51
|
Bernardo-Castro S, Sousa JA, Brás A, Cecília C, Rodrigues B, Almendra L, Machado C, Santo G, Silva F, Ferreira L, Santana I, Sargento-Freitas J. Pathophysiology of Blood-Brain Barrier Permeability Throughout the Different Stages of Ischemic Stroke and Its Implication on Hemorrhagic Transformation and Recovery. Front Neurol 2020; 11:594672. [PMID: 33362697 PMCID: PMC7756029 DOI: 10.3389/fneur.2020.594672] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/09/2020] [Indexed: 12/25/2022] Open
Abstract
The blood-brain barrier (BBB) is a dynamic interface responsible for maintaining the central nervous system homeostasis. Its unique characteristics allow protecting the brain from unwanted compounds, but its impairment is involved in a vast number of pathological conditions. Disruption of the BBB and increase in its permeability are key in the development of several neurological diseases and have been extensively studied in stroke. Ischemic stroke is the most prevalent type of stroke and is characterized by a myriad of pathological events triggered by an arterial occlusion that can eventually lead to fatal outcomes such as hemorrhagic transformation (HT). BBB permeability seems to follow a multiphasic pattern throughout the different stroke stages that have been associated with distinct biological substrates. In the hyperacute stage, sudden hypoxia damages the BBB, leading to cytotoxic edema and increased permeability; in the acute stage, the neuroinflammatory response aggravates the BBB injury, leading to higher permeability and a consequent risk of HT that can be motivated by reperfusion therapy; in the subacute stage (1-3 weeks), repair mechanisms take place, especially neoangiogenesis. Immature vessels show leaky BBB, but this permeability has been associated with improved clinical recovery. In the chronic stage (>6 weeks), an increase of BBB restoration factors leads the barrier to start decreasing its permeability. Nonetheless, permeability will persist to some degree several weeks after injury. Understanding the mechanisms behind BBB dysregulation and HT pathophysiology could potentially help guide acute stroke care decisions and the development of new therapeutic targets; however, effective translation into clinical practice is still lacking. In this review, we will address the different pathological and physiological repair mechanisms involved in BBB permeability through the different stages of ischemic stroke and their role in the development of HT and stroke recovery.
Collapse
Affiliation(s)
| | - João André Sousa
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Ana Brás
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Carla Cecília
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Bruno Rodrigues
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Luciano Almendra
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Cristina Machado
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Gustavo Santo
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Fernando Silva
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - João Sargento-Freitas
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
52
|
Devos D, Hirsch E, Wyse R. Seven Solutions for Neuroprotection in Parkinson's Disease. Mov Disord 2020; 36:306-316. [PMID: 33184908 DOI: 10.1002/mds.28379] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/07/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by loss of dopaminergic neurons in the substantia nigra and accumulation of iron and alpha-synuclein; it follows a characteristic pattern throughout the nervous system. Despite decades of successful preclinical neuroprotective studies, no drug has then shown efficacy in clinical trials. Considering this dilemma, we have reviewed and organized solutions of varying importance that can be exclusive or additive, and we outline approaches to help generate successful development of neuroprotective drugs for PD: (1) select patients in which the targeted mechanism is involved in the pathological process associated with the monitoring of target engagement, (2) combine treatments that target multiple pathways, (3) establish earliest interventions and develop better prodromal biomarkers, (4) adopt rigorous methodology and specific disease-relevant designs for disease-modifying clinical trials, (5) customize drug with better brain biodistribution, (6) prioritize repurposed drugs as a first line approach, and (7) adapt preclinical models to the targeted mechanisms with translational biomarkers to increase their predictive value. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- David Devos
- Department of Medical Pharmacology, Expert Center for Parkinson, CHU-Lille, Lille Neuroscience & Cognition, Inserm, zUMR-S1172, LICEND, University of Lille, Lille, France
| | - Etienne Hirsch
- Institut du Cerveau-ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| | - Richard Wyse
- The Cure Parkinson's Trust, London, United Kingdom
| |
Collapse
|
53
|
Blood-Brain Barrier Modulation to Improve Glioma Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12111085. [PMID: 33198244 PMCID: PMC7697580 DOI: 10.3390/pharmaceutics12111085] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
The blood-brain barrier (BBB) is formed by brain microvascular endothelial cells that are sealed by tight junctions, making it a significant obstacle for most brain therapeutics. The poor BBB penetration of newly developed therapeutics has therefore played a major role in limiting their clinical success. A particularly challenging therapeutic target is glioma, which is the most frequently occurring malignant brain tumor. Thus, to enhance therapeutic uptake in tumors, researchers have been developing strategies to modulate BBB permeability. However, most conventional BBB opening strategies are difficult to apply in the clinical setting due to their broad, non-specific modulation of the BBB, which can result in damage to normal brain tissue. In this review, we have summarized strategies that could potentially be used to selectively and efficiently modulate the tumor BBB for more effective glioma treatment.
Collapse
|
54
|
Schlachet I, Moshe Halamish H, Sosnik A. Mixed Amphiphilic Polymeric Nanoparticles of Chitosan, Poly(vinyl alcohol) and Poly(methyl methacrylate) for Intranasal Drug Delivery: A Preliminary In Vivo Study. Molecules 2020; 25:molecules25194496. [PMID: 33008001 PMCID: PMC7582691 DOI: 10.3390/molecules25194496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 01/10/2023] Open
Abstract
Intranasal (i.n.) administration became an alternative strategy to bypass the blood-brain barrier and improve drug bioavailability in the brain. The main goal of this work was to preliminarily study the biodistribution of mixed amphiphilic mucoadhesive nanoparticles made of chitosan-g-poly(methyl methacrylate) and poly(vinyl alcohol)-g-poly(methyl methacrylate) and ionotropically crosslinked with sodium tripolyphosphate in the brain after intravenous (i.v.) and i.n. administration to Hsd:ICR mice. After i.v. administration, the highest nanoparticle accumulation was detected in the liver, among other peripheral organs. After i.n. administration of a 10-times smaller nanoparticle dose, the accumulation of the nanoparticles in off-target organs was much lower than after i.v. injection. In particular, the accumulation of the nanoparticles in the liver was 20 times lower than by i.v. When brains were analyzed separately, intravenously administered nanoparticles accumulated mainly in the "top" brain, reaching a maximum after 1 h. Conversely, in i.n. administration, nanoparticles were detected in the "bottom" brain and the head (maximum reached after 2 h) owing to their retention in the nasal mucosa and could serve as a reservoir from which the drug is released and transported to the brain over time. Overall, results indicate that i.n. nanoparticles reach similar brain bioavailability, though with a 10-fold smaller dose, and accumulate in off-target organs to a more limited extent and only after redistribution through the systemic circulation. At the same time, both administration routes seem to lead to differential accumulation in brain regions, and thus, they could be beneficial in the treatment of different medical conditions.
Collapse
|
55
|
Targeting Glioblastoma: Advances in Drug Delivery and Novel Therapeutic Approaches. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000124] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
56
|
Manthe RL, Loeck M, Bhowmick T, Solomon M, Muro S. Intertwined mechanisms define transport of anti-ICAM nanocarriers across the endothelium and brain delivery of a therapeutic enzyme. J Control Release 2020; 324:181-193. [PMID: 32389778 PMCID: PMC7720842 DOI: 10.1016/j.jconrel.2020.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
The interaction of drug delivery systems with tissues is key for their application. An example is drug carriers targeted to endothelial barriers, which can be transported to intra-endothelial compartments (lysosomes) or transcellularly released at the tissue side (transcytosis). Although carrier targeting valency influences this process, the mechanism is unknown. We studied this using polymer nanocarriers (NCs) targeted to intercellular adhesion molecule-1 (ICAM-1), an endothelial-surface glycoprotein whose expression is increased in pathologies characterized by inflammation. A bell-shaped relationship was found between NC targeting valency and the rate of transcytosis, where high and low NC valencies rendered less efficient transcytosis rates than an intermediate valency formulation. In contrast, an inverted bell-shape relationship was found for NC valency and lysosomal trafficking rates. Data suggested a model where NC valency plays an opposing role in the two sub-processes involved in transcytosis: NC binding-uptake depended directly on valency and exocytosis-detachment was inversely related to this parameter. This is because the greater the avidity of the NC-receptor interaction the more efficient uptake becomes, but NC-receptor detachment post-transport is more compromised. Cleavage of the receptor at the basolateral side of endothelial cells facilitated NC transcytosis, likely by helping NC detachment post-transport. Since transcytosis encompasses both sets of events, the full process finds an optimum at the intersection of these inverted relationships, explaining the bell-shaped behavior. NCs also trafficked to lysosomes from the apical side and, additionally, from the basolateral side in the case of high valency NCs which are slower at detaching from the receptor. This explains the opposite behavior of NC valency for transcytosis vs. lysosomal transport. Anti-ICAM NCs were verified to traffic into the brain after intravenous injection in mice, and both cellular and in vivo data showed that intermediate valency NCs resulted in higher delivery of a therapeutic enzyme, acid sphingomyelinase, required for types A and B Niemann-Pick disease.
Collapse
Affiliation(s)
- Rachel L Manthe
- Institute for Bioscience and Biotechnology Research (IBBR) and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Maximilian Loeck
- Institute for Bioengineering of Catalonia (IBEC) of the Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Tridib Bhowmick
- Institute for Bioscience and Biotechnology Research (IBBR) and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research (IBBR) and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research (IBBR) and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742-4450, USA; Institute for Bioengineering of Catalonia (IBEC) of the Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain; Institution of Catalonia for Research and Advanced Studies (ICREA), Barcelona 08910, Spain.
| |
Collapse
|
57
|
Tam DY, Ho JWT, Chan MS, Lau CH, Chang TJH, Leung HM, Liu LS, Wang F, Chan LLH, Tin C, Lo PK. Penetrating the Blood-Brain Barrier by Self-Assembled 3D DNA Nanocages as Drug Delivery Vehicles for Brain Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:28928-28940. [PMID: 32432847 DOI: 10.1021/acsami.0c02957] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The development of biocompatible drug delivery vehicles for cancer therapy in the brain remains a big challenge. In this study, we designed self-assembled DNA nanocages functionalized with or without blood-brain barrier (BBB)-targeting ligands, d and we investigated their penetration across the BBB. Our DNA nanocages were not cytotoxic and they were substantially taken up in brain capillary endothelial cells and Uppsala 87 malignant glioma (U-87 MG) cells. We found that ligand modification is not essential for this DNA system as the ligand-free DNA nanocages (LF-NCs) could still cross the BBB by endocytosis inin vitro and in vivo models. Our spherical DNA nanocages were more permeable across the BBB compared with tubular DNA nanotubes. Remarkably, in vivo studies revealed that DNA nanocages could carry anticancer drugs across the BBB and inhibit the tumor growth in a U-87 MG xenograft mouse model. This is the first example showing the potential of DNA nanocages as innovative delivery vehicles to the brain for cancer therapy. Unlike other delivery systems, our work suggest that a DNA nanocage-based platform provides a safe and cost-effective tool for targeted delivery to the brain and therapy for brain tumors.
