151
|
Samaridou E, Alonso MJ. Nose-to-brain peptide delivery - The potential of nanotechnology. Bioorg Med Chem 2017; 26:2888-2905. [PMID: 29170026 DOI: 10.1016/j.bmc.2017.11.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/26/2017] [Accepted: 11/02/2017] [Indexed: 12/11/2022]
Abstract
Nose-to-brain (N-to-B) delivery offers to protein and peptide drugs the possibility to reach the brain in a non-invasive way. This article is a comprehensive review of the state-of-the-art of this emerging peptide delivery route, as well as of the challenges associated to it. Emphasis is given on the potential of nanosized drug delivery carriers to enhance the direct N-to-B transport of protein or peptide drugs. In particular, polymer- and lipid- based nanocarriers are comparatively analyzed in terms of the influence of their physicochemical characteristics and composition on their in vivo fate and efficacy. The use of biorecognitive ligands and permeation enhancers in order to enhance their brain targeting efficiency is also discussed. The article concludes highlighting the early stage of this research field and its still unveiled potential. The final message is that more explicatory PK/PD studies are required in order to achieve the translation from preclinical to the clinical development phase.
Collapse
Affiliation(s)
- Eleni Samaridou
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Maria José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
152
|
Lehocky CA, Fellows-Mayle W, Engh JA, Riviere CN. Tip Design for Safety of Steerable Needles for Robot-Controlled Brain Insertion. ROBOTIC SURGERY : RESEARCH AND REVIEWS 2017; 4:107-114. [PMID: 29170740 PMCID: PMC5695876 DOI: 10.2147/rsrr.s141085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Current practice in neurosurgical needle insertion is limited by the straight trajectories inherent with rigid probes. One technique allowing curvilinear trajectories involves flexible bevel-tipped needles, which bend during insertion due to their asymmetry. In the brain, safety will require avoidance of the sharp tips often used in laboratory studies, in favor of a more rounded profile. Steering performance, on the other hand, requires maximal asymmetry. Design of safe bevel-tipped brain needles thus involves management of this tradeoff by adjusting needle gauge, bevel angle, and fillet (or tip) radius to arrive at a design that is suitably asymmetrical while producing strain, strain rate, and stress below the levels that would damage brain tissue. Methods Designs with a variety of values of needle radius, bevel angle, and fillet radius were evaluated in finite-element simulations of simultaneous insertion and rotation. Brain tissue was modeled as a hyperelastic, linear viscoelastic material. Based on the literature available, safety thresholds of 0.19 strain, 10 s-1 strain rate, and 120 kPa stress were used. Safe values of needle radius, bevel angle, and fillet radius were selected, along with an appropriate velocity envelope for safe operation. The resulting needle was fabricated and compared with a Sedan side-cutting brain biopsy needle in a study in the porcine model in vivo (N=3). Results The prototype needle selected was 1.66 mm in diameter, with bevel angle of 10° and fillet radius of 0.25 mm. Upon examination of postoperative CT and histological images, no differences in tissue trauma or hemorrhage were noted between the prototype needle and the Sedan needle. Conclusions The study indicates a general design technique for safe bevel-tipped brain needles based on comparison with relevant damage thresholds for strain, strain rate, and stress. The full potential of the technique awaits the determination of more exact safety thresholds.
Collapse
Affiliation(s)
- Craig A Lehocky
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Wendy Fellows-Mayle
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Johnathan A Engh
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Cameron N Riviere
- The Robotics Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
153
|
Urban A, Golgher L, Brunner C, Gdalyahu A, Har-Gil H, Kain D, Montaldo G, Sironi L, Blinder P. Understanding the neurovascular unit at multiple scales: Advantages and limitations of multi-photon and functional ultrasound imaging. Adv Drug Deliv Rev 2017; 119:73-100. [PMID: 28778714 DOI: 10.1016/j.addr.2017.07.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/17/2017] [Accepted: 07/22/2017] [Indexed: 02/07/2023]
Abstract
Developing efficient brain imaging technologies by combining a high spatiotemporal resolution and a large penetration depth is a key step for better understanding the neurovascular interface that emerges as a main pathway to neurodegeneration in many pathologies such as dementia. This review focuses on the advances in two complementary techniques: multi-photon laser scanning microscopy (MPLSM) and functional ultrasound imaging (fUSi). MPLSM has become the gold standard for in vivo imaging of cellular dynamics and morphology, together with cerebral blood flow. fUSi is an innovative imaging modality based on Doppler ultrasound, capable of recording vascular brain activity over large scales (i.e., tens of cubic millimeters) at unprecedented spatial and temporal resolution for such volumes (up to 10μm pixel size at 10kHz). By merging these two technologies, researchers may have access to a more detailed view of the various processes taking place at the neurovascular interface. MPLSM and fUSi are also good candidates for addressing the major challenge of real-time delivery, monitoring, and in vivo evaluation of drugs in neuronal tissue.
