1
|
Sharma G, Wadhwa K, Kumar S, Singh G, Pahwa R. Revolutionizing Parkinson's treatment: Harnessing the potential of intranasal nanoemulsions for targeted therapy. Drug Deliv Transl Res 2025:10.1007/s13346-024-01770-z. [PMID: 39777646 DOI: 10.1007/s13346-024-01770-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2024] [Indexed: 01/11/2025]
Abstract
Parkinson's disease (PD) is the most prominent and highly prevalent chronic neuro-degenerative disease generally recognized by classical motor symptoms which are linked with genetic mutation, Lewy bodies, and subsequently selective loss of nigrostriatal dopaminergic neurons. The blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier protect the central nervous system against toxins and are the most significant barriers to effective brain drug delivery in managing Parkinsonism. In recent years, intranasal delivery has attracted remarkable attention for brain targeting as the drug can be administered to the brain directly from the nose employing the trigeminal and olfactory pathways. For brain targeting through nasal delivery, several advanced and promising formulation techniques have been investigated globally. Nanoemulsions are regarded as an innovative carrier approach for PD, where these provide targeted administration and enhanced bioavailability of neurotherapeutics. This manuscript provides deeper insight into the pathophysiology of PD, various drug delivery strategies to overcome BBB, and the potential role of nanoemulsions via the intranasal route. Various research findings on the intranasal administration of nanoemulsions and their pivotal applications in the treatment of PD have also been embarked. The potential role of phytoconstituents and surface-modified nanoemulsions for the effective treatment of PD has also been reflected along with current challenges and future perspectives in this avenue.
Collapse
Affiliation(s)
- Gulshan Sharma
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, Haryana, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| | - Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology (MIET), NH-58 Delhi-Roorkee Highway, Meerut, 250005, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Rakesh Pahwa
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, Haryana, India.
| |
Collapse
|
2
|
Botan MVG, da Silva JB, Bruschi ML. Technological Strategies Applied to Pharmaceutical Systems for Intranasal Administration of Drugs Intended for Neurological Treatments: A Review. AAPS PharmSciTech 2024; 25:258. [PMID: 39487374 DOI: 10.1208/s12249-024-02974-9] [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: 05/17/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
The complexity of treating neurological diseases has meant that new strategies have had to be developed to deliver drugs to the brain more efficiently and safely. Intranasal drug delivery is characterized by its ease of administration, safety, and rapid delivery directly from the nose to the brain. Several strategies have been developed to improve the delivery of drugs to the brain via nasal administration. These include the use of mucoadhesive and thermoresponsive polymers and their combination into polymer blends, as well as the use of liposomes, niosomes, and nano- and microemulsions. Therefore, this review focuses on technologies for developing pharmaceutical systems aimed at delivery via the nose to the brain, contributing to new treatments for difficult neurological disorders. Some of the most common and difficult-to-treat neurological conditions, the intranasal route of administration, and the anatomy of the nasal cavity have been discussed, as well as factors that may influence the absorption of drugs administered into the nose. The types of intranasal formulations and the devices that can be used to administer these products are also discussed in this review. Strategies for improving the transport of bioactive agents and increasing bioavailability are highlighted. The technologies discussed in this review can facilitate the development of formulations with improved properties, such as drug release and mucoadhesiveness, which have several advantages for patients requiring complex neurological treatments.
Collapse
Affiliation(s)
- Maria Vitoria Gouveia Botan
- Postgraduate Program in Pharmaceutical Sciences, Laboratory of Research and Development of Drug Delivery Systems, Department of Pharmacy, State University of Maringa, Maringa, PR, Brazil
| | - Jéssica Bassi da Silva
- Postgraduate Program in Pharmaceutical Sciences, Laboratory of Research and Development of Drug Delivery Systems, Department of Pharmacy, State University of Maringa, Maringa, PR, Brazil
| | - Marcos Luciano Bruschi
- Postgraduate Program in Pharmaceutical Sciences, Laboratory of Research and Development of Drug Delivery Systems, Department of Pharmacy, State University of Maringa, Maringa, PR, Brazil.
| |
Collapse
|
3
|
Satapathy BS, Mishra A, Biswal SK, Pattnaik S, Parida R, Biswal B, Shaw TK. Encapsulation of Alpinia leaf essential oil in nanophytosome-embedded gel as novel strategy to treat periodontal infections: evaluation of antimicrobial effectiveness, pharmacokinetic, in vitro-ex vivo correlation and in silico studies. J Microencapsul 2024; 41:327-344. [PMID: 38829223 DOI: 10.1080/02652048.2024.2354234] [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: 12/22/2023] [Accepted: 05/08/2024] [Indexed: 06/05/2024]
Abstract
AIM The work reports a novel nanophytosomal gel encapsulating Alpinia galanga (L.) Willd leaf essential oil to treat periodontal infections. METHODS Alpinia oil-loaded nanophytosomes (ANPs) were formulated by lipid layer hydration technique and were evaluated by FESEM, cryo-TEM, loading efficiency, zeta potential, particle size, release profile etc. Selected ANPs-loaded gel (ANPsG) was evaluated by both in vitro and in vivo methods. RESULTS Selected ANPs were spherical, unilamellar, 49.32 ± 2.1 nm size, 0.45 PDI, -46.7 ± 0.8 mV zeta potential, 9.8 ± 0.5% (w/w) loading, 86.4 ± 3.02% (w/w) loading efficiency with sustained release profile. ANPsG showed good spreadability (6.8 ± 0.3 gm.cm/sec), extrudability (79.33 ± 1.5%), viscosity (36522 ± 0.82 cps), mucoadhesive strength (44.56 ± 3.5 gf) with sustained ex vivo release tendency. Satisfied ZOI and MIC was observed for ANPsG against periodontal bacteria vs. standard/control. ANPsG efficiently treated infection in ligature induced periodontitis model. Key pharmacokinetic parameters like AUC, MRT, Vd were enhanced for ANPsG. CONCLUSION ANPsG may be investigated for futuristic clinical studies.
Collapse
Affiliation(s)
| | - Abhishek Mishra
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, India
| | | | - Snigdha Pattnaik
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, India
| | - Reena Parida
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, India
| | - Biswabhusan Biswal
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, India
| | - Tapan Kumar Shaw
- Department of Pharmaceutical Technology, JIS University, Kolkata, India
| |
Collapse
|
4
|
Zhao P, Wu T, Tian Y, You J, Cui X. Recent advances of focused ultrasound induced blood-brain barrier opening for clinical applications of neurodegenerative diseases. Adv Drug Deliv Rev 2024; 209:115323. [PMID: 38653402 DOI: 10.1016/j.addr.2024.115323] [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: 09/29/2023] [Revised: 12/21/2023] [Accepted: 04/20/2024] [Indexed: 04/25/2024]
Abstract
With the aging population on the rise, neurodegenerative disorders have taken center stage as a significant health concern. The blood-brain barrier (BBB) plays an important role to maintain the stability of central nervous system, yet it poses a formidable obstacle to delivering drugs for neurodegenerative disease therapy. Various methods have been devised to confront this challenge, each carrying its own set of limitations. One particularly promising noninvasive approach involves the utilization of focused ultrasound (FUS) combined with contrast agents-microbubbles (MBs) to achieve transient and reversible BBB opening. This review provides a comprehensive exploration of the fundamental mechanisms behind FUS/MBs-mediated BBB opening and spotlights recent breakthroughs in its application for neurodegenerative diseases. Furthermore, it addresses the current challenges and presents future perspectives in this field.
Collapse
Affiliation(s)
- Pengxuan Zhao
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Pharmacy, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Tiantian Wu
- School of Pharmacy, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Yu Tian
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai 200000, China
| | - Jia You
- School of Pharmacy, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Xinwu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
5
|
Kline-Schoder AR, Chintamen S, Willner MJ, DiBenedetto MR, Noel RL, Batts AJ, Kwon N, Zacharoulis S, Wu CC, Menon V, Kernie SG, Konofagou EE. Characterization of the responses of brain macrophages to focused ultrasound-mediated blood-brain barrier opening. Nat Biomed Eng 2024; 8:650-663. [PMID: 37857722 PMCID: PMC11734153 DOI: 10.1038/s41551-023-01107-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 09/16/2023] [Indexed: 10/21/2023]
Abstract
The opening of the blood-brain barrier (BBB) by focused ultrasound (FUS) coupled with intravenously injected microbubbles can be leveraged as a form of immunotherapy for the treatment of neurodegenerative disorders. However, how FUS BBB opening affects brain macrophages is not well understood. Here by using single-cell sequencing to characterize the distinct responses of microglia and central nervous system-associated macrophages (CAMs) to FUS-mediated BBB opening in mice, we show that the treatment remodels the immune landscape via the recruitment of CAMs and the proliferation of microglia and via population size increases in disease-associated microglia. Both microglia and CAMs showed early and late increases in population sizes, yet only the proliferation of microglia increased at both timepoints. The population of disease-associated microglia also increased, accompanied by the upregulation of genes associated with gliogenesis and phagocytosis, with the depletion of brain macrophages significantly decreasing the duration of BBB opening.
Collapse
Affiliation(s)
| | - Sana Chintamen
- Department of Neurobiology and Behaviour, Columbia University, New York, NY, USA
| | - Moshe J Willner
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | | - Rebecca L Noel
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Alec J Batts
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Cheng-Chia Wu
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Vilas Menon
- Department of Neurology, Columbia University, New York, NY, USA
| | - Steven G Kernie
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
- Department of Radiology, Columbia University, New York, NY, USA.
| |
Collapse
|
6
|
Vishwas S, Bashir B, Birla D, Khandale N, Chaitanya MVNL, Chellappan DK, Gupta G, Negi P, Dua K, Singh SK. Neuroprotective Role of Phytoconstituents-based Nanoemulsion for the Treatment of Alzheimer's Disease. Curr Top Med Chem 2024; 24:1683-1698. [PMID: 38676489 DOI: 10.2174/0115680266296001240327090111] [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: 11/30/2023] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 04/29/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent form of neurodegenerative disorder (ND), affecting more than 44 million individuals globally as of 2023. It is characterized by cognitive dysfunction and an inability to perform daily activities. The progression of AD is associated with the accumulation of amyloid beta (Aβ), the formation of neurofibrillary tangles (NFT), increased oxidative stress, neuroinflammation, mitochondrial dysfunction, and endoplasmic reticulum stress. Presently, various phytomedicines and their bioactive compounds have been identified for their neuroprotective effects in reducing oxidative stress, alleviating neuroinflammation, and mitigating the accumulation of Aβ and acetylcholinesterase enzymes in the hippocampus and cerebral cortex regions of the brain. However, despite demonstrating promising anti-Alzheimer's effects, the clinical utilization of phytoconstituents remains limited in scope. The key factor contributing to this limitation is the challenges inherent in traditional drug delivery systems, which impede their effectiveness and efficiency. These difficulties encompass insufficient drug targeting, restricted drug solubility and stability, brief duration of action, and a lack of control over drug release. Consequently, these constraints result in diminished bioavailability and insufficient permeability across the blood-brain barrier (BBB). In response to these challenges, novel drug delivery systems (NDDS) founded on nanoformulations have emerged as a hopeful strategy to augment the bioavailability and BBB permeability of bioactive compounds with poor solubility. Among these systems, nanoemulsion (NE) have been extensively investigated for their potential in targeting AD. NE offers several advantages, such as ease of preparation, high drug loading, and high stability. Due to their nanosize droplets, NE also improves gut and BBB permeability leading to enhanced permeability of the drug in systemic circulation and the brain. Various studies have reported the testing of NE-based phytoconstituents and their bioactives in different animal species, including transgenic, Wistar, and Sprague-Dawley (SD) rats, as well as mice. However, transgenic mice are commonly employed in AD research to analyze the effects of Aβ. In this review, various aspects such as the neuroprotective role of various phytoconstituents, the challenges associated with conventional drug delivery, and the need for NDDS, particularly NE, are discussed. Various studies involving phytoconstituent-based NE for the treatment of AD are also discussed.