Collapse
Affiliation(s)
- Dick Yan Tam
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Jonathan Weng-Thim Ho
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Miu Shan Chan
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Cia Hin Lau
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Tristan Juin Han Chang
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Hoi Man Leung
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Ling Sum Liu
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Fei Wang
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Leanne Lai Hang Chan
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Chung Tin
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Pik Kwan Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
58
|
Naqvi S, Panghal A, Flora SJS. Nanotechnology: A Promising Approach for Delivery of Neuroprotective Drugs. Front Neurosci 2020; 14:494. [PMID: 32581676 PMCID: PMC7297271 DOI: 10.3389/fnins.2020.00494] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) disorders especially neurodegenerative disorders are the major challenge for public health and demand the great attention of researchers to protect people against them. In past few decades, different treatment strategies have been adopted, but their therapeutic efficacy are not enough and have only shown partial mitigation of symptoms. Blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BSCFB) guard the CNS from harmful substances and pose as the major challenges in delivering drugs into CNS for treatment of CNS complications such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), stroke, epilepsy, brain tumors, multiple sclerosis (MS), and encephalitis, etc. Nanotechnology has come out as an exciting and promising new platform of treating neurological disorders and has shown great potential to overcome problems related to the conventional treatment approaches. Molecules can be nanoengineered to carry out multiple specific functions such as to cross the BBB, target specific cell or signaling pathway, respond to endogenous stimuli, and act as a vehicle for gene delivery, support nerve regeneration and cell survival. In present review, the role of nanocarrier systems such as liposomes, micelles, solid lipid nanoparticles (SLNPs), dendrimers, and nanoemulsions for delivery of various neurotherapeutic agents has been discussed, besides this, their mechanism of action, and nanoformulation of different neuroprotective agents like curcumin, edaravone, nerve growth factors in CNS disorders like Alzheimer’s, Parkinsonism, epilepsy, stroke, and brain tumors has been reviewed.
Collapse
Affiliation(s)
- Saba Naqvi
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - Archna Panghal
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - S J S Flora
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, India
| |
Collapse
|
59
|
Cano A, Sánchez-López E, Ettcheto M, López-Machado A, Espina M, Souto EB, Galindo R, Camins A, García ML, Turowski P. Current advances in the development of novel polymeric nanoparticles for the treatment of neurodegenerative diseases. Nanomedicine (Lond) 2020; 15:1239-1261. [PMID: 32370600 DOI: 10.2217/nnm-2019-0443] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Effective intervention is essential to combat the coming epidemic of neurodegenerative (ND) diseases. Nanomedicine can overcome restrictions of CNS delivery imposed by the blood-brain barrier, and thus be instrumental in preclinical discovery and therapeutic intervention of ND diseases. Polymeric nanoparticles (PNPs) have shown great potential and versatility to encapsulate several compounds simultaneously in controlled drug-delivery systems and target them to the deepest brain regions. Here, we critically review recent advances in the development of drugs incorporated into PNPs and summarize the molecular changes and functional effects achieved in preclinical models of the most common ND disorders. We also briefly discuss the many challenges remaining to translate these findings and technological advances successfully to current clinical settings.
Collapse
Affiliation(s)
- Amanda Cano
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Elena Sánchez-López
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacology, Toxicology & Therapeutic Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Unit of Biochemistry & Pharmacology, Faculty of Medicine & Health Sciences, University of Rovira i Virgili, Reus (Tarragona), Spain
| | - Ana López-Machado
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain
| | - Marta Espina
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar 4710-057, Braga, Portugal
| | - Ruth Galindo
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Unit of Synthesis & Biomedical Applications of Peptides, Department of Biomedical Chemistry, Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacology, Toxicology & Therapeutic Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain
| | - Maria Luisa García
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Patric Turowski
- UCL Institute of Ophthalmology, University College of London, London, UK
| |
Collapse
|
60
|
Rabiei M, Kashanian S, Samavati SS, Jamasb S, McInnes SJ. Active Targeting Towards and Inside the Brain based on Nanoparticles: A Review. Curr Pharm Biotechnol 2020; 21:374-383. [DOI: 10.2174/1389201020666191203094057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/08/2019] [Accepted: 11/22/2019] [Indexed: 11/22/2022]
Abstract
Background:
Treatment of neurological diseases using systemic and non-surgical techniques
presents a significant challenge in medicine. This challenge is chiefly associated with the condensation
and coherence of the brain tissue.
Methods:
The coherence structure of the brain is due to the presence of the blood-brain barrier (BBB),
which consists of a continuous layer of capillary endothelial cells. The BBB prevents most drugs from
entering the brain tissue and is highly selective, permitting only metabolic substances and nutrients to
pass through.
Results:
Although this challenge has caused difficulties for the treatment of neurological diseases, it
has opened up a broad research area in the field of drug delivery. Through the utilization of nanoparticles
(NPs), nanotechnology can provide the ideal condition for passing through the BBB.
Conclusion:
NPs with suitable dimensions and optimum hydrophobicity and charge, as well as appropriate
functionalization, can accumulate in the brain. Furthermore, NPs can facilitate the targeted delivery
of therapeutics into the brain areas involved in Alzheimer’s disease, Parkinson’s disease, stroke,
glioma, migraine, and other neurological disorders. This review describes these methods of actively
targeting specific areas of the brain.
Collapse
Affiliation(s)
- Morteza Rabiei
- Department of Nanobiotechnology, Razi University, Kermanshah, Iran
| | | | | | - Shahriar Jamasb
- Department of Biomedical Engineering, Hamedan University of Technology, Hamedan, 65169-13733, Iran
| | - Steven J.P. McInnes
- University of South Australia, Division of Information Technology, Engineering and the Environment, Mawson Lakes, Mawson Lakes 5095, Australia
| |
Collapse
|
61
|
Islam SU, Shehzad A, Ahmed MB, Lee YS. Intranasal Delivery of Nanoformulations: A Potential Way of Treatment for Neurological Disorders. Molecules 2020; 25:molecules25081929. [PMID: 32326318 PMCID: PMC7221820 DOI: 10.3390/molecules25081929] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/17/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Although the global prevalence of neurological disorders such as Parkinson’s disease, Alzheimer’s disease, glioblastoma, epilepsy, and multiple sclerosis is steadily increasing, effective delivery of drug molecules in therapeutic quantities to the central nervous system (CNS) is still lacking. The blood brain barrier (BBB) is the major obstacle for the entry of drugs into the brain, as it comprises a tight layer of endothelial cells surrounded by astrocyte foot processes that limit drugs’ entry. In recent times, intranasal drug delivery has emerged as a reliable method to bypass the BBB and treat neurological diseases. The intranasal route for drug delivery to the brain with both solution and particulate formulations has been demonstrated repeatedly in preclinical models, including in human trials. The key features determining the efficacy of drug delivery via the intranasal route include delivery to the olfactory area of the nares, a longer retention time at the nasal mucosal surface, enhanced penetration of the drugs through the nasal epithelia, and reduced drug metabolism in the nasal cavity. This review describes important neurological disorders, challenges in drug delivery to the disordered CNS, and new nasal delivery techniques designed to overcome these challenges and facilitate more efficient and targeted drug delivery. The potential for treatment possibilities with intranasal transfer of drugs will increase with the development of more effective formulations and delivery devices.
Collapse
Affiliation(s)
- Salman Ul Islam
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (S.U.I.); (M.B.A.)
| | - Adeeb Shehzad
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Muhammad Bilal Ahmed
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (S.U.I.); (M.B.A.)
| | - Young Sup Lee
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (S.U.I.); (M.B.A.)
- Correspondence: ; Tel.: +82-53-950-6353; Fax: +82-53-943-2762
| |
Collapse
|
62
|
LRP-1 Mediated Endocytosis of EFE Across the Blood–Brain Barrier; Protein–Protein Interaction and Molecular Dynamics Analysis. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10065-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
63
|
Aggarwal N, Qamar Z, Rehman S, Baboota S, Ali J. Orally Administered Nanotherapeutics For Parkinson's Disease: An Old Delivery System Yet More Acceptable. Curr Pharm Des 2020; 26:2280-2290. [PMID: 32250217 DOI: 10.2174/1381612826666200406072451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 03/04/2020] [Indexed: 12/17/2022]
Abstract
As per the present global scenario, Parkinson's disease (PD) is considered to be the second most common neurodegenerative disorder which is a keen area of interest among researchers. The conventional therapies generally employed against PD are associated with serious drawbacks including limited transport across selectively permeable BBB, hepatic metabolism, intestinal barrier, etc. This urges the need to develop novel therapeutic alternatives. The oral route being the most preferred route of administration needs to be explored for new and more intelligent drug delivery systems. Nanotechnology has been proposed to play a promising role in reversing the progression of the disease via the oral route. Nanocarriers, namely nanoparticles, lipid nanoparticles, nanoemulsions, nanocrystals, nanomicellar formulations, self-nanoemulsifying drug delivery systems and alginate nanocomposites have been investigated upon to modulate the fate of drugs inside the human body when administered orally. The development of various nanotherapeutics for the treatment of PD has been reviewed, depicting an enhanced bioavailability to provide a desired therapeutic outcome. The new advances in the therapy have been explored and highlighted through the body of this review. However, a therapeutically effective concentration at the target site remains a challenge, therefore extensive exploration in the field of nanotherapeutics may facilitate superior drug delivery to CNS via oral route thereby improving the state of disease progression.
