavlova
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, 162 00, Czech Republic
| | - Miša Škorič
- Department of Pathological Morphology and Parasitology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Brno, 612 42, Czech Republic
| | - Kateřina Trejbalová
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague, 142 20, Czech Republic
| | - Oto Pavliš
- Military Health Institute, Military Medical Agency, Prague, 160 00, Czech Republic
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Faculty of Materials Science and Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| |
Collapse
|
14
|
Mixing efficiency affects the morphology and compactness of chitosan/tripolyphosphate nanoparticles. Carbohydr Polym 2022; 287:119331. [DOI: 10.1016/j.carbpol.2022.119331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/22/2022] [Accepted: 03/06/2022] [Indexed: 11/17/2022]
|
15
|
Awad R, Avital A, Sosnik A. Polymeric nanocarriers for nose-to-brain drug delivery in neurodegenerative diseases and neurodevelopmental disorders. Acta Pharm Sin B 2022; 13:1866-1886. [DOI: 10.1016/j.apsb.2022.07.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 11/01/2022] Open
|
16
|
Cannabidiol-Loaded Mixed Polymeric Micelles of Chitosan/Poly(Vinyl Alcohol) and Poly(Methyl Methacrylate) for Trans-Corneal Delivery. Pharmaceutics 2021; 13:pharmaceutics13122142. [PMID: 34959427 PMCID: PMC8703866 DOI: 10.3390/pharmaceutics13122142] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
Ocular drug delivery is challenging due to the very short drug residence time and low permeability. In this work, we produce and characterize mucoadhesive mixed polymeric micelles (PMs) made of chitosan (CS) and poly(vinyl alcohol) backbones graft-hydrophobized with short poly(methyl methacrylate) blocks and use them to encapsulate cannabidiol (CBD), an anti-inflammatory cannabinoid. CBD-loaded mixed PMs are physically stabilized by ionotropic crosslinking of the CS domains with sodium tripolyphoshate and spray-drying. These mixed PMs display CBD loading capacity of 20% w/w and sizes of 100-200 nm, and spherical morphology (cryogenic-transmission electron microscopy). The good compatibility of the unloaded and CBD-loaded PMs is assessed in a human corneal epithelial cell line. Then, we confirm the permeability of CBD-free PMs and nanoencapsulated CBD in human corneal epithelial cell monolayers under liquid-liquid and air-liquid conditions. Overall, our results highlight the potential of these polymeric nanocarriers for ocular drug delivery.
Collapse
|
17
|
Targeting Systems to the Brain Obtained by Merging Prodrugs, Nanoparticles, and Nasal Administration. Pharmaceutics 2021; 13:pharmaceutics13081144. [PMID: 34452105 PMCID: PMC8399330 DOI: 10.3390/pharmaceutics13081144] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/23/2021] [Indexed: 01/27/2023] Open
Abstract
About 40 years ago the lipidization of hydrophilic drugs was proposed to induce their brain targeting by transforming them into lipophilic prodrugs. Unfortunately, lipidization often transforms a hydrophilic neuroactive agent into an active efflux transporter (AET) substrate, with consequent rejection from the brain after permeation across the blood brain barrier (BBB). Currently, the prodrug approach has greatly evolved in comparison to lipidization. This review describes the evolution of the prodrug approach for brain targeting considering the design of prodrugs as active influx substrates or molecules able to inhibit or elude AETs. Moreover, the prodrug approach appears strategic in optimization of the encapsulation of neuroactive drugs in nanoparticulate systems that can be designed to induce their receptor-mediated transport (RMT) across the BBB by appropriate decorations on their surface. Nasal administration is described as a valuable alternative to obtain the brain targeting of drugs, evidencing that the prodrug approach can allow the optimization of micro or nanoparticulate nasal formulations of neuroactive agents in order to obtain this goal. Furthermore, nasal administration is also proposed for prodrugs characterized by peripheral instability but potentially able to induce their targeting inside cells of the brain.