Collapse
Affiliation(s)
- Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Bushra Bashir
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Devendra Birla
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Nikhil Khandale
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | | | | | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, 302017, Jaipur, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan (Bajhol), Solan, H.P., 173212, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
7
|
Li Z, Dang Q, Wang P, Zhao F, Huang J, Wang C, Liu X, Min W. Food-Derived Peptides: Beneficial CNS Effects and Cross-BBB Transmission Strategies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20453-20478. [PMID: 38085598 DOI: 10.1021/acs.jafc.3c06518] [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: 12/28/2023]
Abstract
Food-derived peptides, as dietary supplements, have significant effects on promoting brain health and relieving central nervous system (CNS) diseases. However, the blood-brain barrier (BBB) greatly limits their in-brain bioavailability. Thus, overcoming the BBB to target the CNS is a major challenge for bioactive peptides in the prevention and treatment of CNS diseases. This review discusses improvement in the neuroprotective function of food-derived active peptides in CNS diseases, as well as the source of BBB penetrating peptides (BBB-shuttles) and the mechanism of transmembrane transport. Notably, this review also discusses various peptide modification methods to overcome the low permeability and stability of the BBB. Lipification, glycosylation, introduction of disulfide bonds, and cyclization are effective strategies for improving the penetration efficiency of peptides through the BBB. This review provides a new prospective for improving their neuroprotective function and developing treatments to delay or even prevent CNS diseases.
Collapse
Affiliation(s)
- Zehui Li
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P.R. China
| | - Qiao Dang
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P.R. China
| | - Peng Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
| | - Fanrui Zhao
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Jianqin Huang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Chongchong Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Xingquan Liu
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
| | - Weihong Min
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| |
Collapse
|
8
|
Zhivodernikov IV, Markina YV, Kirichenko TV, Popov MA, Markin AM. Exosomes as a potential therapeutic approach in osteoimmunology. Front Immunol 2023; 14:1309015. [PMID: 38173718 PMCID: PMC10763248 DOI: 10.3389/fimmu.2023.1309015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Exosomes are natural extracellular vesicles that play a key role in inter- and intracellular communication. Currently they are considered as a promising therapeutic strategy for the treatment of various diseases. In osteoimmunology, exosomes can serve as biomarkers of bone homeostasis disorders and, at the same time, promising therapeutic agents with high stability in the biological environment, low immunogenicity and good bioavailability. In this review, we attempted to examine exosomes as natural mediators of intercellular communication, playing an essential role in the interaction of the immune system and bone tissue, based on an analysis of the PubMed database up to October 2023.
Collapse
Affiliation(s)
- Ivan V. Zhivodernikov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Center of Surgery, Moscow, Russia
| | - Yuliya V. Markina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Center of Surgery, Moscow, Russia
| | - Tatiana V. Kirichenko
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Center of Surgery, Moscow, Russia
| | - Mikhail A. Popov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), Moscow, Russia
| | - Alexander M. Markin
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Center of Surgery, Moscow, Russia
- Medical Institute, Poples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow, Russia
| |
Collapse
|
9
|
Kumar J, Delgado SA, Sarma H, Narayan M. Caffeic acid recarbonization: A green chemistry, sustainable carbon nano material platform to intervene in neurodegeneration induced by emerging contaminants. ENVIRONMENTAL RESEARCH 2023; 237:116932. [PMID: 37598847 PMCID: PMC11285802 DOI: 10.1016/j.envres.2023.116932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/12/2023] [Accepted: 08/18/2023] [Indexed: 08/22/2023]
Abstract
Environmental agents such as pesticides, weedicides and herbicides (collectively referred to as pesticides) are associated with the onset and pathogenesis of neurodegenerative disorders such as Parkinson's (PD) and Alzheimer's (AD) diseases. The development of blood-brain barrier (BBB)-penetrating therapeutic candidates to both prevent and treat the aforementioned xenotoxicant-induced neurodegenerative disorders remains an unmet need. Here, we examine whether caffeic-acid based Carbon Quantum Dots (CACQDs) can intervene in pesticide-associated onset and progress of the PD phenotype. Pulse-chase fluorescence analyses revealed that CACQDs intervene in the soluble-to-toxic transformation of the amyloid-forming protein model Hen Egg White Lysozyme (HEWL). The sp2-rich CACQDs also scavenged free radicals, a milestone along the PD trajectory. In-vitro, CACQDs introduced into a human neuroblastoma-derived cell line (SH-SY5Y) demonstrated negligible cytotoxicity up to 5 mg/mL and protected the cell line against oxidative stress-induced neuronal injury induced by the pesticide and potent neurotoxin, paraquat. Our findings suggest that the potentially BBB-penetrating CACQDs derived from caffeic acid hold promise for mitigating neurodegenerative disorders associated with environmental pesticides and xenobiotic neurotoxicants. Importantly, CACQDs sourced from coffee, coupled with their facile synthesis, represent a sustainable, green chemistry platform for generating interventional candidates in neurodegeneration.
Collapse
Affiliation(s)
- Jyotish Kumar
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, TX, 79968, United States
| | - Sofia A Delgado
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, TX, 79968, United States
| | - Hemen Sarma
- Department of Botany, Bodoland University, Rangalikhata, Deborgaon, Kokrajhar (BTR), Assam, 783370, India.
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, TX, 79968, United States.
| |
Collapse
|
10
|
Reyes-Esteves S, Nong J, Glassman PM, Omo-Lamai S, Ohashi S, Myerson JW, Zamora ME, Ma X, Kasner SE, Sansing L, Muzykantov VR, Marcos-Contreras OA, Brenner JS. Targeted drug delivery to the brain endothelium dominates over passive delivery via vascular leak in experimental intracerebral hemorrhage. J Control Release 2023; 356:185-195. [PMID: 36868517 PMCID: PMC10519578 DOI: 10.1016/j.jconrel.2023.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is one of the most common causes of fatal stroke, yet has no specific drug therapies. Many attempts at passive intravenous (IV) delivery in ICH have failed to deliver drugs to the salvageable area around the hemorrhage. The passive delivery method assumes vascular leak through the ruptured blood-brain barrier will allow drug accumulation in the brain. Here we tested this assumption using intrastriatal injection of collagenase, a well-established experimental model of ICH. Fitting with hematoma expansion in clinical ICH, we showed that collagenase-induced blood leak drops significantly by 4 h after ICH onset and is gone by 24 h. We observed passive-leak brain accumulation also declines rapidly over ∼4 h for 3 model IV therapeutics (non-targeted IgG; a protein therapeutic; PEGylated nanoparticles). We compared these passive leak results with targeted brain delivery by IV monoclonal antibodies (mAbs) that actively bind vascular endothelium (anti-VCAM, anti-PECAM, anti-ICAM). Even at early time points after ICH induction, where there is high vascular leak, brain accumulation via passive leak is dwarfed by brain accumulation of endothelial-targeted agents: At 4 h after injury, anti-PECAM mAbs accumulate at 8-fold higher levels in the brain vs. non-immune IgG; anti-VCAM nanoparticles (NPs) deliver a protein therapeutic (superoxide dismutase, SOD) at 4.5-fold higher levels than the carrier-free therapeutic at 24 h after injury. These data suggest that relying on passive vascular leak provides inefficient delivery of therapeutics even at early time points after ICH, and that a better strategy might be targeted delivery to the brain endothelium, which serves as the gateway for the immune attack on the peri-hemorrhage inflamed brain region.
Collapse
Affiliation(s)
- Sahily Reyes-Esteves
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Jia Nong
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Patrick M Glassman
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, United States of America
| | - Serena Omo-Lamai
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Sarah Ohashi
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States of America
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Marco E Zamora
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Xiaonan Ma
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Scott E Kasner
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Lauren Sansing
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States of America
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Oscar A Marcos-Contreras
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America.
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America; Division of Pulmonary Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America.
| |
Collapse
|
11
|
Wei W, Cardes F, Hierlemann A, Modena MM. 3D In Vitro Blood-Brain-Barrier Model for Investigating Barrier Insults. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205752. [PMID: 36782313 PMCID: PMC10104638 DOI: 10.1002/advs.202205752] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/18/2023] [Indexed: 06/18/2023]
Abstract
Blood-brain-barrier (BBB) disruption has been associated with a variety of central-nervous-system diseases. In vitro BBB models enable to investigate how the barrier reacts to external injury events, commonly referred to as insults. Here, a human-cell-based BBB platform with integrated, transparent electrodes to monitor barrier tightness in real time at high resolution is presented. The BBB model includes human cerebral endothelial cells and primary pericytes and astrocytes in a 3D arrangement within a pump-free, open-microfluidic platform. With this platform, this study demonstrates that oxygen-glucose deprivation (OGD), which mimics the characteristics of an ischemic insult, induces a rapid remodeling of the cellular actin structures and subsequent morphological changes in the endothelial cells. High-resolution live imaging shows the formation of large actin stress-fiber bundles in the endothelial layer during OGD application, which ultimately leads to cell shrinkage and barrier breakage. Simultaneous electrical measurements evidence a rapid decrease of the barrier electrical resistance before the appearance of stress fibers, which indicates that the barrier function is compromised already before the appearance of drastic morphological changes. The results demonstrate that the BBB platform recapitulates the main barrier functions in vitro and can be used to investigate rapid reorganization of the BBB upon application of external stimuli.
Collapse
Affiliation(s)
- Wei Wei
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Fernando Cardes
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Andreas Hierlemann
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Mario M. Modena
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| |
Collapse
|
12
|
Kim K, Lee J, Park MH. Microbubble Delivery Platform for Ultrasound-Mediated Therapy in Brain Cancers. Pharmaceutics 2023; 15:pharmaceutics15020698. [PMID: 36840020 PMCID: PMC9959315 DOI: 10.3390/pharmaceutics15020698] [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: 01/25/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The blood-brain barrier (BBB) is one of the most selective endothelial barriers that protect the brain and maintains homeostasis in neural microenvironments. This barrier restricts the passage of molecules into the brain, except for gaseous or extremely small hydrophobic molecules. Thus, the BBB hinders the delivery of drugs with large molecular weights for the treatment of brain cancers. Various methods have been used to deliver drugs to the brain by circumventing the BBB; however, they have limitations such as drug diversity and low delivery efficiency. To overcome this challenge, microbubbles (MBs)-based drug delivery systems have garnered a lot of interest in recent years. MBs are widely used as contrast agents and are recently being researched as a vehicle for delivering drugs, proteins, and gene complexes. The MBs are 1-10 μm in size and consist of a gas core and an organic shell, which cause physical changes, such as bubble expansion, contraction, vibration, and collapse, in response to ultrasound. The physical changes in the MBs and the resulting energy lead to biological changes in the BBB and cause the drug to penetrate it, thus enhancing the therapeutic effect. Particularly, this review describes a state-of-the-art strategy for fabricating MB-based delivery platforms and their use with ultrasound in brain cancer therapy.