Collapse
Affiliation(s)
- Nidhi Aggarwal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Zufika Qamar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Saleha Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| |
Collapse
|
64
|
Xia X, Pollock N, Zhou J, Rossi J. Tissue-Specific Delivery of Oligonucleotides. Methods Mol Biol 2020; 2036:17-50. [PMID: 31410789 DOI: 10.1007/978-1-4939-9670-4_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
From the initial discovery of short-interfering RNA (siRNA) and antisense oligonucleotides for specific gene knockdown at the posttranscriptional level to the current CRISPR-Cas9 system offering gene editing at the genomic level, oligonucleotides, in addition to their biological functions in storing and conveying genetic information, provide the most prominent solutions to targeted gene therapies. Nonetheless, looking into the future of curing cancer and acute diseases, researchers are only cautiously optimistic as the cellular delivery of these polyanionic biomacromolecules is still the biggest hurdle for their therapeutic realization. To overcome the delivery obstacle, oligonucleotides have been encapsulated within or conjugated with delivery vehicles for enhanced membrane penetration, improved payload, and tissue-specific delivery. Such delivery systems include but not limited to virus-based vehicles, gold-nanoparticle vehicles, formulated liposomes, and synthetic polymers. In this chapter, delivery challenges imposed by biological barriers are briefly discussed; followed by recent advances in tissue-specific oligonucleotide delivery utilizing both viral and nonviral delivery vectors, discussing their advantages, and how judicious design and formulation could improve and expand their potential as delivery vehicles.
Collapse
Affiliation(s)
- Xin Xia
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Nicolette Pollock
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Jiehua Zhou
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - John Rossi
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
65
|
Jin GZ, Chakraborty A, Lee JH, Knowles JC, Kim HW. Targeting with nanoparticles for the therapeutic treatment of brain diseases. J Tissue Eng 2020; 11:2041731419897460. [PMID: 32180936 PMCID: PMC7057401 DOI: 10.1177/2041731419897460] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Brain diseases including neurodegenerative disorders and tumours are among the most serious health problems, degrading the quality of life and causing massive economic cost. Nanoparticles that load and deliver drugs and genes have been intensively studied for the treatment of brain diseases, and have demonstrated some biological effects in various animal models. Among other efforts taken in the nanoparticle development, targeting of blood brain barrier, specific cell type or local intra-/extra-cellular space is an important strategy to enhance the therapeutic efficacy of the nanoparticle delivery systems. This review underlies the targeting issue in the nanoparticle development for the treatment of brain diseases, taking key exemplar studies carried out in various in vivo models.
Collapse
Affiliation(s)
- Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Atanu Chakraborty
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| | - Jonathan C Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, London, UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
66
|
Fowler S, Chen WLK, Duignan DB, Gupta A, Hariparsad N, Kenny JR, Lai WG, Liras J, Phillips JA, Gan J. Microphysiological systems for ADME-related applications: current status and recommendations for system development and characterization. LAB ON A CHIP 2020; 20:446-467. [PMID: 31932816 DOI: 10.1039/c9lc00857h] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Over the last decade, progress has been made on the development of microphysiological systems (MPS) for absorption, distribution, metabolism, and excretion (ADME) applications. Central to this progress has been proof of concept data generated by academic and industrial institutions followed by broader characterization studies, which provide evidence for scalability and applicability to drug discovery and development. In this review, we describe some of the advances made for specific tissue MPS and outline the desired functionality for such systems, which are likely to make them applicable for practical use in the pharmaceutical industry. Single organ MPS platforms will be valuable for modelling tissue-specific functions. However, dynamic organ crosstalk, especially in the context of disease or toxicity, can only be obtained with the use of inter-linked MPS models which will enable scientists to address questions at the intersection of pharmacokinetics (PK) and efficacy, or PK and toxicity. In the future, successful application of MPS platforms that closely mimic human physiology may ultimately reduce the need for animal models to predict ADME outcomes and decrease the overall risk and cost associated with drug development.
Collapse
Affiliation(s)
- Stephen Fowler
- Pharma Research and Early Development, F.Hoffmann-La Roche Ltd, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | | | - David B Duignan
- Department of Drug Metabolism, Pharmacokinetics & Bioanalysis, AbbVie Bioresearch Center, Worcester, Massachusetts 01605, USA
| | - Anshul Gupta
- Amgen Research, 360 Binney St, Cambridge, MA 02141, USA
| | - Niresh Hariparsad
- Department of Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA, USA
| | - Jane R Kenny
- DMPK, Genentech, 1 DNA Way, South San Francisco 94080, USA
| | | | - Jennifer Liras
- Medicine Design, Pfizer Inc, 1 Portland Ave, Cambridge, MA 02139, USA
| | | | - Jinping Gan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb R&D, PO Box 4000, Princeton, NJ 08543-4000, USA.
| |
Collapse
|
67
|
Qin M, Wang L, Wu D, Williams CK, Xu D, Kranz E, Guo Q, Guan J, Vinters HV, Lee Y, Xie Y, Luo Y, Sun G, Sun X, He Z, Lu Y, Kamata M, Wen J, Chen ISY. Enhanced Delivery of Rituximab Into Brain and Lymph Nodes Using Timed-Release Nanocapsules in Non-Human Primates. Front Immunol 2020; 10:3132. [PMID: 32047498 PMCID: PMC6996053 DOI: 10.3389/fimmu.2019.03132] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor metastasis into the central nervous system (CNS) and lymph nodes (LNs) is a major obstacle for effective therapies. Therapeutic monoclonal antibodies (mAb) have revolutionized tumor treatment; however, their efficacy for treating metastatic tumors-particularly, CNS and LN metastases-is poor due to inefficient penetration into the CNS and LNs following intravenous injection. We recently reported an effective delivery of mAb to the CNS by encapsulating the anti-CD20 mAb rituximab (RTX) within a thin shell of polymer that contains the analogs of choline and acetylcholine receptors. This encapsulated RTX, denoted as n-RTX, eliminated lymphoma cells systemically in a xenografted humanized mouse model using an immunodeficient mouse as a recipient of human hematopoietic stem/progenitor cells and fetal thymus more effectively than native RTX; importantly, n-RTX showed notable anti-tumor effect on CNS metastases which is unable to show by native RTX. As an important step toward future clinical translation of this technology, we further analyzed the properties of n-RTX in immunocompetent animals, rats, and non-human primates (NHPs). Our results show that a single intravenous injection of n-RTX resulted in 10-fold greater levels in the CNS and 2-3-fold greater levels in the LNs of RTX, respectively, than the injection of native RTX in both rats and NHPs. In addition, we demonstrate the enhanced delivery and efficient B-cell depletion in lymphoid organs of NHPs with n-RTX. Moreover, detailed hematological analysis and liver enzyme activity tests indicate n-RTX treatment is safe in NHPs. As this nanocapsule platform can be universally applied to other therapeutic mAbs, it holds great promise for extending mAb therapy to poorly accessible body compartments.
Collapse
Affiliation(s)
- Meng Qin
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States.,UCLA AIDS Institute, Los Angeles, CA, United States
| | - Lan Wang
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States.,UCLA AIDS Institute, Los Angeles, CA, United States
| | - Di Wu
- Department of Chemical and Biomolecular Engineering, School of Engineering, UCLA, Los Angeles, CA, United States
| | - Christopher K Williams
- Departments of Pathology & Laboratory Medicine (Neuropathology) and Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Duo Xu
- Department of Chemical and Biomolecular Engineering, School of Engineering, UCLA, Los Angeles, CA, United States
| | - Emiko Kranz
- UCLA AIDS Institute, Los Angeles, CA, United States.,Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Qi Guo
- UCLA AIDS Institute, Los Angeles, CA, United States.,School of Nursing, UCLA, Los Angeles, CA, United States
| | - Jiaoqiong Guan
- Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, China
| | - Harry V Vinters
- Departments of Pathology & Laboratory Medicine (Neuropathology) and Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - YooJin Lee
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States.,UCLA AIDS Institute, Los Angeles, CA, United States
| | - Yiming Xie
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States.,UCLA AIDS Institute, Los Angeles, CA, United States
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhanlong He
- Institute of Medical Biology, Peking Union Medical College, Chinese Academy of Medical Sciences, Kunming, China
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, School of Engineering, UCLA, Los Angeles, CA, United States
| | - Masakazu Kamata
- UCLA AIDS Institute, Los Angeles, CA, United States.,Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States.,UCLA AIDS Institute, Los Angeles, CA, United States
| | - Irvin S Y Chen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States.,UCLA AIDS Institute, Los Angeles, CA, United States
| |
Collapse
|
68
|
Mahmoud BS, AlAmri AH, McConville C. Polymeric Nanoparticles for the Treatment of Malignant Gliomas. Cancers (Basel) 2020; 12:E175. [PMID: 31936740 PMCID: PMC7017235 DOI: 10.3390/cancers12010175] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/19/2019] [Accepted: 01/06/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant gliomas are one of the deadliest forms of brain cancer and despite advancements in treatment, patient prognosis remains poor, with an average survival of 15 months. Treatment using conventional chemotherapy does not deliver the required drug dose to the tumour site, owing to insufficient blood brain barrier (BBB) penetration, especially by hydrophilic drugs. Additionally, low molecular weight drugs cannot achieve specific accumulation in cancerous tissues and are characterized by a short circulation half-life. Nanoparticles can be designed to cross the BBB and deliver their drugs within the brain, thus improving their effectiveness for treatment when compared to administration of the free drug. The efficacy of nanoparticles can be enhanced by surface PEGylation to allow more specificity towards tumour receptors. This review will provide an overview of the different therapeutic strategies for the treatment of malignant gliomas, risk factors entailing them as well as the latest developments for brain drug delivery. It will also address the potential of polymeric nanoparticles in the treatment of malignant gliomas, including the importance of their coating and functionalization on their ability to cross the BBB and the chemistry underlying that.