Collapse
|
18
|
Baldassi D, Gabold B, Merkel O. Air-liquid interface cultures of the healthy and diseased human respiratory tract: promises, challenges and future directions. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000111. [PMID: 34345878 PMCID: PMC7611446 DOI: 10.1002/anbr.202000111] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Air-liquid interface (ALI) culture models currently represent a valid instrument to recreate the typical aspects of the respiratory tract in vitro in both healthy and diseased state. They can help reducing the number of animal experiments, therefore, supporting the 3R principle. This review discusses ALI cultures and co-cultures derived from immortalized as well as primary cells, which are used to study the most common disorders of the respiratory tract, in terms of both pathophysiology and drug screening. The article displays ALI models used to simulate inflammatory lung diseases such as chronic obstructive pulmonary disease (COPD), asthma, cystic fibrosis, lung cancer, and viral infections. It also includes a focus on ALI cultures described in literature studying respiratory viruses such as SARS-CoV-2 causing the global Covid-19 pandemic at the time of writing this review. Additionally, commercially available models of ALI cultures are presented. Ultimately, the aim of this review is to provide a detailed overview of ALI models currently available and to critically discuss them in the context of the most prevalent diseases of the respiratory tract.
Collapse
Affiliation(s)
- Domizia Baldassi
- Pharmaceutical Technology and Biopharmacy, LMU Munich Butenandtstr. 5-13, 81377 Munich, Germany
| | - Bettina Gabold
- Pharmaceutical Technology and Biopharmacy, LMU Munich Butenandtstr. 5-13, 81377 Munich, Germany
| | - Olivia Merkel
- Pharmaceutical Technology and Biopharmacy, LMU Munich Butenandtstr. 5-13, 81377 Munich, Germany
| |
Collapse
|
19
|
Li S, Guo C, Zhang X, Liu X, Mu J, Liu C, Peng Y, Chang M. Self-assembling modified neuropeptide S enhances nose-to-brain penetration and exerts a prolonged anxiolytic-like effect. Biomater Sci 2021; 9:4765-4777. [PMID: 34037635 DOI: 10.1039/d1bm00380a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Anxiety disorders are the most common mental diseases and can greatly disrupt everyday life. Although there has been substantial research on anxiety disorders, novel therapeutics are needed. Neuropeptide S (NPS) is a potential therapeutic candidate owing to its strong anxiolytic activity; however, some disadvantages, such as its poor metabolic stability and inability to cross the blood-brain barrier (BBB), limit its use in the clinic. Herein, inspired by nose-to-brain drug delivery strategies, an endogenous 20-amino-acid-long mNPS peptide was modified by incorporating palmitic acid into its functional Lys12 side chain (M-3), which was expected to facilitate nose-to-brain penetration and exert a prolonged anxiolytic-like effect compared to mNPS. We found that M-3 assembled into nanofibers that retained the bioactivity of NPS and exhibited obvious improvements in metabolic stability. Notably, as expected, self-assembled M-3 was able to penetrate into the brain and exert anxiolytic effects. The elevated plus-maze (EPM) results further revealed that M-3 could produce prolonged anxiolytic-like effects in mice. In vivo imaging studies revealed that self-assembled M-3 could be efficiently transported from the nasal cavity to the brain. Furthermore, when intranasally administered, this molecule exhibited a significantly prolonged anxiolytic-like effect, which further illustrated that this molecule has a potent nose-to-brain penetration in vivo. Overall, this self-assembled nanofiber showed potent nose-to-brain penetration ability and prolonged bioactivity.
Collapse
Affiliation(s)
- Shu Li
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Chen Guo
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Xingjiao Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Xiaojing Liu
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Jing Mu
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Chunxia Liu
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Yali Peng
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| | - Min Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
20
|
Kumarasamy M, Sosnik A. Heterocellular spheroids of the neurovascular blood-brain barrier as a platform for personalized nanoneuromedicine. iScience 2021; 24:102183. [PMID: 33718835 PMCID: PMC7921813 DOI: 10.1016/j.isci.2021.102183] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/03/2021] [Accepted: 02/09/2021] [Indexed: 12/22/2022] Open
Abstract
Nanoneuromedicine investigates nanotechnology to target the brain and treat neurological diseases. In this work, we biofabricated heterocellular spheroids comprising human brain microvascular endothelial cells, brain vascular pericytes and astrocytes combined with primary cortical neurons and microglia isolated from neonate rats. The structure and function are characterized by confocal laser scanning and light sheet fluorescence microscopy, electron microscopy, western blotting, and RNA sequencing. The spheroid bulk is formed by neural cells and microglia and the surface by endothelial cells and they upregulate key structural and functional proteins of the blood-brain barrier. These cellular constructs are utilized to preliminary screen the permeability of polymeric, metallic, and ceramic nanoparticles (NPs). Findings reveal that penetration and distribution patterns depend on the NP type and that microglia would play a key role in this pathway, highlighting the promise of this platform to investigate the interaction of different nanomaterials with the central nervous system in nanomedicine, nanosafety and nanotoxicology.