Collapse
Affiliation(s)
- Kibeom Kim
- Department of Chemistry and Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| | - Jungmin Lee
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University, Seoul 01795, Republic of Korea
| | - Myoung-Hwan Park
- Department of Chemistry and Life Science, Sahmyook University, Seoul 01795, Republic of Korea
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Convergence Science, Sahmyook University, Seoul 01795, Republic of Korea
- N to B Co., Ltd., Seoul 01795, Republic of Korea
- Correspondence:
| |
Collapse
|
13
|
Exosome-Based Carrier for RNA Delivery: Progress and Challenges. Pharmaceutics 2023; 15:pharmaceutics15020598. [PMID: 36839920 PMCID: PMC9964211 DOI: 10.3390/pharmaceutics15020598] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
In the last few decades, RNA-based drugs have emerged as a promising candidate to specifically target and modulate disease-relevant genes to cure genetic defects. The key to applying RNA therapy in clinical trials is developing safe and effective delivery systems. Exosomes have been exploited as a promising vehicle for drug delivery due to their nanoscale size, high stability, high biocompatibility, and low immunogenicity. We reviewed and summarized the progress in the strategy and application of exosome-mediated RNA therapy. The challenges of exosomes as a carrier for RNA drug delivery are also elucidated in this article. RNA molecules can be loaded into exosomes and then delivered to targeted cells or tissues via various biochemical or physical approaches. So far, exosome-mediated RNA therapy has shown potential in the treatment of cancer, central nervous system disorders, COVID-19, and other diseases. To further exploit the potential of exosomes for RNA delivery, more efforts should be made to overcome both technological and logistic problems.
Collapse
|
14
|
Lo Celso F, Barone G, Maiuolo L, Algieri V, Cretu C, Calandra P. Dissolution of nitrones in alkylphosphates: A structural study. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
15
|
Li J, Zhou Y, Song L, Yang S, Wang Q, Zhou Y, Zhang XB, Qing Z, Yang R. Brain-targeted Near-Infrared Nanobeacon for In Situ Monitoring H 2S Fluctuation during Epileptic Seizures. Anal Chem 2022; 94:15085-15092. [PMID: 36266763 DOI: 10.1021/acs.analchem.2c03254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Epilepsy is a neurological brain disease, and its recurrent seizures are related to the reductive substance-powered antioxidant defense system (ADS). However, until now, there has been no report on the study of in situ antioxidant fluctuation during epilepsy of varying severity. In this work, hydrogen sulfide (H2S) was selected as the model target, a H2S-responsive near-infrared fluorophore was designed and synthesized, and an amphiphilic molecule was synthesized and modified with angiopep-2 peptide at its hydrophilic terminus. A nanobeacon termed as BFPP was prepared by the formation of micelles with the package of the fluorophore. The nanobeacon was sensitive to H2S, with a low detection limit of 17 nM. The H2S fluctuation in cells can be monitored by fluorescence imaging. In addition, angiopep-2 peptide at the surface of BFPP helps it cross the blood-brain barrier, and near-infrared fluorescence improves in vivo imaging. BFPP revealed that H2S was at a moderate level in the normal brain, but its level was obviously elevated during mild epilepsy because of the activation of the ADS while significantly suppressed during severe epilepsy due to neuronal damage. This approach is generally accessible for other targets by altering the responsive fluorophore, with significance for in situ analysis of brain pathology.
Collapse
Affiliation(s)
- Junbin Li
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Ying Zhou
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Lifei Song
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Sheng Yang
- Laboratory of Chemical Biology & Traditional Chinese Medicine Research, Ministry of Education, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Qianqian Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Yibo Zhou
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Xiao-Bing Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Zhihe Qing
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Ronghua Yang
- Laboratory of Chemical Biology & Traditional Chinese Medicine Research, Ministry of Education, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| |
Collapse
|
16
|
Rhaman MM, Islam MR, Akash S, Mim M, Noor alam M, Nepovimova E, Valis M, Kuca K, Sharma R. Exploring the role of nanomedicines for the therapeutic approach of central nervous system dysfunction: At a glance. Front Cell Dev Biol 2022; 10:989471. [PMID: 36120565 PMCID: PMC9478743 DOI: 10.3389/fcell.2022.989471] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022] Open
Abstract
In recent decades, research scientists, molecular biologists, and pharmacologists have placed a strong emphasis on cutting-edge nanostructured materials technologies to increase medicine delivery to the central nervous system (CNS). The application of nanoscience for the treatment of neurodegenerative diseases (NDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), Huntington’s disease (HD), brain cancer, and hemorrhage has the potential to transform care. Multiple studies have indicated that nanomaterials can be used to successfully treat CNS disorders in the case of neurodegeneration. Nanomedicine development for the cure of degenerative and inflammatory diseases of the nervous system is critical. Nanoparticles may act as a drug transporter that can precisely target sick brain sub-regions, boosting therapy success. It is important to develop strategies that can penetrate the blood–brain barrier (BBB) and improve the effectiveness of medications. One of the probable tactics is the use of different nanoscale materials. These nano-based pharmaceuticals offer low toxicity, tailored delivery, high stability, and drug loading capacity. They may also increase therapeutic effectiveness. A few examples of the many different kinds and forms of nanomaterials that have been widely employed to treat neurological diseases include quantum dots, dendrimers, metallic nanoparticles, polymeric nanoparticles, carbon nanotubes, liposomes, and micelles. These unique qualities, including sensitivity, selectivity, and ability to traverse the BBB when employed in nano-sized particles, make these nanoparticles useful for imaging studies and treatment of NDs. Multifunctional nanoparticles carrying pharmacological medications serve two purposes: they improve medication distribution while also enabling cell dynamics imaging and pharmacokinetic study. However, because of the potential for wide-ranging clinical implications, safety concerns persist, limiting any potential for translation. The evidence for using nanotechnology to create drug delivery systems that could pass across the BBB and deliver therapeutic chemicals to CNS was examined in this study.
Collapse
Affiliation(s)
- Md. Mominur Rhaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mobasharah Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Noor alam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Martin Valis
- Department of Neurology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital, Hradec Králové, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| |
Collapse
|
17
|
Yokel RA. Direct nose to the brain nanomedicine delivery presents a formidable challenge. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1767. [PMID: 34957707 DOI: 10.1002/wnan.1767] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/29/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022]
Abstract
This advanced review describes the anatomical and physiological barriers and mechanisms impacting nanomedicine translocation from the nasal cavity directly to the brain. There are significant physiological and anatomical differences in the nasal cavity, olfactory area, and airflow reaching the olfactory epithelium between humans and experimentally studied species that should be considered when extrapolating experimental results to humans. Mucus, transporters, and tight junction proteins present barriers to material translocation across the olfactory epithelium. Uptake of nanoparticles through the olfactory mucosa and translocation to the brain can be intracellular via cranial nerves (intraneuronal) or other cells of the olfactory epithelium, or extracellular along cranial nerve pathways (perineural) and surrounding blood vessels (perivascular, the glymphatic system). Transport rates vary greatly among the nose to brain pathways. Nanomedicine physicochemical properties (size, surface charge, surface coating, and particle stability) can affect uptake efficiency, which is usually less than 5%. Incorporation of therapeutic agents in nanoparticles has been shown to produce pharmacokinetic and pharmacodynamic benefits. Assessment of adverse effects has included olfactory mucosa toxicity, ciliotoxicity, and olfactory bulb and brain neurotoxicity. The results have generally suggested the investigated nanomedicines do not present significant toxicity. Research needs to advance the understanding of nanomedicine translocation and its drug cargo after intranasal administration is presented. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials.
Collapse
Affiliation(s)
- Robert A Yokel
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
18
|
Bai X, Bian Z. MicroRNA-21 Is a Versatile Regulator and Potential Treatment Target in Central Nervous System Disorders. Front Mol Neurosci 2022; 15:842288. [PMID: 35173580 PMCID: PMC8841607 DOI: 10.3389/fnmol.2022.842288] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of endogenous, non-coding, single-stranded RNAs with a length of approximately 22 nucleotides that are found in eukaryotes. miRNAs are involved in the regulation of cell differentiation, proliferation, invasion, apoptosis, and metabolism by regulating the expression of their target genes. Emerging studies have suggested that various miRNAs play key roles in the pathogenesis of central nervous system (CNS) disorders and may be viable therapeutic targets. In particular, miR-21 has prominently emerged as a focus of increasing research on the mechanisms of its involvement in CNS disorders. Herein, we reviewed recent studies on the critical roles of miR-21, including its dysregulated expression and target genes, in the regulation of pathophysiological processes of CNS disorders, with a special focus on apoptosis and inflammation. Collectively, miR-21 is a versatile regulator in the progression of CNS disorders and could be a promising biomarker and therapeutic target for these diseases. An in-depth understanding of the mechanisms by which miR-21 affects the pathogenesis of CNS disorders could pave the way for miR-21 to serve as a therapeutic target for these conditions.
Collapse
Affiliation(s)
- Xue Bai
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhigang Bian
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Zhigang Bian,
| |
Collapse
|
19
|
Gulcan HO, Kosar M. The hybrid compounds as multi-target ligands for the treatment of Alzheimer's Disease: Considerations on Donepezil. Curr Top Med Chem 2021; 22:395-407. [PMID: 34766890 DOI: 10.2174/1568026621666211111153626] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 08/31/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
The strategies to combat Alzheimer's Disease (AD) have been changing with respect to the failures of many drug candidates assessed in clinical studies, the complex pathophysiology of AD, and the limitations of the current drugs employed. So far, none of the targets, either validated or nonvalidated, have been shown to be purely causative in the generation and development of AD. Considering the progressive and the neurodegenerative characteristics of the disease, the main strategy has been based on the design of molecules capable of showing activity on more than one receptor, and it is defined as multi-target ligand design strategy. The hybrid molecule concept is an outcome of this approach. Donepezil, as one of the currently employed drugs for AD therapy, has also been utilized in hybrid drug design studies. This review has aimed to present the promising donepezil-like hybrid molecules introduced in the recent period. Particularly, multi-target ligands with additional activities concomitant to cholinesterase inhibition are preferred.
Collapse
Affiliation(s)
- Hayrettin Ozan Gulcan
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, T.R. North Cyprus, via Mersin 10, Turkey
| | - Muberra Kosar
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, T.R. North Cyprus, via Mersin 10, Turkey
| |
Collapse
|
20
|
Schwering C, Kammler G, Wibbeler E, Christner M, Knobloch JKM, Nickel M, Denecke J, Baehr M, Schulz A. Development of the "Hamburg Best Practice Guidelines for ICV-Enzyme Replacement therapy (ERT) in CLN2 Disease" Based on 6 Years Treatment Experience in 48 Patients. J Child Neurol 2021; 36:635-641. [PMID: 33543660 PMCID: PMC8255505 DOI: 10.1177/0883073821989154] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Intracerebroventricular enzyme replacement therapy (ICV-ERT) for CLN2 disease represents the first approved treatment for neuronal ceroid lipofuscinosis (NCL) diseases. It is the first treatment where a recombinant lysosomal enzyme, cerliponase alfa, is administered into the lateral cerebral ventricles to reach the central nervous system, the organ affected in CLN2 disease. If untreated, CLN2 children show first symptoms such as epilepsy and language developmental delay at 2-4 years followed by rapid loss of motor and language function, vision loss, and early death. Treatment with cerliponase alfa has shown to slow the rapid neurologic decline. However, the mode of administration by 4 hour-long intracerebroventricular infusions every 14 days represents a potentially greater risk of infection compared to intravenous enzyme replacement therapies. The Hamburg NCL Specialty Clinic was the first site worldwide to perform intracerebroventricular enzyme replacement therapy in children with CLN2 disease. In order to ensure maximum patient safety, we analysed data from our center from more than 3000 intracerebroventricular enzyme replacement therapies in 48 patients over 6 years with regard to the occurrence of device-related adverse events and device infections. Since starting intracerebroventricular enzyme replacement therapy, we have also developed and continuously improved the "Hamburg Best Practice Guidelines for ICV-Enzyme Replacement Therapy (ERT) in CLN2 Disease." Results from this study showed low rates for device-related adverse events and infections with 0.27% and 0.33%, respectively. Therefore, following our internal procedural guidelines has shown to improve standardization and patient safety of intracerebroventricular enzyme replacement therapy for CLN2 disease.