Collapse
Affiliation(s)
- Basant Salah Mahmoud
- College of Medical and Dental Sciences, School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK; (B.S.M.); or
- Hormones Department, Medical Research Division, National Research Centre, El Buhouth St., Dokki, Cairo 12622, Egypt
| | - Ali Hamod AlAmri
- College of Medical and Dental Sciences, School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK; (B.S.M.); or
- College of Pharmacy, King Khalid University, Abha 62585, Saudi Arabia
| | - Christopher McConville
- College of Medical and Dental Sciences, School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK; (B.S.M.); or
| |
Collapse
|
69
|
Mittal S, Ashhar MU, Qizilbash FF, Qamar Z, Narang JK, Kumar S, Ali J, Baboota S. Ligand Conjugated Targeted Nanotherapeutics for Treatment of Neurological Disorders. Curr Pharm Des 2020; 26:2291-2305. [PMID: 32303160 DOI: 10.2174/1381612826666200417141600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/26/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Human brain is amongst the most complex organs in human body, and delivery of therapeutic agents across the brain is a tedious task. Existence of blood brain barrier (BBB) protects the brain from invasion of undesirable substances; therefore it hinders the transport of various drugs used for the treatment of different neurological diseases including glioma, Parkinson's disease, Alzheimer's disease, etc. To surmount this barrier, various approaches have been used such as the use of carrier mediated drug delivery; use of intranasal route, to avoid first pass metabolism; and use of ligands (lactoferrin, apolipoprotein) to transport the drug across the BBB. Ligands bind with proteins present on the cell and facilitate the transport of drug across the cell membrane via. receptor mediated, transporter mediated or adsorptive mediated transcytosis. OBJECTIVE The main focus of this review article is to illustrate various studies performed using ligands for delivering drug across BBB; it also describes the procedure used by various researchers for conjugating the ligands to the formulation to achieve targeted action. METHODS Research articles that focused on the used of ligand conjugation for brain delivery and compared the outcome with unconjugated formulation were collected from various search engines like PubMed, Science Direct and Google Scholar, using keywords like ligands, neurological disorders, conjugation, etc. Results and Conclusion: Ligands have shown great potential in delivering drug across BBB for treatment of various diseases, yet extensive research is required so that the ligands can be used clinically for treating neurological diseases.
Collapse
Affiliation(s)
- Saurabh Mittal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Muhammad U Ashhar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Farheen F Qizilbash
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Zufika Qamar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Jasjeet K Narang
- Department of Pharmaceutics, Khalsa College of Pharmacy, Amritsar, Punjab, India
| | - Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Uttar Pradesh, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi-110062, India
| |
Collapse
|
70
|
Ghosh S, Lalani R, Patel V, Bhowmick S, Misra A. Surface engineered liposomal delivery of therapeutics across the blood brain barrier: recent advances, challenges and opportunities. Expert Opin Drug Deliv 2019; 16:1287-1311. [DOI: 10.1080/17425247.2019.1676721] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Formulation Development Department-Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
71
|
Xi J, Liu H. Recent Advances in the Design of Self‐Delivery Amphiphilic Drugs and Vaccines. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jingchao Xi
- Department of Chemical Engineering and Materials Science Wayne State University Detroit MI 48202 USA
| | - Haipeng Liu
- Department of Chemical Engineering and Materials Science Wayne State University Detroit MI 48202 USA
- Department of Oncology Wayne State University Detroit MI 48201 United States
- Tumor Biology and Microenvironment Program Barbara Ann Karmanos Cancer Institute Detroit MI 48201 United States
| |
Collapse
|
72
|
Varini K, Lécorché P, Sonnette R, Gassiot F, Broc B, Godard M, David M, Faucon A, Abouzid K, Ferracci G, Temsamani J, Khrestchatisky M, Jacquot G. Target engagement and intracellular delivery of mono- and bivalent LDL receptor-binding peptide-cargo conjugates: Implications for the rational design of new targeted drug therapies. J Control Release 2019; 314:141-161. [PMID: 31644939 DOI: 10.1016/j.jconrel.2019.10.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
Targeted delivery to specific tissues and subcellular compartments is of paramount importance to optimize therapeutic or diagnostic interventions while minimizing side-effects. Using recently identified LDL receptor (LDLR) -targeting small synthetic peptide-vectors conjugated to model cargos of different nature and size, we investigated in LDLR-expressing cells the impact of vector-cargo molecular engineering and coupling valency, as well as the cellular exposure duration on their target engagement and intracellular trafficking and delivery profiles. All vector-cargo conjugates evaluated were found to be delivered to late compartments together with the natural ligand LDL, although to varying extents and with different kinetics. Partial recycling together with the LDLR was also consistently observed. Under continuous cellular exposure, the extent of intracellular vector-cargo delivery primarily relies on their endosomal unloading potential. In this condition, the highest intracellular delivery potential was observed with a monovalent conjugate displaying a rather high LDLR dissociation rate. On the contrary, under transient cellular exposure followed by chase, low dissociation-rate bivalent conjugates revealed a higher intracellular delivery potential than the monovalent conjugate. This was shown to rely on their ability to undergo multiple endocytosis-recycling rounds, with limited release in the ligand-free medium. The absence of reciprocal competition with the natural ligand LDL on their respective intracellular trafficking was also demonstrated, which is essential in terms of potential safety liabilities. These results demonstrate that not only molecular engineering of new therapeutic conjugates of interest, but also the cellular exposure mode used during in vitro evaluations are critical to anticipate and optimize their delivery potential.
Collapse
Affiliation(s)
- K Varini
- VECT-HORUS SAS, Marseille, France; Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | | | | | | - B Broc
- VECT-HORUS SAS, Marseille, France
| | - M Godard
- VECT-HORUS SAS, Marseille, France
| | - M David
- VECT-HORUS SAS, Marseille, France
| | - A Faucon
- VECT-HORUS SAS, Marseille, France
| | | | - G Ferracci
- Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | | - M Khrestchatisky
- Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | |
Collapse
|
73
|
Meenu M, Reeta KH, Dinda AK, Kottarath SK, Gupta YK. Evaluation of sodium valproate loaded nanoparticles in acute and chronic pentylenetetrazole induced seizure models. Epilepsy Res 2019; 158:106219. [PMID: 31726286 DOI: 10.1016/j.eplepsyres.2019.106219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/27/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Efficacy of sodium valproate in epilepsy is limited by its poor blood brain barrier penetration and side effects. Nanoparticles may offer a better drug delivery system to overcome these limitations. This study evaluated the efficacy of sodium valproate encapsulated in nanoparticles in pentylenetetrazole (PTZ) induced acute and kindling models of seizures in male Wistar rats. METHODS Poly lactic-co-glycolic acid (PLGA) based, polysorbate 80 stabilized sodium valproate loaded nanoparticles (nano sodium valproate) and rhodamine loaded nanoparticles (RLN) were formulated by double emulsion- solvent evaporation method and characterized for their size, shape, zeta potential and drug loading percentage. RLN was used to demonstrate blood brain barrier (BBB) permeability of nanoparticles. Serum drug levels were estimated using high performance liquid chromatography. The efficacy of standard sodium valproate (300 mg/kg) and nano sodium valproate (∼300, ∼150 and ∼75 mg/kg of sodium valproate) were evaluated in experimental animal models of seizures along with their effects on behavioral and oxidative stress parameters. Drugs were administered 60 min before PTZ in acute model. In the kindling model, drugs were administered every day while PTZ was administered on alternate days 60 min after drug administration. All the study drugs/compounds were administered intraperitoneally. RESULTS RLN were observed to be clustered in cortex which implied that the nanoparticles crossed BBB. Both standard sodium valproate and nano sodium valproate reached therapeutic serum level at 15 min and 1 h, but were undetectable in serum at 24 h. In acute PTZ (60 mg/kg) model, nano sodium valproate (∼300 mg/kg of sodium valproate) and standard sodium valproate showed protection against seizures till 6 h and 4 h, respectively. There were significant behavioral impairment and oxidative stress with standard sodium valproate in acute model as compared to nano sodium valproate at 6 h. In kindling model, induced with PTZ (30 mg/kg, every alternate day for 42 days), complete protection from seizures was observed with nano sodium valproate (∼150 mg/kg and ∼75 mg/kg of sodium valproate) and standard sodium valproate (300 mg/kg). Similarly, significant protection from behavioral impairment and oxidative stress was observed with standard sodium valproate and nano sodium valproate as compared to PTZ. CONCLUSION When compared to conventional therapy, nano sodium valproate showed protection from seizures at reduced doses and for a longer duration in animal models of epilepsy. This study suggests the potential of nano sodium valproate in the treatment of epilepsy.
Collapse
Affiliation(s)
- Meenakshi Meenu
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| | - Amit Kumar Dinda
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Yogendra Kumar Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
74
|
Su S, Wang J, Qiu J, Martinez-Zaguilan R, Sennoune SR, Wang S. In vitro study of transportation of porphyrin immobilized graphene oxide through blood brain barrier. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 107:110313. [PMID: 31761227 DOI: 10.1016/j.msec.2019.110313] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/03/2019] [Accepted: 10/13/2019] [Indexed: 01/09/2023]
Abstract
Blood brain barrier (BBB), a barrier formed by endothelial cells, separates the brain from the circulatory system and protects the stability of central neural system normally, however, it also results in low permeability of vast majority of drugs for brain disease therapy. In this work, the cytotoxicity, uptake and transportation of 2D graphene nanosheet through BBB were investigated in in vitro models of BBB constructed by human brain microvascular endothelia cells (hBMECs). Permeability of two types of graphene nanosheet, including graphene oxide (GO) and porphyrin conjugated graphene oxide (PGO) through BBB were studied. With hydrophobic chemicals conjugation on its surface, permeability of PGO was greatly improved compared to GO. Furthermore, transportation behavior of assorted sizes of PGO obtained by differential velocity centrifugation through BBB was also explored, revealing that PGO with larger size has higher permeability than smaller-size PGO. The significant improved permeability of 2D graphene nanosheet through BBB compared to traditional drugs provides promising applications in drug delivery and disease therapy for brain disease in the near future.