Collapse
Affiliation(s)
- Alan Urban
- Neuroelectronics Research Flanders, Leuven, Belgium; VIB, Leuven, Belgium and/or IMEC, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium; Neurobiology Dept., Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Lior Golgher
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Clément Brunner
- Neuroelectronics Research Flanders, Leuven, Belgium; VIB, Leuven, Belgium and/or IMEC, Leuven, Belgium
| | - Amos Gdalyahu
- Neurobiology Dept., Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Hagai Har-Gil
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - David Kain
- Neurobiology Dept., Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Gabriel Montaldo
- Neuroelectronics Research Flanders, Leuven, Belgium; VIB, Leuven, Belgium and/or IMEC, Leuven, Belgium
| | - Laura Sironi
- Physics Dept., Universita degli Studi di Milano Bicocca, Italy
| | - Pablo Blinder
- Neurobiology Dept., Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel; Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
154
|
Azria D, Blanquer S, Verdier JM, Belamie E. Nanoparticles as contrast agents for brain nuclear magnetic resonance imaging in Alzheimer's disease diagnosis. J Mater Chem B 2017; 5:7216-7237. [PMID: 32264173 DOI: 10.1039/c7tb01599b] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nuclear Magnetic Resonance Imaging (MRI) of amyloid plaques is a powerful non-invasive approach for the early and accurate diagnosis of Alzheimer's disease (AD) along with clinical observations of behavioral changes and cognitive impairment. The present article aims at giving a critical and comprehensive review of recent advances in the development of nanoparticle-based contrast agents for brain MRI. Nanoparticles considered for the MRI of AD must comply with a highly stringent set of requirements including low toxicity and the ability to cross the blood-brain-barrier. In addition, to reach an optimal signal-to-noise ratio, they must exhibit a specific ability to target amyloid plaques, which can be achieved by grafting antibodies, peptides or small molecules. Finally, we propose to consider new directions for the future of MRI in the context of Alzheimer's disease, in particular by enhancing the performances of contrast agents and by including therapeutic functionalities following a theranostic strategy.
Collapse
Affiliation(s)
- David Azria
- Institut Charles Gerhardt Montpellier, UMR 5253 CNRS-UM-ENSCM, Equipe Matériaux Avancés pour la Catalyse et la Santé, 8 rue de l'Ecole Normale, 34296 Montpellier Cedex 5, France.
| | | | | | | |
Collapse
|
155
|
Giuffrida MC, Pignatello R, Castelli F, Sarpietro MG. Amphiphilic naproxen prodrugs: differential scanning calorimetry study on their interaction with phospholipid bilayers†. J Pharm Pharmacol 2017. [DOI: 10.1111/jphp.12754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Abstract
Objectives
Naproxen, a nonsteroid anti-inflammatory drug studied for Alzheimer's disease, was conjugated with lipoamino acids (LAA) directly or through a diethylamine (EDA) spacer to improve the drug lipophilicity and the interaction with phospholipid bilayers.
Methods
The interaction of naproxen and its prodrugs with biomembrane models consisting of dimyristoylphosphatidylcholine multilamellar vesicles was studied by differential scanning calorimetry. The transfer of prodrugs from a lipophilic carrier to a biomembrane model was also studied.
Key findings
Naproxen conjugation to lipoamino acids improves its interaction with biomembrane models and affects the transfer from a lipophilic carrier to biomembrane model. LAA portion may localize between the phospholipid chains; the entity of the interaction depends not only on the presence of the spacer but also on the LAA chain length.