Collapse
Affiliation(s)
- Christoph Schwering
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Christoph Schwering, Department of Pediatrics, Children’s Hospital, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.
| | - Gertrud Kammler
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Wibbeler
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Christner
- Institute of Medical Microbiology, University Medical Center Hamburg-Eppendorf, Virology and Hygiene, Hamburg, Germany
| | - Johannes K.-M. Knobloch
- Institute of Medical Microbiology, University Medical Center Hamburg-Eppendorf, Virology and Hygiene, Hamburg, Germany
| | - Miriam Nickel
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Denecke
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Baehr
- University Medical Center Hamburg-Eppendorf, Pharmacy, Hamburg, Germany
| | - Angela Schulz
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
Gratpain V, Mwema A, Labrak Y, Muccioli GG, van Pesch V, des Rieux A. Extracellular vesicles for the treatment of central nervous system diseases. Adv Drug Deliv Rev 2021; 174:535-552. [PMID: 33991589 DOI: 10.1016/j.addr.2021.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/14/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
The interest in extracellular vesicles (EVs) increased during the last decade. It is now established that these vesicles play a role in the pathogenesis of central nervous system diseases (CNS), which explains why they are studied as biomarkers in these pathologies. On the other hand, EVs can also present therapeutic properties, often similar to their parent cells, as observed with mesenchymal stem cell-derived EVs. They can then be used as therapeutics, alone or combined with a bioactive molecule, for the treatment of CNS diseases, as they can cross the blood-brain barrier more easily than synthetic nanomedicines and are less immunogenic. A few clinical trials are currently on-going but there are still challenges to overcome for further clinical translation such as the scale-up of the production, the lack of standardization for isolation and characterization methods and the low encapsulation efficiency.
Collapse
Affiliation(s)
- Viridiane Gratpain
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium
| | - Ariane Mwema
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium; Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium
| | - Yasmine Labrak
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium; Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium
| | - Giulio G Muccioli
- Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium
| | - Vincent van Pesch
- Institute of Neuroscience, Neurochemistry Unit, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium; Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Anne des Rieux
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium.
| |
Collapse
|
22
|
Guerrab W, El Jemli M, Akachar J, Demirtaş G, Mague JT, Taoufik J, Ibrahimi A, Ansar M, Alaoui K, Ramli Y. Design, synthesis, structural and molecular characterization, toxicity, psychotropic activity and molecular docking evaluation of a novel phenytoin derivative: 3-decyl-5,5-diphenylimidazolidine-2,4-dione. J Biomol Struct Dyn 2021; 40:8765-8782. [PMID: 33970810 DOI: 10.1080/07391102.2021.1922096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The hydantoin scaffold is of substantial importance and it is commonly used in drug discovery. Herein, we report the synthesis of a novel phenytoine (a hydantoin derivative) with high yield by the reaction of phenytoin with 1-bromodecyl agent. Namely, 3-decyl-5,5- diphenylimidazolidine-2,4-dione (3DDID). The optimized geometry of the compound was calculated using density functional theory (DFT) method by B3LYP with 6-311++G(d,p) basis set. For this calculation, the X-ray data were used as initial values. Molecular electrostatic potential (MEP) surface and Frontier molecular orbitals (FOMs) were prepared for the compound. The crystal structure of the title compound contains intermolecular N-H···O, C-H···O hydrogen bonds and weak C-H···π interactions. Hirshfeld surface analysis and 2D fingerprint plots of the molecule aid comparison of intermolecular interactions and these analysis reveals that two close contacts are associated with intermolecular hydrogen bonds. The psychotropic activity evaluation of the synthesized compound was further explored using hole bored test for exploratory behaviors, dark//light box test for anxiolytic activity and Rota-road, traction, chimney testes were used to assess the myrelaxant effect. In addition, molecular modeling study was also conducted to rationalize the potential as neurotherapeutic drugs of our synthesized compound by predicting their binding modes, binding affinities and optimal orientation at the active site of the GABA-A receptor and Na+ channel. Finally, in silico ADMET predictions was also examined. HighlightsSynthesis, structural, and molecular characterization of a novel phenytoin derivative.DFT, XRD, and the Hirshfeld surface analysis of crystal structure was studied.Acute toxicity and psychotropic activity evaluation of 3-decyl-5,5 diphenylimidazolidine-2,4-dione (3DDID).Molecular modeling studies have been conducted to rationalize the obtained data and to determine the probable binding mode.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Walid Guerrab
- Laboratory of Medicinal Chemistry, Drug Sciences Research Center, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Meryem El Jemli
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Jihane Akachar
- Laboratoire of Biotechnologie, Faculté de Médecine et de Pharmacie de rabat, Université Mohamed V, Rabat, Morocco
| | - Güneş Demirtaş
- Faculty of Arts and Sciences, Department of Physics, Ondokuz Mayıs University, Samsun, Turkey
| | - Joel T Mague
- Department of Chemistry, Tulane University, New Orleans, LA, USA
| | - Jamal Taoufik
- Laboratory of Medicinal Chemistry, Drug Sciences Research Center, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Azeddine Ibrahimi
- Laboratoire of Biotechnologie, Faculté de Médecine et de Pharmacie de rabat, Université Mohamed V, Rabat, Morocco
| | - M'Hammed Ansar
- Laboratory of Medicinal Chemistry, Drug Sciences Research Center, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Katim Alaoui
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Youssef Ramli
- Laboratory of Medicinal Chemistry, Drug Sciences Research Center, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
23
|
Zhou X, Smith QR, Liu X. Brain penetrating peptides and peptide-drug conjugates to overcome the blood-brain barrier and target CNS diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1695. [PMID: 33470550 DOI: 10.1002/wnan.1695] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Nearly one in six people worldwide suffer from disorders of the central nervous system (CNS). There is an urgent need for effective strategies to improve the success rates in CNS drug discovery and development. The lack of effective technologies for delivering drugs and genes to the brain due to the blood-brain barrier (BBB), a structural barrier that effectively blocks most neurotherapeutic agents from reaching the brain, has posed a formidable hurdle for CNS drug development. Brain-homing and brain-penetrating molecular transport vectors, such as brain permeable peptides or BBB shuttle peptides, have shown promise in overcoming the BBB and ferrying the drug molecules to the brain. The BBB shuttle peptides are discovered by phage display technology or derived from natural neurotropic proteins or certain viruses and harness the receptor-mediated transcytosis molecular machinery for crossing the BBB. Brain permeable peptide-drug conjugates (PDCs), composed of BBB shuttle peptides, linkers, and drug molecules, have emerged as a promising CNS drug delivery system by taking advantage of the endogenous transcytosis mechanism and tricking the brain into allowing these bioactive molecules to pass the BBB. Here, we examine the latest development of brain-penetrating peptide shuttles and brain-permeable PDCs as molecular vectors to deliver small molecule drug payloads across the BBB to reach brain parenchyma. Emerging knowledge of the contribution of the peptides and their specific receptors expressed on the brain endothelial cells, choice of drug payloads, the design of PDCs, brain entry mechanisms, and delivery efficiency to the brain are highlighted. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Quentin R Smith
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| |
Collapse
|
24
|
Bellotti E, Schilling AL, Little SR, Decuzzi P. Injectable thermoresponsive hydrogels as drug delivery system for the treatment of central nervous system disorders: A review. J Control Release 2021; 329:16-35. [PMID: 33259851 DOI: 10.1016/j.jconrel.2020.11.049] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
Abstract
The central nervous system (CNS), consisting of the brain, spinal cord, and retina, superintends to the acquisition, integration and processing of peripheral information to properly coordinate the activities of the whole body. Neurodegenerative and neurodevelopmental disorders, trauma, stroke, and brain tumors can dramatically affect CNS functions resulting in serious and life-long disabilities. Globally, the societal and economic burden associated with CNS disorders continues to grow with the ageing of the population thus demanding for more effective and definitive treatments. Despite the variety of clinically available therapeutic molecules, medical interventions on CNS disorders are mostly limited to treat symptoms rather than halting or reversing disease progression. This is attributed to the complexity of the underlying disease mechanisms as well as to the unique biological microenvironment. Given its central importance, multiple barriers, including the blood brain barrier and the blood cerebrospinal fluid barrier, protect the CNS from external agents. This limits the access of drug molecules to the CNS thus contributing to the modest therapeutic successes. Loco-regional therapies based on the deposition of thermoresponsive hydrogels loaded with therapeutic agents and cells are receiving much attention as an alternative and potentially more effective approach to manage CNS disorders. In this work, the current understanding and challenges in the design of thermoresponsive hydrogels for CNS therapy are reviewed. First, the biological barriers that hinder mass and drug transport to the CNS are described, highlighting the distinct features of each barrier. Then, the realization, characterization and biomedical application of natural and synthetic thermoresponsive hydrogels are critically presented. Advantages and limitations of each design and application are discussed with the objective of identifying general rules that could enhance the effective translation of thermoresponsive hydrogel-based therapies for the treatment of CNS disorders.
Collapse
Affiliation(s)
- Elena Bellotti
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genova, Italy.
| | - Andrea L Schilling
- Department of Chemical Engineering, University of Pittsburgh, 427 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15261, USA
| | - Steven R Little
- Department of Chemical Engineering, University of Pittsburgh, 427 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15261, USA; Department of Bioengineering, University of Pittsburgh, 302 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15216, USA; Department of Clinical and Translational Science, University of Pittsburgh, Forbes tower, Suite 7057, Pittsburgh, PA 15213, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA; Department of Immunology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15213, USA; Department of Pharmaceutical Science, University of Pittsburgh, 3501 Terrace Street, Pittsburgh, PA, 15213, USA
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genova, Italy
| |
Collapse
|
25
|
Akhtar A, Andleeb A, Waris TS, Bazzar M, Moradi AR, Awan NR, Yar M. Neurodegenerative diseases and effective drug delivery: A review of challenges and novel therapeutics. J Control Release 2020; 330:1152-1167. [PMID: 33197487 DOI: 10.1016/j.jconrel.2020.11.021] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022]
Abstract
The central nervous system (CNS) encompasses the brain and spinal cord and is considered the processing center and the most vital part of human body. The central nervous system (CNS) barriers are crucial interfaces between the CNS and the periphery. Among all these biological barriers, the blood-brain barrier (BBB) strongly impede hurdle for drug transport to brain. It is a semi-permeable diffusion barrier against the noxious chemicals and harmful substances present in the blood stream and regulates the nutrients delivery to the brain for its proper functioning. Neurological diseases owing to the existence of the BBB and the blood-spinal cord barrier have been terrible and threatening challenges all over the world and can rarely be directly mediated. In fact, drug delivery to brain remained a challenge in the treatment of neurodegenerative (ND) disorders, for these different approaches have been proposed. Nano-fabricated smart drug delivery systems and implantable drug loaded biomaterials for brain repair are among some of these latest approaches. In current review, modern approaches developed to deal with the challenges associated with transporting drugs to the CNS are included. Recent studies on neural drug discovery and injectable hydrogels provide a potential new treatment option for neurological disorders. Moreover, induced pluripotent stem cells used to model ND diseases are discussed to evaluate drug efficacy. These protocols and recent developments will enable discovery of more effective drug delivery systems for brain.