Collapse
Affiliation(s)
- Murali Kumarasamy
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Bldg. Office 607, Technion City, 3200003 Haifa, Israel
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Bldg. Office 607, Technion City, 3200003 Haifa, Israel
| |
Collapse
|
21
|
das Neves J, Sverdlov Arzi R, Sosnik A. Molecular and cellular cues governing nanomaterial-mucosae interactions: from nanomedicine to nanotoxicology. Chem Soc Rev 2021; 49:5058-5100. [PMID: 32538405 DOI: 10.1039/c8cs00948a] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mucosal tissues constitute the largest interface between the body and the surrounding environment and they regulate the access of molecules, supramolecular structures, particulate matter, and pathogens into it. All mucosae are characterized by an outer mucus layer that protects the underlying cells from physicochemical, biological and mechanical insults, a mono-layered or stratified epithelium that forms tight junctions and controls the selective transport of solutes across it and associated lymphoid tissues that play a sentinel role. Mucus is a gel-like material comprised mainly of the glycoprotein mucin and water and it displays both hydrophilic and hydrophobic domains, a net negative charge, and high porosity and pore interconnectivity, providing an efficient barrier for the absorption of therapeutic agents. To prolong the residence time, absorption and bioavailability of a broad spectrum of active compounds upon mucosal administration, mucus-penetrating and mucoadhesive particles have been designed by tuning the chemical composition, the size, the density, and the surface properties. The benefits of utilizing nanomaterials that interact intimately with mucosae by different mechanisms in the nanomedicine field have been extensively reported. To ensure the safety of these nanosystems, their compatibility is evaluated in vitro and in vivo in preclinical and clinical trials. Conversely, there is a growing concern about the toxicity of nanomaterials dispersed in air and water effluents that unintentionally come into contact with the airways and the gastrointestinal tract. Thus, deep understanding of the key nanomaterial properties that govern the interplay with mucus and tissues is crucial for the rational design of more efficient drug delivery nanosystems (nanomedicine) and to anticipate the fate and side-effects of nanoparticulate matter upon acute or chronic exposure (nanotoxicology). This review initially overviews the complex structural features of mucosal tissues, including the structure of mucus, the epithelial barrier, the mucosal-associated lymphatic tissues and microbiota. Then, the most relevant investigations attempting to identify and validate the key particle features that govern nanomaterial-mucosa interactions and that are relevant in both nanomedicine and nanotoxicology are discussed in a holistic manner. Finally, the most popular experimental techniques and the incipient use of mathematical and computational models to characterize these interactions are described.
Collapse
Affiliation(s)
- José das Neves
- i3S - Instituto de Investigação e Inovação em Saúde & INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Roni Sverdlov Arzi
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Haifa, 3200003, Israel.
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Haifa, 3200003, Israel.
| |
Collapse
|
22
|
Shoueir KR, El-Desouky N, Rashad MM, Ahmed MK, Janowska I, El-Kemary M. Chitosan based-nanoparticles and nanocapsules: Overview, physicochemical features, applications of a nanofibrous scaffold, and bioprinting. Int J Biol Macromol 2021; 167:1176-1197. [PMID: 33197477 DOI: 10.1016/j.ijbiomac.2020.11.072] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/29/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022]
Abstract
Recent advancements in the synthesis, properties, and applications of chitosan as the second after cellulose available biopolymer in nature were discussed in this review. A general overview of processing and production procedures from A to Z was highlighted. Chitosan exists in three polymorphic forms which differ in degree of crystallinity (α, β, and γ). Thus, the degree of deacetylation, crystallinity, surface area, and molecular mass significantly affect most applications. Otherwise, the synthesis of chitosan nanofibers is suffering from many drawbacks that were recently treated by co-electrospun with other polymers such as polyvinyl alcohol (PVA), polyethylene oxide (PEO), and polycaprolactone (PCL). Ultimately, this review focuses on the area of new trend utilization of chitosan nanoparticles as nanospheres and nanocapsules, in cartilage and bone regenerative medicine. Owing to its biocompatibility, bioavailability, biodegradability, and costless synthesis, chitosan is a promising biopolymeric structure for water remediation, drug delivery, antimicrobials, and tissue engineering.