Conclusions
Variation of LAA portion can modulate the naproxen prodrugs affinity towards the biological membrane as well as towards the lipophilic carrier.
Collapse
Affiliation(s)
| | - Rosario Pignatello
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Catania, Italy
| | - Francesco Castelli
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Catania, Italy
| | | |
Collapse
|
156
|
Huang L, Hu J, Huang S, Wang B, Siaw-Debrah F, Nyanzu M, Zhang Y, Zhuge Q. Nanomaterial applications for neurological diseases and central nervous system injury. Prog Neurobiol 2017; 157:29-48. [PMID: 28743465 DOI: 10.1016/j.pneurobio.2017.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022]
Abstract
The effectiveness of noninvasive treatment for neurological disease is generally limited by the poor entry of therapeutic agents into the central nervous system (CNS). Most CNS drugs cannot permeate into the brain parenchyma because of the blood-brain barrier thus, overcoming this problem has become one of the most significant challenges in the development of neurological therapeutics. Nanotechnology has emerged as an innovative alternative for treating neurological diseases. In fact, rapid advances in nanotechnology have provided promising solutions to this challenge. This review highlights the applications of nanomaterials in the developing neurological field and discusses the evidence for their efficacies.
Collapse
Affiliation(s)
- Lijie Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Jiangnan Hu
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Shengwei Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Brian Wang
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Felix Siaw-Debrah
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Mark Nyanzu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Yu Zhang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Qichuan Zhuge
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China.
| |
Collapse
|
157
|
Te Brake LHM, de Knegt GJ, de Steenwinkel JE, van Dam TJP, Burger DM, Russel FGM, van Crevel R, Koenderink JB, Aarnoutse RE. The Role of Efflux Pumps in Tuberculosis Treatment and Their Promise as a Target in Drug Development: Unraveling the Black Box. Annu Rev Pharmacol Toxicol 2017; 58:271-291. [PMID: 28715978 DOI: 10.1146/annurev-pharmtox-010617-052438] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Insight into drug transport mechanisms is highly relevant to the efficacious treatment of tuberculosis (TB). Major problems in TB treatment are related to the transport of antituberculosis (anti-TB) drugs across human and mycobacterial membranes, affecting the concentrations of these drugs systemically and locally. Firstly, transporters located in the intestines, liver, and kidneys all determine the pharmacokinetics and pharmacodynamics of anti-TB drugs, with a high risk of drug-drug interactions in the setting of concurrent use of antimycobacterial, antiretroviral, and antidiabetic agents. Secondly, human efflux transporters limit the penetration of anti-TB drugs into the brain and cerebrospinal fluid, which is especially important in the treatment of TB meningitis. Finally, efflux transporters located in the macrophage and Mycobacterium tuberculosis cell membranes play a pivotal role in the emergence of phenotypic tolerance and drug resistance, respectively. We review the role of efflux transporters in TB drug disposition and evaluate the promise of efflux pump inhibition from a novel holistic perspective.
Collapse
Affiliation(s)
- Lindsey H M Te Brake
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; .,Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Gerjo J de Knegt
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Jurriaan E de Steenwinkel
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Teunis J P van Dam
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jan B Koenderink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Rob E Aarnoutse
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| |
Collapse
|
158
|
Guerra M, Blázquez JL, Rodríguez EM. Blood-brain barrier and foetal-onset hydrocephalus, with a view on potential novel treatments beyond managing CSF flow. Fluids Barriers CNS 2017; 14:19. [PMID: 28701191 PMCID: PMC5508761 DOI: 10.1186/s12987-017-0067-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/24/2017] [Indexed: 12/12/2022] Open
Abstract
Despite decades of research, no compelling non-surgical therapies have been developed for foetal hydrocephalus. So far, most efforts have pointed to repairing disturbances in the cerebrospinal fluid (CSF) flow and to avoid further brain damage. There are no reports trying to prevent or diminish abnormalities in brain development which are inseparably associated with hydrocephalus. A key problem in the treatment of hydrocephalus is the blood–brain barrier that restricts the access to the brain for therapeutic compounds or systemically grafted cells. Recent investigations have started to open an avenue for the development of a cell therapy for foetal-onset hydrocephalus. Potential cells to be used for brain grafting include: (1) pluripotential neural stem cells; (2) mesenchymal stem cells; (3) genetically-engineered stem cells; (4) choroid plexus cells and (5) subcommissural organ cells. Expected outcomes are a proper microenvironment for the embryonic neurogenic niche and, consequent normal brain development.