Collapse
Affiliation(s)
- Amna Akhtar
- Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan; Department of Chemical Engineering, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Anisa Andleeb
- Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Tayyba Sher Waris
- Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Masoomeh Bazzar
- School of Chemistry, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Ali-Reza Moradi
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran; School of Nano Science, Institute for Research in Fundamental Sciences (IPM), P.O. Box 19395-5531, Tehran 19395, Iran
| | - Nasir Raza Awan
- Department of Neurosciences, Sharif Medical and Dental College, Lahore, Pakistan; Spinacure, 63-A Block E1, Gulberg III, Lahore, Pakistan
| | - Muhammad Yar
- Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan.
| |
Collapse
|
26
|
Sharma D, Cartar H, Law N, Giles A, Farhat G, Oelze M, Czarnota GJ. Optimization of microbubble enhancement of hyperthermia for cancer therapy in an in vivo breast tumour model. PLoS One 2020; 15:e0237372. [PMID: 32797049 PMCID: PMC7428078 DOI: 10.1371/journal.pone.0237372] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 07/24/2020] [Indexed: 11/18/2022] Open
Abstract
We have demonstrated that exposing human breast tumour xenografts to ultrasound-stimulated microbubbles enhances tumour cell death and vascular disruption resulting from hyperthermia treatment. The aim of this study was to investigate the effect of varying the hyperthermia and ultrasound-stimulated microbubbles treatment parameters in order to optimize treatment bioeffects. Human breast cancer (MDA-MB-231) tumour xenografts in severe combined immunodeficiency (SCID) mice were exposed to varying microbubble concentrations (0%, 0.1%, 1% or 3% v/v) and ultrasound sonication durations (0, 1, 3 or 5 min) at 570 kPa peak negative pressure and central frequency of 500 kHz. Five hours later, tumours were immersed in a 43°C water bath for varying hyperthermia treatment durations (0, 10, 20, 30, 40, 50 or 60 minutes). Results indicated a significant increase in tumour cell death reaching 64 ± 5% with combined treatment compared to 11 ± 3% and 26 ± 5% for untreated and USMB-only treated tumours, respectively. A similar but opposite trend was observed in the vascular density of the tumours receiving the combined treatment. Optimal treatment parameters were found to consist of 40 minutes of heat with low power ultrasound treatment microbubble parameters of 1 minute of sonification and a 1% microbubble concentration.
Collapse
Affiliation(s)
- Deepa Sharma
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Departments of Medical Biophysics, and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Holliday Cartar
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Niki Law
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Anoja Giles
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Golnaz Farhat
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Michael Oelze
- Department of Electrical and Computer Engineering, Beckman Institute, University of Chicago Illinois at Urbana Champaign, Illinois, United States of America
| | - Gregory J. Czarnota
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Departments of Medical Biophysics, and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
27
|
Le Floc'h J, Lu HD, Lim TL, Démoré C, Prud'homme RK, Hynynen K, Foster FS. Transcranial Photoacoustic Detection of Blood-Brain Barrier Disruption Following Focused Ultrasound-Mediated Nanoparticle Delivery. Mol Imaging Biol 2020; 22:324-334. [PMID: 31286352 PMCID: PMC7197023 DOI: 10.1007/s11307-019-01397-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE Blood-brain barrier disruption (BBBD) is of interest for treating neurodegenerative diseases and tumors by enhancing drug delivery. Focused ultrasound (FUS) is a powerful method to alleviate BBB challenges; however, the detection of BBB opening by non-invasive methods remains limited. The purpose of this work is to demonstrate that 3D transcranial color Doppler (3DCD) and photoacoustic imaging (PAI) combined with custom-made nanoparticle (NP)-mediated FUS delivery can detect BBBD in mice. PROCEDURES We use MRI and stereotactic ultrasound-mediated BBBD to create and confirm four openings in the left hemisphere and inject intravenously indocyanine green (ICG) and three sizes (40 nm, 100 nm, and 240 nm in diameter) of fluorophore-labeled NPs. We use PAI and fluorescent imaging (FI) to assess the spatial distribution of ICG/NPs in tissues. RESULTS A reversible 41 ± 12 % (n = 8) decrease in diameter of the left posterior cerebral artery (PCA) relative to the right after FUS treatment is found using CD images. The spectral unmixing of photoacoustic images of the in vivo (2 h post FUS), perfused, and ex vivo brain reveals a consistent distribution pattern of ICG and NPs at *FUS locations. Ex vivo spectrally unmixed photoacoustic images show that the opening width is, on average, 1.18 ± 0.12 mm and spread laterally 0.49 ± 0.05 mm which correlated well with the BBB opening locations on MR images. In vivo PAI confirms a deposit of NPs in tissues for hours and potentially days, is less sensitive to NPs of lower absorbance at a depth greater than 3 mm and too noisy with NPs above an absorbance of 85.4. FI correlates well with ex vivo PAI to a depth of 3 mm in tissues for small NPs and 4.74 mm for large NPs. CONCLUSIONS 3DCD can monitor BBBD over time by detecting reversible anatomical changes in the PCA. In vivo 3DPAI at 15 MHz combined with circulating ICG and/or NPs with suitable properties can assess BBB opening 2 h post FUS.
Collapse
Affiliation(s)
- Johann Le Floc'h
- Department of Medical Biophysics, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.
| | - Hoang D Lu
- Department of Chemical and Biological Engineering, Princeton University, 50-70 Olden St, Princeton, NJ, 08540, USA
| | - Tristan L Lim
- Department of Chemical and Biological Engineering, Princeton University, 50-70 Olden St, Princeton, NJ, 08540, USA
| | - Christine Démoré
- Department of Medical Biophysics, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Robert K Prud'homme
- Department of Chemical and Biological Engineering, Princeton University, 50-70 Olden St, Princeton, NJ, 08540, USA
| | - Kullervo Hynynen
- Department of Medical Biophysics, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - F Stuart Foster
- Department of Medical Biophysics, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| |
Collapse
|
28
|
Nozohouri S, Sifat AE, Vaidya B, Abbruscato TJ. Novel approaches for the delivery of therapeutics in ischemic stroke. Drug Discov Today 2020; 25:535-551. [PMID: 31978522 DOI: 10.1016/j.drudis.2020.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
Here, we review novel approaches to deliver neuroprotective drugs to salvageable penumbral brain areas of stroke injury with the goals of offsetting ischemic brain injury and enhancing recovery.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
29
|
Ji R, Smith M, Niimi Y, Karakatsani ME, Murillo MF, Jackson-Lewis V, Przedborski S, Konofagou EE. Focused ultrasound enhanced intranasal delivery of brain derived neurotrophic factor produces neurorestorative effects in a Parkinson's disease mouse model. Sci Rep 2019; 9:19402. [PMID: 31852909 PMCID: PMC6920380 DOI: 10.1038/s41598-019-55294-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/26/2019] [Indexed: 01/11/2023] Open
Abstract
Focused ultrasound-enhanced intranasal (IN + FUS) delivery is a noninvasive approach that utilizes the olfactory pathway to administer pharmacological agents directly to the brain, allowing for a more homogenous distribution in targeted locations compared to IN delivery alone. However, whether such a strategy has therapeutic values, especially in neurodegenerative disorders such as Parkinson’s disease (PD), remains to be established. Herein, we evaluated whether the expression of tyrosine hydroxylase (TH), the rate limiting enzyme in dopamine catalysis, could be enhanced by IN + FUS delivery of brain-derived neurotrophic factor (BDNF) in a toxin-based PD mouse model. Mice were put on the subacute dosing regimen of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), producing bilateral degeneration of the nigrostriatal pathway consistent with early-stage PD. MPTP mice then received BDNF intranasally followed by multiple unilateral FUS-induced blood-brain barrier (BBB) openings in the left basal ganglia for three consecutive weeks. Subsequently, mice were survived for two months and were evaluated morphologically and behaviorally to determine the integrity of their nigrostriatal dopaminergic pathways. Mice receiving IN + FUS had significantly increased TH immunoreactivity in the treated hemisphere compared to the untreated hemisphere while mice receiving only FUS-induced BBB opening or no treatment at all did not show any differences. Additionally, behavioral changes were only observed in the IN + FUS treated mice, indicating improved motor control function in the treated hemisphere. These findings demonstrate the robustness of the method and potential of IN + FUS for the delivery of bioactive factors for treatment of neurodegenerative disorder.
Collapse
Affiliation(s)
- Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Morgan Smith
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Yusuke Niimi
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Maria E Karakatsani
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Maria F Murillo
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Vernice Jackson-Lewis
- Department of Pathology & Cell Biology, Columbia University, New York, New York, USA.,Department of the Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of the Columbia Translational Neuroscience Initiative, Columbia University, New York, New York, USA
| | - Serge Przedborski
- Department of Pathology & Cell Biology, Columbia University, New York, New York, USA.,Department of Neurology, Columbia University, New York, New York, USA.,Department of the Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of the Columbia Translational Neuroscience Initiative, Columbia University, New York, New York, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, New York, USA. .,Department of Radiology, Columbia University, New York, New York, USA.
| |
Collapse
|
30
|
Karakatsani ME, Blesa J, Konofagou EE. Blood-brain barrier opening with focused ultrasound in experimental models of Parkinson's disease. Mov Disord 2019; 34:1252-1261. [PMID: 31361356 PMCID: PMC7213581 DOI: 10.1002/mds.27804] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease has many symptomatic treatments, but there is no neuroprotective therapy currently available. The evolution of this disease is inexorably progressive, and halting or stopping the neurodegenerative process is a major unmet need. Parkinson's disease motor features at onset are typically limited to 1 body segment, that is, focal signs, and the nigrostriatal degeneration is highly asymmetrical and mainly present in the caudal putamen. Thus, clinically and neurobiologically the process is fairly limited early in its evolution. Tentatively, this would allow the possibility of intervening to halt neurodegeneration at the most vulnerable site. The recent use of new technologies such as focused ultrasound provides interesting prospects. In particular, the possibility of transiently opening the blood-brain barrier to facilitate penetrance of putative neuroprotective agents is a highly attractive approach that could be readily applied to Parkinson's disease. However, because there are currently effective treatments available (ie, dopaminergic pharmacological therapy), more experimental evidence is needed to construct a feasible and practical therapeutic approach to be tested early in the evolution of Parkinson's disease patients. In this review, we provide the current evidence for the application of blood-brain barrier opening in experimental models of Parkinson's disease and discuss its potential clinical applicability. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Elisa Evgenia Konofagou
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
- Department of Radiology, Columbia University, New York, New York, USA
| |
Collapse
|
31
|
Nanoformulation properties, characterization, and behavior in complex biological matrices: Challenges and opportunities for brain-targeted drug delivery applications and enhanced translational potential. Adv Drug Deliv Rev 2019; 148:146-180. [PMID: 30797956 DOI: 10.1016/j.addr.2019.02.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/08/2019] [Accepted: 02/12/2019] [Indexed: 12/20/2022]
Abstract
Nanocarriers (synthetic/cell-based have attracted enormous interest for various therapeutic indications, including neurodegenerative disorders. A broader understanding of the impact of nanomedicines design is now required to enhance their translational potential. Nanoformulations in vivo journey is significantly affected by their physicochemical properties including the size, shape, hydrophobicity, elasticity, and surface charge/chemistry/morphology, which play a role as an interface with the biological environment. Understanding protein corona formation is crucial in characterizing nanocarriers and evaluating their interactions with biological systems. In this review, the types and properties of the brain-targeted nanocarriers are discussed. The biological factors and nanocarriers properties affecting their in vivo behavior are elaborated. The compositional description of cell culture and biological matrices, including proteins potentially relevant to protein corona built-up on nanoformulation especially for brain administration, is provided. Analytical techniques of characterizing nanocarriers in complex matrices, their advantages, limitations, and implementation challenges in industrial GMP environment are discussed. The uses of orthogonal complementary characterization approaches of nanocarriers are also covered.