Collapse
Affiliation(s)
- Siheng Su
- Department of Mechanical Engineering, California State University at Fullerton, 800 N State College Blvd, Fullerton, CA, 92831, USA.
| | - Jilong Wang
- Key Laboratory of Textile Science & Technology of Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, PR China
| | - Jingjing Qiu
- Department of Mechanical Engineering, Texas Tech University, 2500 Broadway, Lubbock, TX, 79409, USA.
| | - Raul Martinez-Zaguilan
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA
| | - Souad R Sennoune
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA
| | - Shiren Wang
- Department of Industrial & Systems Engineering, Texas A&M University, 400 Bizzell Street, College Station, TX, 77843, USA.
| |
Collapse
|
75
|
Bender H, Noyes N, Annis JL, Hitpas A, Mollnow L, Croak K, Kane S, Wagner K, Dow S, Zabel M. PrPC knockdown by liposome-siRNA-peptide complexes (LSPCs) prolongs survival and normal behavior of prion-infected mice immunotolerant to treatment. PLoS One 2019; 14:e0219995. [PMID: 31329627 PMCID: PMC6645518 DOI: 10.1371/journal.pone.0219995] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/05/2019] [Indexed: 11/29/2022] Open
Abstract
Prion diseases are members of neurodegenerative protein misfolding diseases (NPMDs) that include Alzheimer's, Parkinson's and Huntington diseases, amyotrophic lateral sclerosis, tauopathies, traumatic brain injuries, and chronic traumatic encephalopathies. No known therapeutics extend survival or improve quality of life of humans afflicted with prion disease. We and others developed a new approach to NPMD therapy based on reducing the amount of the normal, host-encoded protein available as substrate for misfolding into pathologic forms, using RNA interference, a catabolic pathway that decreases levels of mRNA encoding a particular protein. We developed a therapeutic delivery system consisting of small interfering RNA (siRNA) complexed to liposomes and addressed to the central nervous system using a targeting peptide derived from rabies virus glycoprotein. These liposome-siRNA-peptide complexes (LSPCs) cross the blood-brain barrier and deliver PrP siRNA to neuronal cells to decrease expression of the normal cellular prion protein, PrPC, which acts as a substrate for prion replication. Here we show that LSPCs can extend survival and improve behavior of prion-infected mice that remain immunotolerant to treatment. LSPC treatment may be a viable therapy for prion and other NPMDs that can improve the quality of life of patients at terminal disease stages.
Collapse
Affiliation(s)
- Heather Bender
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Noelle Noyes
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States of America
| | - Jessica L. Annis
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Amanda Hitpas
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Luke Mollnow
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Kendra Croak
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Sarah Kane
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Kaitlyn Wagner
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Steven Dow
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Mark Zabel
- Prion Research Center, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States of America
| |
Collapse
|
76
|
Kang JH, Ko YT. Dual-selective photodynamic therapy with a mitochondria-targeted photosensitizer and fiber optic cannula for malignant brain tumors. Biomater Sci 2019; 7:2812-2825. [PMID: 31066391 DOI: 10.1039/c9bm00403c] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Among brain tumors, glioblastoma multiforme (GBM) is the most common and aggressive form (WHO grade IV) with a median survival of only 14.6 months in adults. Photodynamic therapy (PDT) is a combination of a photosensitizer (PS), light and molecular oxygen, and considered a promising treatment for GBM. Therapeutic outcomes of PDT rely on ROS generation in a tumor microenvironment, which can be controlled with dual selectivity by localization of the photosensitizer and confinement of light to the targeted tumor microenvironment. We previously demonstrated the photodynamic anticancer efficacy of mitochondrial-targeted photosensitizer-loaded albumin nanoparticles (PS@chol-BSA NPs). In this study, the photodynamic therapeutic effect of PS@chol-BSA NPs was further enhanced by confinement of light using a fiber optic cannula in orthotopic GBM-xenografted mice. In vitro cellular uptake and phototoxicity of PS@chol-BSA NPs were evaluated in brain tumor (U87MG) and endothelial (bEnd.3) cells. In vivo biodistribution was determined by an in vivo imaging system (IVIS) and the photodynamic efficacy was evaluated with confined laser irradiation. PS@chol-BSA NPs showed higher cellular uptake and phototoxicity in U87MG cells than in bEnd.3 cells. PS@chol-BSA NPs showed a brain tumor accumulation of 0.2%ID within 2 h and remain in the brain tumor for 22 h. When compared to the control group, there was remarkable suppression in tumor growth by laser irradiation with and without the fiber optic cannula at a dose of 1 mg kg-1, in which significant tumor suppression up to 40% was observed with confined laser irradiation. Together, dual-selective photodynamic therapy with a mitochondria-targeted photosensitizer and fiber optic cannula provides a promising therapeutic strategy for malignant brain tumors.
Collapse
Affiliation(s)
- Ji Hee Kang
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, Republic of Korea 21936.
| | - Young Tag Ko
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, Republic of Korea 21936.
| |
Collapse
|
77
|
Peviani M, Capasso Palmiero U, Cecere F, Milazzo R, Moscatelli D, Biffi A. Biodegradable polymeric nanoparticles administered in the cerebrospinal fluid: Brain biodistribution, preferential internalization in microglia and implications for cell-selective drug release. Biomaterials 2019; 209:25-40. [PMID: 31026609 DOI: 10.1016/j.biomaterials.2019.04.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 12/31/2022]
Abstract
Cell-selective drug release in the central nervous system (CNS) holds great promise for the treatment of many CNS disorders but it is still challenging. We previously demonstrated that polymeric nanoparticles (NPs) injected intra-parenchyma in the CNS can be internalized specifically in microglia/macrophages surrounding the injection site. Here, we explored NPs administration in the cerebrospinal fluid (CSF) to achieve a wider spreading and increased cell targeting throughout the CNS; we generated new NPs variants and studied the effect of modifying size and surface charge on NPs biodistribution and cellular uptake. Intra-cerebroventricular administration resulted in prevalent localization of the NPs in proximity to stem-cell niches, such as around the lateral ventricles, the subventricular zone and the rostral migratory stream. NPs internalization occurred preferentially in brain myeloid cells/microglia. We demonstrated that brain biodistribution and extent of internalization in microglia are influenced by NPs dimensions and can be improved by applying a transient disruption of the blood-brain barrier with mannitol, leading to NPs internalization in up to 25% of brain myeloid/microglia cells. A fraction of the targeted cells was positive for markers of proliferation or stained positive for stemness/progenitor-cell markers such as Nestin, c-kit, or NG2. Interestingly, through these newly formulated NPs we obtained controlled and selective release of drugs otherwise difficult to formulate (such as busulfan and etoposide) to the target cells, preventing unwanted side effects and the toxicity obtained by direct brain delivery of the not encapsulated drugs. Overall, these data provide proof of concept of the applicability of these novel NP-based drug formulations for achieving internalization not only in mature microglia but also possibly in more immature myeloid cells in the brain and pave the way for brain-restricted microglia-targeted drug delivery regimens.
Collapse
Affiliation(s)
- Marco Peviani
- Gene Therapy Program, Dana Farber/Boston Children's Cancer and Blood Disorders Center, 450 Brookline Ave., 02215, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 48, 20156, Milan, Italy.
| | - Umberto Capasso Palmiero
- Dipartimento di Chimica, Materiali ed Ingegneria Chimica, Politecnico di Milano, 20131, Milan, Italy; Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg 1, 8093 Zürich, Switzerland
| | - Francesca Cecere
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 48, 20156, Milan, Italy
| | - Rita Milazzo
- San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 48, 20156, Milan, Italy
| | - Davide Moscatelli
- Dipartimento di Chimica, Materiali ed Ingegneria Chimica, Politecnico di Milano, 20131, Milan, Italy
| | - Alessandra Biffi
- Gene Therapy Program, Dana Farber/Boston Children's Cancer and Blood Disorders Center, 450 Brookline Ave., 02215, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Via Olgettina 48, 20156, Milan, Italy.
| |
Collapse
|
78
|
Henrich-Noack P, Nikitovic D, Neagu M, Docea AO, Engin AB, Gelperina S, Shtilman M, Mitsias P, Tzanakakis G, Gozes I, Tsatsakis A. The blood–brain barrier and beyond: Nano-based neuropharmacology and the role of extracellular matrix. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:359-379. [DOI: 10.1016/j.nano.2019.01.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/11/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
|
79
|
Nanomaterials for Drug Delivery to the Central Nervous System. NANOMATERIALS 2019; 9:nano9030371. [PMID: 30841578 PMCID: PMC6474019 DOI: 10.3390/nano9030371] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 12/25/2022]
Abstract
The intricate microstructure of the blood-brain barrier (BBB) is responsible for the accurate intrinsic regulation of the central nervous system (CNS), in terms of neuronal pathophysiological phenomena. Any disruption to the BBB can be associated with genetic defects triggering or with local antigenic invasion (either neurotoxic blood-derived metabolites and residues or microbial pathogens). Such events can be further related to systemic inflammatory or immune disorders, which can subsequently initiate several neurodegenerative pathways. Any degenerative process related to the CNS results in progressive and yet incurable impairment of neuronal cells. Since these particular neurons are mostly scanty or incapable of self-repair and regeneration processes, there is tremendous worldwide interest in novel therapeutic strategies for such specific conditions. Alzheimer’s and Parkinson’s diseases (AD and PD, respectively) are conditions found worldwide, being considered the most rampant degenerative pathologies related to CNS. The current therapy of these conditions, including both clinical and experimental approaches, mainly enables symptom management and subsidiary neuronal protection and even less disease regression. Still, a thorough understanding of the BBB pathophysiology and an accurate molecular and sub-molecular management of AD and PD will provide beneficial support for more specific and selective therapy. Since nanotechnology-derived materials and devices proved attractive and efficient platforms for modern biomedicine (including detection, imaging, diagnosis, medication, restoration and regeneration), a particular approach for AD and PD management relies on nanoparticle-based therapy. In this paper we will discuss relevant aspects related to the BBB and its impact on drug-based treatment and emphasize that nanoparticles are suitable and versatile candidates for the development of novel and performance-enhanced nanopharmaceuticals for neurodegenerative conditions therapy.