Collapse
Affiliation(s)
- Kamel R Shoueir
- Institute of Nanoscience & Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt; Institut de Chimie et Procédés pour l'Énergie, l'Environnement et la Santé (ICPEES), CNRS UMR 7515-Université de Strasbourg, 25 rue Becquerel 67087 Strasbourg, France.
| | - Nagwa El-Desouky
- Institute of Nanoscience & Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt
| | - Moataz M Rashad
- Institute of Nanoscience & Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt
| | - M K Ahmed
- Department of Physics, Faculty of Science, Suez University, Suez, 43518, Egypt
| | - Izabela Janowska
- Institut de Chimie et Procédés pour l'Énergie, l'Environnement et la Santé (ICPEES), CNRS UMR 7515-Université de Strasbourg, 25 rue Becquerel 67087 Strasbourg, France
| | - Maged El-Kemary
- Institute of Nanoscience & Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt; Pharos University, Alexandria, Egypt.
| |
Collapse
|
23
|
Schlachet I, Moshe Halamish H, Sosnik A. Mixed Amphiphilic Polymeric Nanoparticles of Chitosan, Poly(vinyl alcohol) and Poly(methyl methacrylate) for Intranasal Drug Delivery: A Preliminary In Vivo Study. Molecules 2020; 25:molecules25194496. [PMID: 33008001 PMCID: PMC7582691 DOI: 10.3390/molecules25194496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 01/10/2023] Open
Abstract
Intranasal (i.n.) administration became an alternative strategy to bypass the blood-brain barrier and improve drug bioavailability in the brain. The main goal of this work was to preliminarily study the biodistribution of mixed amphiphilic mucoadhesive nanoparticles made of chitosan-g-poly(methyl methacrylate) and poly(vinyl alcohol)-g-poly(methyl methacrylate) and ionotropically crosslinked with sodium tripolyphosphate in the brain after intravenous (i.v.) and i.n. administration to Hsd:ICR mice. After i.v. administration, the highest nanoparticle accumulation was detected in the liver, among other peripheral organs. After i.n. administration of a 10-times smaller nanoparticle dose, the accumulation of the nanoparticles in off-target organs was much lower than after i.v. injection. In particular, the accumulation of the nanoparticles in the liver was 20 times lower than by i.v. When brains were analyzed separately, intravenously administered nanoparticles accumulated mainly in the "top" brain, reaching a maximum after 1 h. Conversely, in i.n. administration, nanoparticles were detected in the "bottom" brain and the head (maximum reached after 2 h) owing to their retention in the nasal mucosa and could serve as a reservoir from which the drug is released and transported to the brain over time. Overall, results indicate that i.n. nanoparticles reach similar brain bioavailability, though with a 10-fold smaller dose, and accumulate in off-target organs to a more limited extent and only after redistribution through the systemic circulation. At the same time, both administration routes seem to lead to differential accumulation in brain regions, and thus, they could be beneficial in the treatment of different medical conditions.