Collapse
|
32
|
Karakatsani ME, Kugelman T, Ji R, Murillo M, Wang S, Niimi Y, Small SA, Duff KE, Konofagou EE. Unilateral Focused Ultrasound-Induced Blood-Brain Barrier Opening Reduces Phosphorylated Tau from The rTg4510 Mouse Model. Am J Cancer Res 2019; 9:5396-5411. [PMID: 31410223 PMCID: PMC6691580 DOI: 10.7150/thno.28717] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 06/21/2019] [Indexed: 11/21/2022] Open
Abstract
The neuropathological hallmarks of Alzheimer's disease include amyloid plaques and neurofibrillary tangles. Tau pathology correlates well with impaired neuronal activity and dementia. Focused ultrasound coupled with systemic administration of microbubbles has previously been shown to open the blood-brain barrier and induce an immune response, which, in an amyloid AD mouse model, resulted in the reduction of the amyloid brain load. Methods: In this study, we investigated the effect of focused ultrasound at the early stages of tau pathology (pre-tangle) in the rTg4510 mouse model. Results: Reduction of phosphorylated tau from the hippocampal formation processes, and particularly the pyramidal CA1 neurons, was noted in the ultrasound-treated brains without an associated increase in the phosphorylated tau-affected cell somas, typically associated with disease progression. Attenuation of the pathology was found to correlate well with the ultrasound-initiated immune response without compromising neuronal integrity. Unilateral ultrasound application resulted in a bilateral effect indicating a broader reduction of the phosphorylated tau. Conclusion: Findings presented herein reinforce the premise of ultrasound in reducing tau pathology and thus curbing the progression of Alzheimer's disease.
Collapse
|
33
|
Karakatsani ME, Wang S, Samiotaki G, Kugelman T, Olumolade OO, Acosta C, Sun T, Han Y, Kamimura HAS, Jackson-Lewis V, Przedborski S, Konofagou E. Amelioration of the nigrostriatal pathway facilitated by ultrasound-mediated neurotrophic delivery in early Parkinson's disease. J Control Release 2019; 303:289-301. [PMID: 30953664 PMCID: PMC6618306 DOI: 10.1016/j.jconrel.2019.03.030] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/30/2019] [Accepted: 03/29/2019] [Indexed: 01/01/2023]
Abstract
The blood-brain barrier (BBB) prevents most drugs from gaining access to the brain parenchyma, which is a recognized impediment to the treatment of neurodegenerative disorders like Parkinson's disease (PD). Focused ultrasound (FUS), in conjunction with systemically administered microbubbles, opens the BBB locally, reversibly and non-invasively. Herein, we show that neither FUS applied over both the striatum and the ventral midbrain, without neurotrophic factors, nor intravenous administration of neurotrophic factors (either through protein or gene delivery) without FUS, ameliorates the damage to the nigrostriatal dopaminergic pathway in the sub-acute MPTP mouse model of early-stage PD. Conversely, the combination of FUS and intravenous neurotrophic (protein or gene) delivery attenuates the damage to the nigrostriatal dopaminergic pathway, by allowing the entry of these agents into the brain parenchyma. Our findings provide evidence that the application of FUS at the early stages of PD facilitates critical neurotrophic delivery that can curb the rapid progression of neurodegeneration while improving the neuronal function, seemingly opening new therapeutic avenues for the early treatment of diseases of the central nervous system.
Collapse
Affiliation(s)
| | - Shutao Wang
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Gesthimani Samiotaki
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Tara Kugelman
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Oluyemi O Olumolade
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Camilo Acosta
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Tao Sun
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Yang Han
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Hermes A S Kamimura
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Vernice Jackson-Lewis
- Departments of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA; Departments of Neurology, Columbia University, New York, NY 10032, USA; the Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; the Columbia Translational Neuroscience Initiative, Columbia University, New York, NY 10032, USA
| | - Serge Przedborski
- Departments of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA; Departments of Neurology, Columbia University, New York, NY 10032, USA; the Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; the Columbia Translational Neuroscience Initiative, Columbia University, New York, NY 10032, USA.
| | - Elisa Konofagou
- Departments of Biomedical Engineering, Columbia University, New York, NY 10032, USA; Departments of Radiology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
34
|
Kunze R, Marti HH. Angioneurins - Key regulators of blood-brain barrier integrity during hypoxic and ischemic brain injury. Prog Neurobiol 2019; 178:101611. [PMID: 30970273 DOI: 10.1016/j.pneurobio.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
The loss of blood-brain barrier (BBB) integrity leading to vasogenic edema and brain swelling is a common feature of hypoxic/ischemic brain diseases such as stroke, but is also central to the etiology of other CNS disorders. In the past decades, numerous proteins, belonging to the family of angioneurins, have gained increasing attention as potential therapeutic targets for ischemic stroke, but also other CNS diseases attributed to BBB dysfunction. Angioneurins encompass mediators that affect both neuronal and vascular function. Recently, increasing evidence has been accumulated that certain angioneurins critically determine disease progression and outcome in stroke among others through multifaceted effects on the compromised BBB. Here, we will give a concise overview about the family of angioneurins. We further describe the most important cellular and molecular components that contribute to structural integrity and low permeability of the BBB under steady-state conditions. We then discuss BBB alterations in ischemic stroke, and highlight underlying cellular and molecular mechanisms. For the most prominent angioneurin family members including vascular endothelial growth factors, angiopoietins, platelet-derived growth factors and erythropoietin, we will summarize current scientific literature from experimental studies in animal models, and if available from clinical trials, on the following points: (i) spatiotemporal expression of these factors in the healthy and hypoxic/ischemic CNS, (ii) impact of loss- or gain-of-function during cerebral hypoxia/ischemia for BBB integrity and beyond, and (iii) potential underlying molecular mechanisms. Moreover, we will highlight novel therapeutic strategies based on the activation of endogenous angioneurins that might improve BBB dysfuntion during ischemic stroke.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany.
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany
| |
Collapse
|
35
|
Bruggeman KF, Moriarty N, Dowd E, Nisbet DR, Parish CL. Harnessing stem cells and biomaterials to promote neural repair. Br J Pharmacol 2019; 176:355-368. [PMID: 30444942 PMCID: PMC6329623 DOI: 10.1111/bph.14545] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 10/16/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023] Open
Abstract
With the limited capacity for self-repair in the adult CNS, efforts to stimulate quiescent stem cell populations within discrete brain regions, as well as harness the potential of stem cell transplants, offer significant hope for neural repair. These new cells are capable of providing trophic cues to support residual host populations and/or replace those cells lost to the primary insult. However, issues with low-level adult neurogenesis, cell survival, directed differentiation and inadequate reinnervation of host tissue have impeded the full potential of these therapeutic approaches and their clinical advancement. Biomaterials offer novel approaches to stimulate endogenous neurogenesis, as well as for the delivery and support of neural progenitor transplants, providing a tissue-appropriate physical and trophic milieu for the newly integrating cells. In this review, we will discuss the various approaches by which bioengineered scaffolds may improve stem cell-based therapies for repair of the CNS.
Collapse
Affiliation(s)
- K F Bruggeman
- Laboratory of Advanced Biomaterials, Research School of EngineeringThe Australian National UniversityCanberraACTAustralia
| | - N Moriarty
- Pharmacology and Therapeutics and Galway Neuroscience CentreNational University of Ireland GalwayGalwayIreland
| | - E Dowd
- Pharmacology and Therapeutics and Galway Neuroscience CentreNational University of Ireland GalwayGalwayIreland
| | - D R Nisbet
- Laboratory of Advanced Biomaterials, Research School of EngineeringThe Australian National UniversityCanberraACTAustralia
| | - C L Parish
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVICAustralia
| |
Collapse
|
36
|
Brighi C, Puttick S, Rose S, Whittaker AK. The potential for remodelling the tumour vasculature in glioblastoma. Adv Drug Deliv Rev 2018; 136-137:49-61. [PMID: 30308226 DOI: 10.1016/j.addr.2018.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 12/19/2022]
Abstract
Despite significant improvements in the clinical management of glioblastoma, poor delivery of systemic therapies to the entire population of tumour cells remains one of the biggest challenges in the achievement of more effective treatments. On the one hand, the abnormal and dysfunctional tumour vascular network largely limits blood perfusion, resulting in an inhomogeneous delivery of drugs to the tumour. On the other hand, the presence of an intact blood-brain barrier (BBB) in certain regions of the tumour prevents chemotherapeutic drugs from permeating through the tumour vessels and reaching the diseased cells. In this review we analyse in detail the implications of the presence of a dysfunctional vascular network and the impenetrable BBB on drug transport. We discuss advantages and limitations of the currently available strategies for remodelling the tumour vasculature aiming to ameliorate the above mentioned limitations. Finally we review research methods for visualising vascular dysfunction and highlight the power of DCE- and DSC-MRI imaging to assess changes in blood perfusion and BBB permeability.
Collapse
|
37
|
Ganipineni LP, Danhier F, Préat V. Drug delivery challenges and future of chemotherapeutic nanomedicine for glioblastoma treatment. J Control Release 2018; 281:42-57. [PMID: 29753958 DOI: 10.1016/j.jconrel.2018.05.008] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/20/2022]
Abstract
Glioblastoma (GBM) is one of the most aggressive and deadliest central nervous system tumors, and the current standard treatment is surgery followed by radiotherapy with concurrent chemotherapy. Nevertheless, the survival period is notably low. Although ample research has been performed to develop an effective therapeutic strategy for treating GBM, the success of extending patients' survival period and quality of life is limited. This review focuses on the strategies developed to address the challenges associated with drug delivery in GBM, particularly nanomedicine. The first part describes major obstacles to the development of effective GBM treatment strategies. The second part focuses on the conventional chemotherapeutic nanomedicine strategies, their limitations and the novel and advanced strategies of nanomedicine, which could be promising for GBM treatment. We also highlighted the prominence of nanomedicine clinical translation. The near future looks bright following the beginning of clinical translation of nanochemotherapy for GBM.
Collapse
Affiliation(s)
- Lakshmi Pallavi Ganipineni
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73 bte B1 73.12, 1200 Brussels, Belgium
| | - Fabienne Danhier
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73 bte B1 73.12, 1200 Brussels, Belgium
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73 bte B1 73.12, 1200 Brussels, Belgium.
| |
Collapse
|
38
|
Enhancement of brain-targeting delivery of danshensu in rat through conjugation with pyrazine moiety to form danshensu-pyrazine ester. Drug Deliv Transl Res 2018. [PMID: 29524164 DOI: 10.1007/s13346-018-0501-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Tetramethylpyrazine was introduced to the structure of danshensu (DSS) as P-glycoprotein (P-gp)-inhibiting carrier, designing some novel brain-targeting DSS-pyrazine derivatives via prodrug delivery strategy. Following the virtual screening, three DSS-pyrazine esters (DT1, DT2, DT3) were selected because of their better prediction parameters related to brain-targeting. Among them, DT3 was thought to be a promising candidate due to its appropriate bioreversible property in vitro release assay. Further investigation with regard to DT3's brain-targeting effects in vivo was also reported in this study. High-performance liquid chromatography-diode array detection (HPLC-DAD) method was established for the quantitative determination of DT3 and DSS in rat plasma, brain homogenate after intravenous injection. In vivo metabolism of DT3 indicated that it was first converted into DT1, DT2, then the generation of DSS, which could be the result of carboxylesterase activity in rat blood and brain tissue. Moreover, the brain pharmacokinetics of DT3 was significantly altered with 2.16 times increase in half-life compared with that of DSS, and its drug targeting index (DTI) was up to 16.95. Above these data demonstrated that DT3 had better tendency of brain-targeting delivery, which would be positive for the treatment of brain-related disorders.