Collapse
|
80
|
Louwies T, Ligon CO, Johnson AC, Greenwood-Van Meerveld B. Targeting epigenetic mechanisms for chronic visceral pain: A valid approach for the development of novel therapeutics. Neurogastroenterol Motil 2019; 31:e13500. [PMID: 30393913 PMCID: PMC7924309 DOI: 10.1111/nmo.13500] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/21/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Chronic visceral pain is persistent pain emanating from thoracic, pelvic, or abdominal origin that is poorly localized with regard to the specific organ affected. The prevalence can range up to 25% in the adult population as chronic visceral pain is a common feature of many visceral disorders, which may or may not be accompanied by distinct structural or histological abnormalities within the visceral organs. Mounting evidence suggests that changes in epigenetic mechanisms are involved in the top-down or bottom-up sensitization of pain pathways and the development of chronic pain. Epigenetic changes can lead to long-term alterations in gene expression profiles of neurons and consequently alter functionality of peripheral neurons, dorsal root ganglia, spinal cord, and brain neurons. However, epigenetic modifications are dynamic, and thus, detrimental changes may be reversible. Hence, external factors/therapeutic interventions may be capable of modulating the epigenome and restore normal gene expression for extended periods of time. PURPOSE The goal of this review is to highlight the latest discoveries made toward understanding the epigenetic mechanisms that are involved in the development or maintenance of chronic visceral pain. Furthermore, this review will provide evidence supporting that targeting these epigenetic mechanisms may represent a novel approach to treat chronic visceral pain.
Collapse
Affiliation(s)
- Tijs Louwies
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Casey O. Ligon
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Beverley Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| |
Collapse
|
81
|
Israel LL, Braubach O, Galstyan A, Chiechi A, Shatalova ES, Grodzinski Z, Ding H, Black KL, Ljubimova JY, Holler E. A Combination of Tri-Leucine and Angiopep-2 Drives a Polyanionic Polymalic Acid Nanodrug Platform Across the Blood-Brain Barrier. ACS NANO 2019; 13:1253-1271. [PMID: 30633492 PMCID: PMC7641102 DOI: 10.1021/acsnano.8b06437] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
One of the major problems facing the treatment of neurological disorders is the poor delivery of therapeutic agents into the brain. Our goal is to develop a multifunctional and biodegradable nanodrug delivery system that crosses the blood-brain barrier (BBB) to access brain tissues affected by neurological disease. In this study, we synthesized a biodegradable nontoxic β-poly(l-malic acid) (PMLA or P) as a scaffold to chemically bind the BBB crossing peptides Angiopep-2 (AP2), MiniAp-4 (M4), and the transferrin receptor ligands cTfRL and B6. In addition, a trileucine endosome escape unit (LLL) and a fluorescent marker (rhodamine or rh) were attached to the PMLA backbone. The pharmacokinetics, BBB penetration, and biodistribution of nanoconjugates were studied in different brain regions and at multiple time points via optical imaging. The optimal nanoconjugate, P/LLL/AP2/rh, produced significant fluorescence in the parenchyma of cortical layers II/III, the midbrain colliculi, and the hippocampal CA1-3 cellular layers 30 min after a single intravenous injection; clearance was observed after 4 h. The nanoconjugate variant P/LLL/rh lacking AP2, or the variant P/AP2/rh lacking LLL, showed significantly less BBB penetration. The LLL moiety appeared to stabilize the nanoconjugate, while AP2 enhanced BBB penetration. Finally, nanoconjugates containing the peptides M4, cTfRL, and B6 displayed comparably little and/or inconsistent infiltration of brain parenchyma, likely due to reduced trans-BBB movement. P/LLL/AP2/rh can now be functionalized with intra-brain targeting and drug treatment moieties that are aimed at molecular pathways implicated in neurological disorders.
Collapse
Affiliation(s)
- Liron L. Israel
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| | - Oliver Braubach
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| | - Anna Galstyan
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| | - Antonella Chiechi
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| | - Ekaterina S. Shatalova
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| | - Zachary Grodzinski
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| | - Hui Ding
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| | - Keith L. Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, United States
| | - Julia Y. Ljubimova
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| | - Eggehard Holler
- Nanomedicine Research Center, Department of Neurosurgery, Los Angeles, California 90048, United States
| |
Collapse
|
82
|
Guan J, Jiang Z, Wang M, Liu Y, Liu J, Yang Y, Ding T, Lu W, Gao C, Qian J, Zhan C. Short Peptide-Mediated Brain-Targeted Drug Delivery with Enhanced Immunocompatibility. Mol Pharm 2019; 16:907-913. [PMID: 30666875 DOI: 10.1021/acs.molpharmaceut.8b01216] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Peptide ligands have been exploited as versatile tools to facilitate targeted delivery of nanocarriers. However, the effects of peptide ligands on immunocompatibility and therapeutic efficacy of liposomes remain intricate. Here, a short and stable brain targeted peptide ligand D8 was modified on the surface of doxorubicin-loaded liposomes (D8-sLip/DOX), demonstrating prolonged blood circulation and lower liver distribution in comparison to the long and stable D-peptide ligand DCDX-modified doxorubicin-loaded liposomes (DCDX-sLip/DOX) by mitigating natural IgM absorption. Despite the improved pharmacokinetic profiles, D8-sLip/DOX exhibited comparable brain targeting capacity in ICR mice and antiglioblastoma efficacy to DCDX-sLip/DOX in nude mice bearing intracranial glioblastoma. However, dramatic accumulation of DCDX-sLip/DOX in liver (especially during the first 8 h after intravenous injection) resulted in pathological symptoms, including nuclei swelling, necrosis of liver cells, and inflammation. These results suggest that short peptide ligand-mediated brain-targeted drug delivery systems possessing enhanced immunocompatibility are promising to facilitate efficient brain transport with improved biosafety.
Collapse
Affiliation(s)
- Juan Guan
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers , Fudan University , Shanghai 200032 , P.R. China
| | - Zhuxuan Jiang
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers , Fudan University , Shanghai 200032 , P.R. China
| | - Mengke Wang
- School of Pharmacy, Ministry of Education, Key Laboratory of Smart Drug Delivery , Fudan University , Shanghai 201203 , P.R. China
| | - Ying Liu
- Department of Pathology, School of Basic Medical Sciences , Fudan University , Shanghai 200032 , P.R. China
| | - Jican Liu
- Department of Pathology, Affiliated Zhongshan Hospital Qingpu Branch , Fudan University , Shanghai 201700 , P.R. China
| | - Yang Yang
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers , Fudan University , Shanghai 200032 , P.R. China
| | - Tianhao Ding
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers , Fudan University , Shanghai 200032 , P.R. China
| | - Weiyue Lu
- School of Pharmacy, Ministry of Education, Key Laboratory of Smart Drug Delivery , Fudan University , Shanghai 201203 , P.R. China
| | - Chunli Gao
- Department of Otolaryngology-Head and Neck Surgery, Eye and ENT Hospital , Fudan University , Shanghai 200032 , P.R. China
| | - Jun Qian
- School of Pharmacy, Ministry of Education, Key Laboratory of Smart Drug Delivery , Fudan University , Shanghai 201203 , P.R. China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers , Fudan University , Shanghai 200032 , P.R. China.,School of Pharmacy, Ministry of Education, Key Laboratory of Smart Drug Delivery , Fudan University , Shanghai 201203 , P.R. China
| |
Collapse
|
83
|
De Jong E, Williams DS, Abdelmohsen LK, Van Hest JC, Zuhorn IS. A filter-free blood-brain barrier model to quantitatively study transendothelial delivery of nanoparticles by fluorescence spectroscopy. J Control Release 2018; 289:14-22. [DOI: 10.1016/j.jconrel.2018.09.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/15/2018] [Accepted: 09/18/2018] [Indexed: 01/21/2023]
|
84
|
Francia V, Aliyandi A, Salvati A. Effect of the development of a cell barrier on nanoparticle uptake in endothelial cells. NANOSCALE 2018; 10:16645-16656. [PMID: 30155550 DOI: 10.1039/c8nr03171a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
In order to improve the current success of nanomedicine, a better understanding of how nano-sized materials interact with and are processed by cells is required. Typical in vitro nanoparticle-cell interaction studies often make use of cells cultured at different cell densities. However, in vivo, for their successful delivery to the target tissue, nanomedicines need to overcome several barriers, such as endothelial and epithelial cell barriers. Unlike sub-confluent or confluent cell cultures, cell barriers are tight cell monolayers, expressing a series of specialized tight junction proteins between adjacent cells to limit paracellular transport and ensure close cell-to-cell interactions. A clear understanding on how the development of cells into a cell barrier may affect the uptake of nano-sized drug carriers is still missing. To this aim, here, human primary umbilical vein endothelial cells (HUVEC) are used as a model cell line to form endothelial cell barriers. Then, nanoparticle uptake is assessed in the developed endothelial barriers and compared to the uptake in sub-confluent or confluent HUVEC cultures. The results clearly show that the organization of cells into a cell barrier leads to a differential gene expression of endocytic markers, and - interestingly - this is accompanied by reduced nanoparticle uptake levels. Transport inhibitors are used to characterise the mechanisms involved in the uptake. However, we show that some of them can strongly compromise barrier integrity, thus impairing the interpretation of the outcomes, and overall, only a partial inhibition of nanoparticle uptake could be obtained.