Collapse
|
24
|
de Oliveira Junior ER, Santos LCR, Salomão MA, Nascimento TL, de Almeida Ribeiro Oliveira G, Lião LM, Lima EM. Nose-to-brain drug delivery mediated by polymeric nanoparticles: influence of PEG surface coating. Drug Deliv Transl Res 2020; 10:1688-1699. [PMID: 32613550 DOI: 10.1007/s13346-020-00816-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Intranasal administration of mucus-penetrating nanoparticles is an emerging trend to increase drug delivery to the brain. In order to overcome rapid nasal mucociliary clearance, low epithelial permeation, and local enzymatic degradation, we investigated the influence of PEGylation on nose-to-brain delivery of polycaprolactone (PCL) nanoparticles (PCL-NPs) encapsulating bexarotene, a potential neuroprotective compound. PEGylation with 1, 3, 5, and 10% PCL-PEG did not affect particle diameter or morphology. Upon incubation with artificial nasal mucus, only 5 and 10% of PCL-PEG coating were able to ensure NP stability and homogeneity in mucus. Rapid mucus-penetrating ability was observed for 98.8% of PCL-PEG5% NPs and for 99.5% of PCL-PEG10% NPs. Conversely, the motion of non-modified PCL-NPs was markedly slower. Fluorescence microscopy showed that the presence of PEG on NP surface did not reduce their uptake by RMPI 2650 cells. Fluorescence tomography images evidenced higher translocation into the brain for PCL-PEG5% NPs. Bexarotene loaded into PCL-PEG5% NPs resulted in area under the curve in the brain (AUCbrain) 3 and 2-fold higher than that for the drug dispersion and for non-PEGylated NPs (p < 0.05), indicating that approximately 4% of the dose was directly delivered to the brain. Combined, these results indicate that PEGylation of PCL-NPs with PCL-PEG5% is able to reduce NP interactions with the mucus, leading to a more efficient drug delivery to the brain following intranasal administration. Graphical abstract.
Collapse
Affiliation(s)
- Edilson Ribeiro de Oliveira Junior
- FarmaTec - Centro de PD&I de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Goiás - UFG, 5ª Avenida c/Rua 240 s/n, Praça Universitária, Goiânia, GO, 74605-170, Brazil
| | - Lílian Cristina Rosa Santos
- FarmaTec - Centro de PD&I de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Goiás - UFG, 5ª Avenida c/Rua 240 s/n, Praça Universitária, Goiânia, GO, 74605-170, Brazil
| | - Mariana Arraes Salomão
- FarmaTec - Centro de PD&I de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Goiás - UFG, 5ª Avenida c/Rua 240 s/n, Praça Universitária, Goiânia, GO, 74605-170, Brazil
| | - Thais Leite Nascimento
- FarmaTec - Centro de PD&I de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Goiás - UFG, 5ª Avenida c/Rua 240 s/n, Praça Universitária, Goiânia, GO, 74605-170, Brazil
| | | | - Luciano Morais Lião
- LabRMN, Instituto de Química, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Eliana Martins Lima
- FarmaTec - Centro de PD&I de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Goiás - UFG, 5ª Avenida c/Rua 240 s/n, Praça Universitária, Goiânia, GO, 74605-170, Brazil.
| |
Collapse
|
25
|
Cronin JG, Jones N, Thornton CA, Jenkins GJS, Doak SH, Clift MJD. Nanomaterials and Innate Immunity: A Perspective of the Current Status in Nanosafety. Chem Res Toxicol 2020; 33:1061-1073. [PMID: 32307980 DOI: 10.1021/acs.chemrestox.0c00051] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human exposure to engineered nanomaterials (ENMs) is inevitable due to the plethora of applications for which they are being manufactured and integrated within. ENMs demonstrate plentiful advantages in terms of industrial approaches as well as from a consumer perspective. However, despite such positives, doubts remain over the human health implications of ENM exposure. In light of the increased research focus upon the potential effects of ENM exposure to human health in recent decades, questions still remain regarding the safety of these highly advanced, precision-tuned physical entities. The risk of short-term, high-dose exposure to humans is considered relatively low, although this has formed the direction of the hazard-assessment community since the turn of the 21st century. However, the possibility of humans being exposed repeatedly over a long period of time to a low-dose of ENMs of varying physicochemical characteristics is of significant concern, and thus, industry, government, academic, and consumer agencies are only now beginning to consider this. Notably, when considering the human health implications of such low-dose, long-term, repeated exposure scenarios, the impact of ENMs upon the human immune system is of primary importance. However, there remains a real need to understand the impact of ENMs upon the human immune system, especially the innate immune system, at all stages of life, given exposure to nanosized particles begins before birth, that is, of the fetus. Therefore, the purpose of this perspective is to summarize what is currently known regarding ENM exposure of different components of the innate immune system and identify knowledge gaps that should be addressed if we are to fully deduce the impact of ENM exposure on innate immune function.