Collapse
|
39
|
Bruggeman KF, Williams RJ, Nisbet DR. Dynamic and Responsive Growth Factor Delivery from Electrospun and Hydrogel Tissue Engineering Materials. Adv Healthc Mater 2018; 7. [PMID: 29193871 DOI: 10.1002/adhm.201700836] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/04/2017] [Indexed: 12/21/2022]
Abstract
Tissue engineering scaffolds are designed to mimic physical, chemical, and biological features of the extracellular matrix, thereby providing a constant support that is crucial to improved regenerative medicine outcomes. Beyond mechanical and structural support, the next generation of these materials must also consider the more dynamic presentation and delivery of drugs or growth factors to guide new and regenerating tissue development. These two aspects are explored expansively separately, but they must interact synergistically to achieve optimal regeneration. This review explores common tissue engineering materials types, electrospun polymers and hydrogels, and strategies used for incorporating drug delivery systems into these scaffolds.
Collapse
Affiliation(s)
- Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials; Research School of Engineering; The Australian National University; Canberra ACT 2601 Australia
| | - Richard J. Williams
- School of Engineering; RMIT University; Melbourne VIC 3001 Australia
- Biofab3D; Aikenhead Center for Medical Discovery; St. Vincent's Hospital; Melbourne VIC 3065 Australia
| | - David R. Nisbet
- Laboratory of Advanced Biomaterials; Research School of Engineering; The Australian National University; Canberra ACT 2601 Australia
- Biofab3D; Aikenhead Center for Medical Discovery; St. Vincent's Hospital; Melbourne VIC 3065 Australia
| |
Collapse
|
40
|
Fontes MAP, Vaz GC, Cardoso TZD, de Oliveira MF, Campagnole-Santos MJ, Dos Santos RAS, Sharma NM, Patel KP, Frézard F. GABA-containing liposomes: neuroscience applications and translational perspectives for targeting neurological diseases. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:781-788. [PMID: 29278747 DOI: 10.1016/j.nano.2017.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/27/2017] [Accepted: 12/10/2017] [Indexed: 11/28/2022]
Abstract
There are multiple challenges for neuropharmacology in the future. Undoubtedly, one of the greatest challenges is the development of strategies for pharmacological targeting of specific brain regions for treatment of diseases. GABA is the main inhibitory neurotransmitter in the central nervous system, and dysfunction of GABAergic mechanisms is associated with different neurological conditions. Liposomes are lipid vesicles that are able to encapsulate chemical compounds and are used for chronic drug delivery. This short review reports our experience with the development of liposomes for encapsulation and chronic delivery of GABA to sites within the brain. Directions for future research regarding the efficacy and practical use of GABA-containing liposomes for extended periods of time as well as understanding and targeting neurological conditions are discussed.
Collapse
Affiliation(s)
| | - Gisele Cristiane Vaz
- Department of Physiology & Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | - Neeru M Sharma
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Frédéric Frézard
- Department of Physiology & Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
41
|
Zhao Y, Zhang L, Peng Y, Yue Q, Hai L, Guo L, Wang Q, Wu Y. GLUT 1 -mediated venlafaxine-thiamine disulfide system-glucose conjugates with "lock-in" function for central nervous system delivery. Chem Biol Drug Des 2017; 91:707-716. [PMID: 29063718 DOI: 10.1111/cbdd.13128] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/15/2017] [Accepted: 10/02/2017] [Indexed: 01/11/2023]
Abstract
Venlafaxine, a novel third-generation antidepressant drug, has been described as a reference treatment for major depression, owing to its ability of inhibiting both noradrenalin and serotonin neuronal reuptake, and inhibiting dopamine reuptake slightly. However, its clinical application is hampered by a limited brain distribution. Glucosylation is an effective way to enhance the brain targeting ability of drugs, but the bidirectional transport of glucose transporter 1 (GLUT1 ) might decrease the concentrations of venlafaxine-glucose (V-G) in brain before the release of parent drug venlafaxine. To conquer this drawback of GLUT1 , "lock-in" thiamine disulfide system (TDS) was introduced to modify the V-G conjugate. Both conjugates could release venlafaxine when incubated with the various buffers, mice plasma, and brain homogenate. The evaluation in vivo demonstrated that venlafaxine-TDS-glucose (V-TDS-G) had an improved targeting ability and significantly increased the level of venlafaxine in brain compared to the naked venlafaxine and V-G. The relative uptake efficiency (RE) and concentration efficiency (CE) were enhanced to 5.69 and 5.70 times higher than that of naked venlafaxine, respectively. The results of this study suggest that the conjugate strategy based on the glucose-TDS (G-TDS) is available to enhance the delivery of central nervous system (CNS) drugs into brain.
Collapse
Affiliation(s)
- Yi Zhao
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Li Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yao Peng
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qiming Yue
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Li Hai
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Li Guo
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qiantao Wang
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yong Wu
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
42
|
Lafuente JV, Requejo C, Carrasco A, Bengoetxea H. Nanoformulation: A Useful Therapeutic Strategy for Improving Neuroprotection and the Neurorestorative Potential in Experimental Models of Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 137:99-122. [PMID: 29132545 DOI: 10.1016/bs.irn.2017.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disorder, but current therapies are only symptomatic. Experimental models are necessary to go deeper in the comprehension of pathophysiological mechanism and to assess new therapeutic strategies. The unilateral 6-hydroxydopamine (6-OHDA) lesion either in medial forebrain bundle (MFB) or into the striatum in rats affords to study various stages of PD depending on the evolution time lapsed. A promising alternative to address the neurodegenerative process is the use of neurotrophic factors; but its clinical use has been limited due to its short half-life and rapid degradation after in vivo administration, along with difficulties for crossing the blood-brain barrier (BBB). Tyrosine hydroxylase (TH) immunostaining revealed a significant decrease of the TH-immunopositive striatal volume in 6-OHDA group from rostral to caudal one. The loss of TH-ir neurons and axodendritic network (ADN) was higher in caudal sections showing a selective vulnerability of the topological distributed dopaminergic system. In addition to a remarkable depletion of dopamine in the nigrostriatal system, the administration of 6-OHDA into MFB induces some other neuropathological changes such as an increase of glial fibrillary acidic protein (GFAP) positive cells in substantia nigra (SN) as well as in striatum. Intrastriatal implantation of micro- or nanosystems delivering neurotrophic factor in parkinsonized rats for bypassing BBB leads to a significative functional and morphological recovery. Neurorestorative morphological changes (preservation of the TH-ir cells and ADN) along the rostrocaudal axis of caudoputamen complex and SN have been probed supporting a selective recovering after the treatment as well. Others innovative therapeutic strategies, such as the intranasal delivery, have been recently assessed in order to search the NTF effects. In addition some others methodological key points are reviewed.
Collapse
Affiliation(s)
- Jose V Lafuente
- Laboratory of Clinical and Experimental Neuroscience (LaNCE), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain.
| | - Catalina Requejo
- Laboratory of Clinical and Experimental Neuroscience (LaNCE), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain
| | - Alejandro Carrasco
- Group Nanoneurosurgery, Institute of Health Research Biocruces, Barakaldo, Spain; Service Neurosurgery, Cruces University Hospital, Barakaldo, Spain
| | - Harkaitz Bengoetxea
- Laboratory of Clinical and Experimental Neuroscience (LaNCE), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain
| |
Collapse
|
43
|
Bruggeman KF, Wang Y, Maclean FL, Parish CL, Williams RJ, Nisbet DR. Temporally controlled growth factor delivery from a self-assembling peptide hydrogel and electrospun nanofibre composite scaffold. NANOSCALE 2017; 9:13661-13669. [PMID: 28876347 DOI: 10.1039/c7nr05004f] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Tissue-specific self-assembling peptide (SAP) hydrogels designed based on biologically relevant peptide sequences have great potential in regenerative medicine. These materials spontaneously form 3D networks of physically assembled nanofibres utilising non-covalent interactions. The nanofibrous structure of SAPs is often compared to that of electrospun scaffolds. These electrospun nanofibers are produced as sheets that can be engineered from a variety of polymers that can be chemically modified to incorporate many molecules including drugs and growth factors. However, their macroscale morphology limits them to wrapping and bandaging applications. Here, for the first time, we combine the benefits of these systems to describe a two-component composite scaffold from these biomaterials, with the design goal of providing a hydrogel scaffold that presents 3D structures, and also has temporal control over drug delivery. Short fibres, cut from electrospun scaffolds, were mixed with our tissue-specific SAP hydrogel to provide a range of nanofibre sizes found in the extracellular matrix (10-300 nm in diameter). The composite material maintained the shear-thinning and void-filling properties of SAP hydrogels that have previously been shown to be effective for minimally invasive material injection, cell delivery and subsequent in vivo integration. Both scaffold components were separately loaded with growth factors, important signaling molecules in tissue regeneration whose rapid degradation limits their clinical efficacy. The two biomaterials provided sequential growth factor delivery profiles: the SAP hydrogel provided a burst release, with the release rate decreasing over 12 hours, while the electrospun nanofibres provided a more constant, sustained delivery. Importantly, this second release commenced 6 days later. The design rules established here to provide temporally distinct release profiles can enable researchers to target specific stages in regeneration, such as the acute immune response versus sustained protection and survival of cells following injury. In summary, this novel composite material combines the physical advantages of SAP hydrogels and electrospun nanofibres, while additionally providing a superior vehicle for the stabilisation and controlled delivery of growth factors necessary for optimal tissue repair.
Collapse
Affiliation(s)
- Kiara F Bruggeman
- Laboratory of Advanced Biomaterials, Research School of Engineering, The Australian National University, Canberra, ACT 2601, Australia.
| | | | | | | | | | | |
Collapse
|
44
|
Zhao MX, Zhu BJ, Yao WJ, Chen DF, Wang C. The delivery of doxorubicin of multifunctional β-cyclodextrin-modified CdSe/ZnS quantum dots for bioactivity and nano-probing. Chem Biol Drug Des 2017; 91:285-293. [PMID: 28791767 DOI: 10.1111/cbdd.13080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/30/2017] [Accepted: 07/27/2017] [Indexed: 01/04/2023]
Abstract
The modified quantum dots (QDs) have been used in intracellular probing and drug delivery because of their special chemical and physical properties. In this paper, two β-cyclodextrin (β-CD)-modified CdSe/ZnS QDs with strong optical emission properties were synthesized as drug carriers to induce apoptosis. The positively charged l-Arginine (l-Arg) and neutral l-Tryptophan (l-Trp) were selected as ligands to compare the effect of charge on bioactivity of QDs nanoparticles. The in vitro assays revealed that these modified QDs showed good Dox carrier ability and significantly high inhibition rate to cancer cells. Especially, the more positively charged β-CD-l-Arg-polyamine-coated CdSe/ZnS QDs could effectively deliver the doxorubicin (Dox) into cells and exhibit excellent cell selectivity in cancer versus normal cells. The Dox-loaded QDs could enter intracellular, which showed that the Dox can efficiently go through the membranes at the existence of β-CD. Several lines of evidence suggest that the Dox-loaded QDs can efficiently induce apoptosis likely related to the production of ROS. We expect that the modified QDs can enhance the amount of hydrophobic antitumor drugs in cells and can also be used as fluorescent imaging agents.