Collapse
Affiliation(s)
- Valentina Francia
- Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | | | | |
Collapse
|
85
|
Tamba B, Streinu V, Foltea G, Neagu A, Dodi G, Zlei M, Tijani A, Stefanescu C. Tailored surface silica nanoparticles for blood-brain barrier penetration: Preparation and in vivo investigation. ARAB J CHEM 2018. [DOI: 10.1016/j.arabjc.2018.03.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
86
|
Soares TB, Loureiro L, Carvalho A, Oliveira MECR, Dias A, Sarmento B, Lúcio M. Lipid nanocarriers loaded with natural compounds: Potential new therapies for age related neurodegenerative diseases? Prog Neurobiol 2018; 168:21-41. [DOI: 10.1016/j.pneurobio.2018.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 04/05/2018] [Indexed: 12/28/2022]
|
87
|
Abstract
Delivery of imaging agents and pharmaceutical payloads to the central nervous system (CNS) is essential for efficient diagnosis and treatment of brain diseases. However, therapeutic delivery is often restricted by the blood-brain barrier (BBB), which prevents transport of clinical compounds to their region of interest. This review discusses the methods that have been used to avoid or overcome this barrier, presenting the use of biologically-derived nanomaterial systems as an efficient strategy for the diagnosis and treatment of CNS diseases. Biological nanomaterials have many advantages over synthetic systems, including being biodegradable, biocompatible, easily surface functionalised for conjugation of targeting moieties, and are often able to self-assemble. These abilities are discussed in relation to various systems, including liposomes, dendrimers, and viral nanoparticles.
Collapse
|
88
|
Nabi B, Rehman S, Khan S, Baboota S, Ali J. Ligand conjugation: An emerging platform for enhanced brain drug delivery. Brain Res Bull 2018; 142:384-393. [DOI: 10.1016/j.brainresbull.2018.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/06/2018] [Accepted: 08/02/2018] [Indexed: 10/28/2022]
|
89
|
Sönmez M, Ficai D, Ficai A, Alexandrescu L, Georgescu M, Trusca R, Gurau D, Titu MA, Andronescu E. Applications of mesoporous silica in biosensing and controlled release of insulin. Int J Pharm 2018; 549:179-200. [PMID: 30016674 DOI: 10.1016/j.ijpharm.2018.07.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 01/22/2023]
Abstract
The development of new oral insulin delivery systems could bring significant benefits to insulin-dependent patients due to the simplicity of the method, avoidance of pain caused by parenteral administration and maintenance of optimal therapeutic levels for a longer period. However, administration of such therapeutic proteins orally remains a challenge because insulin (Ins) is a very sensitive molecule and can be easily degraded under the existing pH conditions in the stomach and intestines. Moreover, due to the large size of insulin, intestinal epithelium permeability is very low. This could be improved by immobilizing insulin in the mesoporous silica pores (MSN), acting as a shield to protect the molecule integrity from the proteolytic degradation existing in the stomach and upper part of the small intestine. Due to the high adsorption capacity of insulin, biocompatibility, ease of functionalization with various organic and/or inorganic groups, high mechanical and chemical resistance, adjustable pore size and volume, MSN is considered an ideal candidate for the development of controlled release systems that are sensitive to various stimuli (pH, temperature) as well as to glucose. Modifying MSN surfaces by coating with various mucoadhesive polymers (chitosan, alginate, etc.) will also facilitate interaction with the intestinal mucus and improve intestinal retention time. Moreover, the development of glucose-responsive systems for achieving MSN-based self-regulated insulin delivery, decorated with various components serving as sensors - glucose oxidase (GODx) and phenylboronic acid (PBA) that can control the insulin dosage, avoiding overdose leading to serious hypoglycemia. MSN have also been tested for application as biosensors for glucose monitoring.
Collapse
Affiliation(s)
- Maria Sönmez
- Research Institute of the University of Bucharest, 36-46 bd. M. Kogalniceanu, Bucharest, Romania
| | - Denisa Ficai
- Politehnica University of Bucharest, Faculty of Applied Chemistry and Material Science, 1-7 Polizu St., Bucharest, Romania
| | - Anton Ficai
- S.C. Metav R&D S.A, 31 C.A. Rosetti Str., Bucharest, Romania
| | - Laurentia Alexandrescu
- National Research & Development Institute for Textiles and Leather-Division: Leather and Footwear Research Institute, 93 Ion Minulescu St., Bucharest, Romania
| | - Mihai Georgescu
- National Research & Development Institute for Textiles and Leather-Division: Leather and Footwear Research Institute, 93 Ion Minulescu St., Bucharest, Romania
| | - Roxana Trusca
- S.C. Metav R&D S.A, 31 C.A. Rosetti Str., Bucharest, Romania
| | - Dana Gurau
- National Research & Development Institute for Textiles and Leather-Division: Leather and Footwear Research Institute, 93 Ion Minulescu St., Bucharest, Romania
| | | | - Ecaterina Andronescu
- Politehnica University of Bucharest, Faculty of Applied Chemistry and Material Science, 1-7 Polizu St., Bucharest, Romania.
| |
Collapse
|
90
|
Hajal C, Campisi M, Mattu C, Chiono V, Kamm RD. In vitro models of molecular and nano-particle transport across the blood-brain barrier. BIOMICROFLUIDICS 2018; 12:042213. [PMID: 29887937 PMCID: PMC5980570 DOI: 10.1063/1.5027118] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/09/2018] [Indexed: 05/11/2023]
Abstract
The blood-brain barrier (BBB) is the tightest endothelial barrier in humans. Characterized by the presence of tight endothelial junctions and adherens junctions, the primary function of the BBB is to maintain brain homeostasis through the control of solute transit across the barrier. The specific features of this barrier make for unique modes of transport of solutes, nanoparticles, and cells across the BBB. Understanding the different routes of traffic adopted by each of these is therefore critical in the development of targeted therapies. In an attempt to move towards controlled experimental assays, multiple groups are now opting for the use of microfluidic systems. A comprehensive understanding of bio-transport processes across the BBB in microfluidic devices is therefore necessary to develop targeted and efficient therapies for a host of diseases ranging from neurological disorders to the spread of metastases in the brain.
Collapse
Affiliation(s)
- Cynthia Hajal
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, MIT Building, Room NE47-321, Cambridge, Massachusetts 02139, USA
| | | | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Roger D. Kamm
- Author to whom correspondence should be addressed: and
| |
Collapse
|
91
|
Enhanced efficacy of combined temozolomide and bromodomain inhibitor therapy for gliomas using targeted nanoparticles. Nat Commun 2018; 9:1991. [PMID: 29777137 PMCID: PMC5959860 DOI: 10.1038/s41467-018-04315-4] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 04/11/2018] [Indexed: 12/15/2022] Open
Abstract
Effective treatment for glioblastoma (GBM) is limited by the presence of the blood–brain barrier (BBB) and rapid resistance to single agent therapies. To address these issues, we developed a transferrin-functionalized nanoparticle (Tf-NP) that can deliver dual combination therapies. Using intravital imaging, we show the ability of Tf-NPs to traverse intact BBB in mice as well as achieve direct tumor binding in two intracranial orthotopic models of GBM. Treatment of tumor-bearing mice with Tf-NPs loaded with temozolomide and the bromodomain inhibitor JQ1 leads to increased DNA damage and apoptosis that correlates with a 1.5- to 2-fold decrease in tumor burden and corresponding increase in survival compared to equivalent free-drug dosing. Immunocompetent mice treated with Tf-NP-loaded drugs also show protection from the effects of systemic drug toxicity, demonstrating the preclinical potential of this nanoscale platform to deliver novel combination therapies to gliomas and other central nervous system tumors. The blood-brain barrier often limits effective delivery of treatments for glioblastoma . In this study, the authors develop transferrin-functionalized nanoparticles able to traverse the intact blood-brain barrier and deliver combination temozolomide and bromodomain inhibitor therapy to glioma-bearing mice.
Collapse
|
92
|
Wang J, Garancher A, Ramaswamy V, Wechsler-Reya RJ. Medulloblastoma: From Molecular Subgroups to Molecular Targeted Therapies. Annu Rev Neurosci 2018; 41:207-232. [PMID: 29641939 DOI: 10.1146/annurev-neuro-070815-013838] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Brain tumors are the leading cause of cancer-related death in children, and medulloblastoma (MB) is the most common malignant pediatric brain tumor. Advances in surgery, radiation, and chemotherapy have improved the survival of MB patients. But despite these advances, 25-30% of patients still die from the disease, and survivors suffer severe long-term side effects from the aggressive therapies they receive. Although MB is often considered a single disease, molecular profiling has revealed a significant degree of heterogeneity, and there is a growing consensus that MB consists of multiple subgroups with distinct driver mutations, cells of origin, and prognosis. Here, we review recent progress in MB research, with a focus on the genes and pathways that drive tumorigenesis, the animal models that have been developed to study tumor biology, and the advances in conventional and targeted therapy.
Collapse
Affiliation(s)
- Jun Wang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| | - Alexandra Garancher
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| | - Vijay Ramaswamy
- Division of Haematology/Oncology and Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| |
Collapse
|
93
|
Erickson MA, Banks WA. Neuroimmune Axes of the Blood-Brain Barriers and Blood-Brain Interfaces: Bases for Physiological Regulation, Disease States, and Pharmacological Interventions. Pharmacol Rev 2018; 70:278-314. [PMID: 29496890 PMCID: PMC5833009 DOI: 10.1124/pr.117.014647] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Central nervous system (CNS) barriers predominantly mediate the immune-privileged status of the brain, and are also important regulators of neuroimmune communication. It is increasingly appreciated that communication between the brain and immune system contributes to physiologic processes, adaptive responses, and disease states. In this review, we discuss the highly specialized features of brain barriers that regulate neuroimmune communication in health and disease. In section I, we discuss the concept of immune privilege, provide working definitions of brain barriers, and outline the historical work that contributed to the understanding of CNS barrier functions. In section II, we discuss the unique anatomic, cellular, and molecular characteristics of the vascular blood-brain barrier (BBB), blood-cerebrospinal fluid barrier, and tanycytic barriers that confer their functions as neuroimmune interfaces. In section III, we consider BBB-mediated neuroimmune functions and interactions categorized as five neuroimmune axes: disruption, responses to immune stimuli, uptake and transport of immunoactive substances, immune cell trafficking, and secretions of immunoactive substances. In section IV, we discuss neuroimmune functions of CNS barriers in physiologic and disease states, as well as pharmacological interventions for CNS diseases. Throughout this review, we highlight many recent advances that have contributed to the modern understanding of CNS barriers and their interface functions.