Collapse
Affiliation(s)
- James G Cronin
- Immunometabolism & Cancer Research Group, Institute of Life Science, Swansea University Medical School, Swansea, Wales SA2 8PP, U.K
| | - Nicholas Jones
- Human Immunology Research Group, Institute of Life Science, Swansea University Medical School, Swansea, Wales SA2 8PP, U.K
| | - Catherine A Thornton
- Human Immunology Research Group, Institute of Life Science, Swansea University Medical School, Swansea, Wales SA2 8PP, U.K
| | - Gareth J S Jenkins
- In Vitro Toxicology Group, Institute of Life Science, Swansea University Medical School, Swansea, Wales SA2 8PP, U.K
| | - Shareen H Doak
- In Vitro Toxicology Group, Institute of Life Science, Swansea University Medical School, Swansea, Wales SA2 8PP, U.K
| | - Martin J D Clift
- In Vitro Toxicology Group, Institute of Life Science, Swansea University Medical School, Swansea, Wales SA2 8PP, U.K
| |
Collapse
|
26
|
Dalpiaz A, Fogagnolo M, Ferraro L, Beggiato S, Hanuskova M, Maretti E, Sacchetti F, Leo E, Pavan B. Bile salt-coating modulates the macrophage uptake of nanocores constituted by a zidovudine prodrug and enhances its nose-to-brain delivery. Eur J Pharm Biopharm 2019; 144:91-100. [PMID: 31521715 DOI: 10.1016/j.ejpb.2019.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/26/2019] [Accepted: 09/08/2019] [Indexed: 02/07/2023]
Abstract
We have previously demonstrated that the ester conjugation of zidovudine (AZT) with ursodeoxycholic acid (UDCA) allows to obtain a prodrug (U-AZT) which eludes the active efflux transporters (AET). This allows the prodrug to more efficiently permeates and remains in murine macrophages than the parent compound. Here we demonstrate that U-AZT can be formulated, by a nanoprecipitation method, as nanoparticle cores coated by bile acid salt (taurocholate or ursodeoxycholate) corona, without any other excipients. The U-AZT nanoparticles appeared spherical with a mean diameter of ∼200 nm and a zeta potential of ∼-55 mV. During the incubation (5 h) in fetal bovine serum, the ursodeoxycholate-coated nanoparticle size did not change. Differently, taurocholate-coated particle size was firstly reduced and then increased up to 800 µm, thus suggesting the high aptitude of these nanoparticles to interact with serum proteins. The in vitro uptake of taurocholate coated particles by murine macrophages was strongly higher than that of ursodeoxycholate-coated particles or free U-AZT (∼500% and ∼7000%, respectively). AZT was also detected in macrophages following the prodrug uptake, with the greatest amounts observed after the taurocholate-coated nanoparticle incubation. As macrophages in the subarachnoid spaces of cerebrospinal fluid (CSF) constitute one of the most unreachable HIV sanctuaries in the body, we also tested the ability of taurocholate-coated nanoparticles (i.e., nanoparticles highly internalized by macrophages) to reach them after their nasal administration in the presence or absence of chitosan. The results indicate that chitosan allowed to obtain a relatively high uptake (up to 4 µg/ml) of U-AZT in CSF. Taking into account that chitosan may promote the direct brain nanoparticle uptake, these findings can be considered an initial step toward the in vivo targeting of the subarachnoid macrophages by U-AZT prodrug.
Collapse
Affiliation(s)
- Alessandro Dalpiaz
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Fossato di Mortara 19, I-44121 Ferrara, Italy.
| | - Marco Fogagnolo
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Fossato di Mortara 19, I-44121 Ferrara, Italy.
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara and LTTA Center, Via L. Borsari 46, I-44121 Ferrara, Italy.
| | - Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara and LTTA Center, Via L. Borsari 46, I-44121 Ferrara, Italy.
| | - Miriam Hanuskova
- "Enzo Ferrari" Engineering Department, University of Modena and Reggio Emilia, Via Pietro Vivarelli 10, I-41125 Modena, Italy.
| | - Eleonora Maretti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, I-41125 Modena, Italy.
| | - Francesca Sacchetti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, I-41125 Modena, Italy.
| | - Eliana Leo
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, I-41125 Modena, Italy.
| | - Barbara Pavan
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy.
| |
Collapse
|