Collapse
Affiliation(s)
- Mei-Xia Zhao
- Key Laboratory of Natural Medicine and Immune Engineering of Henan Province, Henan University, Kaifeng, China
| | - Bing-Jie Zhu
- Key Laboratory of Natural Medicine and Immune Engineering of Henan Province, Henan University, Kaifeng, China
| | - Wen-Jing Yao
- Key Laboratory of Natural Medicine and Immune Engineering of Henan Province, Henan University, Kaifeng, China
| | - Di-Feng Chen
- Key Laboratory of Natural Medicine and Immune Engineering of Henan Province, Henan University, Kaifeng, China
| | - Chaojie Wang
- Key Laboratory of Natural Medicine and Immune Engineering of Henan Province, Henan University, Kaifeng, China
| |
Collapse
|
45
|
Sakamoto K, Shinohara T, Adachi Y, Asami T, Ohtaki T. A novel LRP1-binding peptide L57 that crosses the blood brain barrier. Biochem Biophys Rep 2017; 12:135-139. [PMID: 29090274 PMCID: PMC5645116 DOI: 10.1016/j.bbrep.2017.07.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/26/2017] [Accepted: 07/03/2017] [Indexed: 12/22/2022] Open
Abstract
The blood-brain barrier (BBB) is a major obstacle to drug delivery into the central nervous system (CNS), in particular for macromolecules such as peptides and proteins. However, certain macromolecules can reach the CNS via a receptor-mediated transcytosis (RMT) pathway, and low-density lipoprotein receptor-related protein 1 (LRP1) is one of the promising receptors for RMT. An LRP1 ligand peptide, Angiopep-2, was reported to pass through the BBB and deliver covalently conjugated drugs into the CNS. While conjugation of LRP1 ligands with drugs would be an effective approach for drug delivery to the CNS, no other reliable LRP1 ligands have been reported to date. In this study, we aimed to identify novel LRP1 ligands to further investigate LRP1-mediated RMT. Using phage display technology, we obtained a novel peptide, L57 (TWPKHFDKHTFYSILKLGKH-OH), with an EC50 value of 45 nM for binding to cluster 4 (Ser3332–Asp3779) of LRP1. L57 was stable in mouse plasma for up to 20 min. In situ brain perfusion assay in mice revealed the significantly high BBB permeability of L57. In conclusion, we discovered L57, the first artificial LRP1-binding peptide with BBB permeability. Our findings will contribute to the development of RMT-based drugs for the treatment of CNS diseases. The first artificial LRP1-binding peptide L57 was discovered by phage display. L57 binds the extracellular domain of LRP1 with an EC50 binding value of 24 nM. L57 exhibits brain uptake in in situ brain perfusion and i.v. injection in mice.
Collapse
Affiliation(s)
- Kotaro Sakamoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., Fujisawa 251-8555, Japan
| | - Tokuyuki Shinohara
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., Fujisawa 251-8555, Japan
| | - Yusuke Adachi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., Fujisawa 251-8555, Japan
| | - Taiji Asami
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., Fujisawa 251-8555, Japan
| | - Tetsuya Ohtaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Ltd., Fujisawa 251-8555, Japan
| |
Collapse
|
46
|
Huang L, Hu J, Huang S, Wang B, Siaw-Debrah F, Nyanzu M, Zhang Y, Zhuge Q. Nanomaterial applications for neurological diseases and central nervous system injury. Prog Neurobiol 2017; 157:29-48. [PMID: 28743465 DOI: 10.1016/j.pneurobio.2017.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022]
Abstract
The effectiveness of noninvasive treatment for neurological disease is generally limited by the poor entry of therapeutic agents into the central nervous system (CNS). Most CNS drugs cannot permeate into the brain parenchyma because of the blood-brain barrier thus, overcoming this problem has become one of the most significant challenges in the development of neurological therapeutics. Nanotechnology has emerged as an innovative alternative for treating neurological diseases. In fact, rapid advances in nanotechnology have provided promising solutions to this challenge. This review highlights the applications of nanomaterials in the developing neurological field and discusses the evidence for their efficacies.
Collapse
Affiliation(s)
- Lijie Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Jiangnan Hu
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Shengwei Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Brian Wang
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Felix Siaw-Debrah
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Mark Nyanzu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Yu Zhang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Qichuan Zhuge
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China.
| |
Collapse
|
47
|
Katare YK, Piazza JE, Bhandari J, Daya RP, Akilan K, Simpson MJ, Hoare T, Mishra RK. Intranasal delivery of antipsychotic drugs. Schizophr Res 2017; 184:2-13. [PMID: 27913162 DOI: 10.1016/j.schres.2016.11.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 12/31/2022]
Abstract
Antipsychotic drugs are used to treat psychotic disorders that afflict millions globally and cause tremendous emotional, economic and healthcare burdens. However, the potential of intranasal delivery to improve brain-specific targeting remains unrealized. In this article, we review the mechanisms and methods used for brain targeting via the intranasal (IN) route as well as the potential advantages of improving this type of delivery. We extensively review experimental studies relevant to intranasal delivery of therapeutic agents for the treatment of psychosis and mental illnesses. We also review clinical studies in which intranasal delivery of peptides, like oxytocin (7 studies) and desmopressin (1), were used as an adjuvant to antipsychotic treatment with promising results. Experimental animal studies (17) investigating intranasal delivery of mainstream antipsychotic drugs have revealed successful targeting to the brain as suggested by pharmacokinetic parameters and behavioral effects. To improve delivery to the brain, nanotechnology-based carriers like nanoparticles and nanoemulsions have been used in several studies. However, human studies assessing intranasal delivery of mainstream antipsychotic drugs are lacking, and the potential toxicity of nanoformulations used in animal studies has not been explored. A brief discussion of future directions anticipates that if limitations of low aqueous solubility of antipsychotic drugs can be overcome and non-toxic formulations used, IN delivery (particularly targeting specific tissues within the brain) will gain more importance moving forward given the inherent benefits of IN delivery in comparison to other methods.
Collapse
Affiliation(s)
- Yogesh K Katare
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Justin E Piazza
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Jayant Bhandari
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Ritesh P Daya
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Kosalan Akilan
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Madeline J Simpson
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Ram K Mishra
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
48
|
Escalona-Rayo O, Fuentes-Vázquez P, Leyva-Gómez G, Cisneros B, Villalobos R, Magaña JJ, Quintanar-Guerrero D. Nanoparticulate strategies for the treatment of polyglutamine diseases by halting the protein aggregation process. Drug Dev Ind Pharm 2017; 43:871-888. [PMID: 28142290 DOI: 10.1080/03639045.2017.1281949] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Polyglutamine (polyQ) diseases are a class of neurodegenerative disorders that cause cellular dysfunction and, eventually, neuronal death in specific regions of the brain. Neurodegeneration is linked to the misfolding and aggregation of expanded polyQ-containing proteins, and their inhibition is one of major therapeutic strategies used commonly. However, successful treatment has been limited to date because of the intrinsic properties of therapeutic agents (poor water solubility, low bioavailability, poor pharmacokinetic properties), and difficulty in crossing physiological barriers, including the blood-brain barrier (BBB). In order to solve these problems, nanoparticulate systems with dimensions of 1-1000 nm able to incorporate small and macromolecules with therapeutic value, to protect and deliver them directly to the brain, have recently been developed, but their use for targeting polyQ disease-mediated protein misfolding and aggregation remains scarce. This review provides an update of the polyQ protein aggregation process and the development of therapeutic strategies for halting it. The main features that a nanoparticulate system should possess in order to enhance brain delivery are discussed, as well as the different types of materials utilized to produce them. The final part of this review focuses on the potential application of nanoparticulate system strategies to improve the specific and efficient delivery of therapeutic agents to the brain for the treatment of polyQ diseases.
Collapse
Affiliation(s)
- Oscar Escalona-Rayo
- a Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, Facultad de Estudios Superiores Cuautitlán , Universidad Nacional Autónoma de México (UNAM) , Cuautitlán Izcalli , Mexico
| | - Paulina Fuentes-Vázquez
- a Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, Facultad de Estudios Superiores Cuautitlán , Universidad Nacional Autónoma de México (UNAM) , Cuautitlán Izcalli , Mexico
| | - Gerardo Leyva-Gómez
- b Laboratory of Connective Tissue , CENIAQ, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra , Mexico City , Mexico
| | - Bulmaro Cisneros
- c Department of Genetics and Molecular Biology , CINVESTAV-IPN , Mexico City , Mexico
| | - Rafael Villalobos
- d División de Estudios de Posgrado (Tecnología Farmacéutica), Facultad de Estudios Superiores Cuautitlán , Universidad Nacional Autónoma de México (UNAM) , Cuautitlán Izcalli , Mexico
| | - Jonathan J Magaña
- e Laboratory of Genomic Medicine, Department of Genetics , Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra , Mexico City , Mexico
| | - David Quintanar-Guerrero
- a Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, Facultad de Estudios Superiores Cuautitlán , Universidad Nacional Autónoma de México (UNAM) , Cuautitlán Izcalli , Mexico
| |
Collapse
|
49
|
Tam VH, Sosa C, Liu R, Yao N, Priestley RD. Nanomedicine as a non-invasive strategy for drug delivery across the blood brain barrier. Int J Pharm 2016; 515:331-342. [DOI: 10.1016/j.ijpharm.2016.10.031] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/02/2016] [Accepted: 10/15/2016] [Indexed: 12/13/2022]
|
50
|
Bleier BS, Kohman RE, Guerra K, Nocera AL, Ramanlal S, Kocharyan AH, Curry WT, Han X. Heterotopic Mucosal Grafting Enables the Delivery of Therapeutic Neuropeptides Across the Blood Brain Barrier. Neurosurgery 2016; 78:448-57; discussion 457. [PMID: 26352099 DOI: 10.1227/neu.0000000000001016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The blood-brain barrier represents a fundamental limitation in treating neurological disease because it prevents all neuropeptides from reaching the central nervous system (CNS). Currently, there is no efficient method to permanently bypass the blood-brain barrier. OBJECTIVE To test the feasibility of using nasal mucosal graft reconstruction of arachnoid defects to deliver glial-derived neurotrophic factor (GDNF) for the treatment of Parkinson disease in a mouse model. METHODS The Institutional Animal Care and Use Committee approved this study in an established murine 6-hydroxydopamine Parkinson disease model. A parietal craniotomy and arachnoid defect was repaired with a heterotopic donor mucosal graft. The therapeutic efficacy of GDNF (2 μg/mL) delivered through the mucosal graft was compared with direct intrastriatal GDNF injection (2 μg/mL) and saline control through the use of 2 behavioral assays (rotarod and apomorphine rotation). An immunohistological analysis was further used to compare the relative preservation of substantia nigra cell bodies between treatment groups. RESULTS Transmucosal GDNF was equivalent to direct intrastriatal injection at preserving motor function at week 7 in both the rotarod and apomorphine rotation behavioral assays. Similarly, both transmucosal and intrastriatal GDNF demonstrated an equivalent ratio of preserved substantia nigra cell bodies (0.79 ± 0.14 and 0.78 ± 0.09, respectively, P = NS) compared with the contralateral control side, and both were significantly greater than saline control (0.53 ± 0.21; P = .01 and P = .03, respectively). CONCLUSION Transmucosal delivery of GDNF is equivalent to direct intrastriatal injection at ameliorating the behavioral and immunohistological features of Parkinson disease in a murine model. Mucosal grafting of arachnoid defects is a technique commonly used for endoscopic skull base reconstruction and may represent a novel method to permanently bypass the blood-brain barrier.
Collapse
Affiliation(s)
- Benjamin S Bleier
- ‡Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts; §Department of Biomedical Engineering, Boston University, Boston, Massachusetts; ¶Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | |
Collapse
|