Collapse
Affiliation(s)
- Michelle A Erickson
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - William A Banks
- Geriatric Research and Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington; and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
94
|
Leong J, Teo JY, Aakalu VK, Yang YY, Kong H. Engineering Polymersomes for Diagnostics and Therapy. Adv Healthc Mater 2018; 7:e1701276. [PMID: 29334183 PMCID: PMC6377267 DOI: 10.1002/adhm.201701276] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 11/30/2017] [Indexed: 12/20/2022]
Abstract
Engineered polymer vesicles, termed as polymersomes, confer a flexibility to control their structure, properties, and functionality. Self-assembly of amphiphilic copolymers leads to vesicles consisting of a hydrophobic bilayer membrane and hydrophilic core, each of which is loaded with a wide array of small and large molecules of interests. As such, polymersomes are increasingly being studied as carriers of imaging probes and therapeutic drugs. Effective delivery of polymersomes necessitates careful design of polymersomes. Therefore, this review article discusses the design strategies of polymersomes developed for enhanced transport and efficacy of imaging probes and therapeutic drugs. In particular, the article focuses on overviewing technologies to regulate the size, structure, shape, surface activity, and stimuli- responsiveness of polymersomes and discussing the extent to which these properties and structure of polymersomes influence the efficacy of cargo molecules. Taken together with future considerations, this article will serve to improve the controllability of polymersome functions and accelerate the use of polymersomes in biomedical applications.
Collapse
Affiliation(s)
- Jiayu Leong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA, Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Jye Yng Teo
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA, Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Vinay K. Aakalu
- Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Department of Ophthalmology and Visual Sciences, Chicago, IL 60612, USA
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, Department of Bioengineering, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA,
| |
Collapse
|
95
|
He Q, Liu J, Liang J, Liu X, Li W, Liu Z, Ding Z, Tuo D. Towards Improvements for Penetrating the Blood-Brain Barrier-Recent Progress from a Material and Pharmaceutical Perspective. Cells 2018; 7:cells7040024. [PMID: 29570659 PMCID: PMC5946101 DOI: 10.3390/cells7040024] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/18/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
The blood–brain barrier (BBB) is a critical biological structure that prevents damage to the brain and maintains its bathing microenvironment. However, this barrier is also the obstacle to deliver beneficial drugs to treat CNS (central nervous system) diseases. Many efforts have been made for improvement of delivering drugs across the BBB in recent years to treat CNS diseases. In this review, the anatomical and functional structure of the BBB is comprehensively discussed. The mechanisms of BBB penetration are summarized, and the methods and effects on increasing BBB permeability are investigated in detail. It also elaborates on the physical, chemical, biological and nanocarrier aspects to improve drug delivery penetration to the brain and introduces some specific drug delivery effects on BBB permeability.
Collapse
Affiliation(s)
- Quanguo He
- School of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, China.
| | - Jun Liu
- School of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, China.
| | - Jing Liang
- School of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, China.
| | - Xiaopeng Liu
- School of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, China.
| | - Wen Li
- School of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, China.
| | - Zhi Liu
- School of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, China.
| | - Ziyu Ding
- School of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, China.
| | - Du Tuo
- School of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, China.
| |
Collapse
|
96
|
Young AT, Cornwell N, Daniele MA. Neuro-Nano Interfaces: Utilizing Nano-Coatings and Nanoparticles to Enable Next-Generation Electrophysiological Recording, Neural Stimulation, and Biochemical Modulation. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1700239. [PMID: 33867903 PMCID: PMC8049593 DOI: 10.1002/adfm.201700239] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Neural interfaces provide a window into the workings of the nervous system-enabling both biosignal recording and modulation. Traditionally, neural interfaces have been restricted to implanted electrodes to record or modulate electrical activity of the nervous system. Although these electrode systems are both mechanically and operationally robust, they have limited utility due to the resultant macroscale damage from invasive implantation. For this reason, novel nanomaterials are being investigated to enable new strategies to chronically interact with the nervous system at both the cellular and network level. In this feature article, the use of nanomaterials to improve current electrophysiological interfaces, as well as enable new nano-interfaces to modulate neural activity via alternative mechanisms, such as remote transduction of electromagnetic fields are explored. Specifically, this article will review the current use of nanoparticle coatings to enhance electrode function, then an analysis of the cutting-edge, targeted nanoparticle technologies being utilized to interface with both the electrophysiological and biochemical behavior of the nervous system will be provided. Furthermore, an emerging, specialized-use case for neural interfaces will be presented: the modulation of the blood-brain barrier.
Collapse
Affiliation(s)
- Ashlyn T Young
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Neil Cornwell
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, 911 Oval Dr., Raleigh, NC 27695, USA
| |
Collapse
|
97
|
Mullis AS, Schlichtmann BW, Narasimhan B, Cademartiri R, Mallapragada SK. Ligand-cascading nano-delivery devices to enable multiscale targeting of anti-neurodegenerative therapeutics. Biomed Mater 2018; 13:034102. [DOI: 10.1088/1748-605x/aaa778] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
98
|
Dong X. Current Strategies for Brain Drug Delivery. Am J Cancer Res 2018; 8:1481-1493. [PMID: 29556336 PMCID: PMC5858162 DOI: 10.7150/thno.21254] [Citation(s) in RCA: 558] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023] Open
Abstract
The blood-brain barrier (BBB) has been a great hurdle for brain drug delivery. The BBB in healthy brain is a diffusion barrier essential for protecting normal brain function by impeding most compounds from transiting from the blood to the brain; only small molecules can cross the BBB. Under certain pathological conditions of diseases such as stroke, diabetes, seizures, multiple sclerosis, Parkinson's disease and Alzheimer disease, the BBB is disrupted. The objective of this review is to provide a broad overview on current strategies for brain drug delivery and related subjects from the past five years. It is hoped that this review could inspire readers to discover possible approaches to deliver drugs into the brain. After an initial overview of the BBB structure and function in both healthy and pathological conditions, this review re-visits, according to recent publications, some questions that are controversial, such as whether nanoparticles by themselves could cross the BBB and whether drugs are specifically transferred to the brain by actively targeted nanoparticles. Current non-nanoparticle strategies are also reviewed, such as delivery of drugs through the permeable BBB under pathological conditions and using non-invasive techniques to enhance brain drug uptake. Finally, one particular area that is often neglected in brain drug delivery is the influence of aging on the BBB, which is captured in this review based on the limited studies in the literature.
Collapse
|
99
|
Targeting the leptin receptor: To evaluate therapeutic efficacy and anti-tumor effects of Doxil, in vitro and in vivo in mice bearing C26 colon carcinoma tumor. Colloids Surf B Biointerfaces 2018; 164:107-115. [PMID: 29413587 DOI: 10.1016/j.colsurfb.2018.01.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 12/11/2022]
Abstract
Leptin is an appetite regulatory hormone that is secreted into the blood circulation by the adipose tissue and it functions via its over expressed receptors (Ob-R) in a wide variety of cancers. In the present study, the function of a leptin-derived peptide (LP16, 91-110 of Leptin) was investigated as a targeting ligand to decorate PEGylated liposomal doxorubicin (PLD, Doxil®) surface and the anti-tumor activity and therapeutic efficacy of Doxil in C26 (Colon Carcinoma) tumor model were also evaluated. As a result of this, Doxil with different LP16 peptide density (25, 50, 100 and 200 peptide on the surface of each liposome) was successfully prepared and characterized. In vitro results showed significant enhanced cytotoxicity and cellular binding and uptake of LP16-targeted Doxil formulations (LP16-Doxil) in C26 cells as compared to Doxil. In BALB/c mice bearing C26 murine carcinoma, at a dose of 15 mg/kg, LP16-Doxil groups (100 ligand) significantly suppressed the growth of the tumor and showed higher inclination to tumor as compared to non-targeted Doxil. This study revealed that the potential of LP16 peptide targeting increased the therapeutic efficacy of Doxil and highlighted the importance of optimizing the ligand density to maximize the targeting ability of the nanocarriers and merits further investigations.
Collapse
|
100
|
Pederzoli F, Tosi G, Genovese F, Belletti D, Vandelli MA, Ballestrazzi A, Forni F, Ruozi B. Qualitative and semiquantitative analysis of the protein coronas associated to different functionalized nanoparticles. Nanomedicine (Lond) 2018; 13:407-422. [PMID: 29345202 DOI: 10.2217/nnm-2017-0250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM The investigation on protein coronas (PCs) adsorbed onto nanoparticle (NP) surface is representing an open issue due to difficulties in detection and clear isolation of the adsorbed proteins. In this study, we investigated protocols able to isolate the compositions of PCs of three polymeric NPs. MATERIALS & METHODS Unfunctionalized NPs and two functionalized NPs were considered as proof-of-concept for the qualitative and semiquantitative analysis of both the corona levels (stably or weakly adsorbed coronas [SC/WC]) of these different nanocarriers. RESULTS The protocols applied were able to discriminate between the SC and WC. In particular, experimental results indicated that stably adsorbed coronas are prevalently composed by ApoE, while WC by albumin in all the NPs. Otherwise, some differences in WC could be correlated with surface functionalization. CONCLUSION This experimental approach allows characterizing the whole PCs, proposing a protocol for isolation of different types of proteins composing PCs.
Collapse
Affiliation(s)
- Francesca Pederzoli
- Department of Life Sciences, University of Modena & Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Giovanni Tosi
- Department of Life Sciences, University of Modena & Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Filippo Genovese
- Centro Interdipartimentale Grandi Strumenti, University of Modena & Reggio Emilia, via Campi 185, 41125 Modena, Italy
| | - Daniela Belletti
- Department of Life Sciences, University of Modena & Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Maria Angela Vandelli
- Department of Life Sciences, University of Modena & Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antonio Ballestrazzi
- Department of Scienze Fisiche, Informatiche e Matematiche, University of Modena & Reggio Emilia, Via Campi 213/a, 41125 Modena, Italy
| | - Flavio Forni
- Department of Life Sciences, University of Modena & Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Barbara Ruozi
- Department of Life Sciences, University of Modena & Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| |
Collapse
|