Collapse
Affiliation(s)
- M Guerra
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.
| | - J L Blázquez
- Departamento de Anatomía e Histología Humana, Facultad de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - E M Rodríguez
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
159
|
Gribkoff VK, Kaczmarek LK. The need for new approaches in CNS drug discovery: Why drugs have failed, and what can be done to improve outcomes. Neuropharmacology 2017; 120:11-19. [PMID: 26979921 PMCID: PMC5820030 DOI: 10.1016/j.neuropharm.2016.03.021] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/14/2016] [Accepted: 03/11/2016] [Indexed: 12/31/2022]
Abstract
An important goal of biomedical research is to translate basic research findings into useful medical advances. In the field of neuropharmacology this requires understanding disease mechanisms as well as the effects of drugs and other compounds on neuronal function. Our hope is that this information will result in new or improved treatment for CNS disease. Despite great progress in our understanding of the structure and functions of the CNS, the discovery of new drugs and their clinical development for many CNS disorders has been problematic. As a result, CNS drug discovery and development programs have been subjected to significant cutbacks and eliminations over the last decade. While there has been recent resurgence of interest in CNS targets, these past changes in priority of the pharmaceutical and biotech industries reflect several well-documented realities. CNS drugs in general have higher failure rates than non-CNS drugs, both preclinically and clinically, and in some areas, such as the major neurodegenerative diseases, the clinical failure rate for disease-modifying treatments has been 100%. The development times for CNS drugs are significantly longer for those drugs that are approved, and post-development regulatory review is longer. In this introduction we review some of the reasons for failure, delineating both scientific and technical realities, some unique to the CNS, that have contributed to this. We will focus on major neurodegenerative disorders, which affect millions, attract most of the headlines, and yet have witnessed the fewest successes. We will suggest some changes that, when coupled with the approaches discussed in the rest of this special volume, may improve outcomes in future CNS-targeted drug discovery and development efforts. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".
Collapse
Affiliation(s)
- Valentin K Gribkoff
- Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
160
|
Song Y, Du D, Li L, Xu J, Dutta P, Lin Y. In Vitro Study of Receptor-Mediated Silica Nanoparticles Delivery across Blood-Brain Barrier. ACS APPLIED MATERIALS & INTERFACES 2017; 9:20410-20416. [PMID: 28541655 PMCID: PMC5533093 DOI: 10.1021/acsami.7b03504] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
A brain drug delivery system has been demonstrated by attaching lactoferrin (Lf) on the silica nanoparticles (Si NPs). The nanoparticle surface was modified with polyethylene glycol to reduce protein adsorption. The transport efficiency of Lf attached Si NPs was studied using an in vitro blood-brain barrier (BBB) model consisting of three distinct types of cells: endocytes, pericytes, and astrocytes. Transfer of NPs from the apical side to the basolateral side is observed. The results indicated that Lf attached Si NPs demonstrated enhanced transport efficiency across the BBB with size-dependence compared to bare Si NPs. The maximum transport efficiency of lactoferrin conjugated silica nanoparticle was observed for 25 nm diameter particles. This receptor-mediated transcytosis of Si NPs across the cerebral endothelial cells can be employed to deliver drugs and imaging probes to the brain.
Collapse
Affiliation(s)
- Yang Song
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Dan Du
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Lei Li
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
| | - Jun Xu
- Department of Integrative Physiology and Neuroscience, Washington State University, 1815 Ferdinand’s Lane, Pullman, WA 99164, USA
| | - Prashanta Dutta
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
- Corresponding Author. .
| | - Yuehe Lin
- School of Mechanical and Materials Engineering, Washington State University, Pullman, Washington 99164, United States
- Corresponding Author. .
| |
Collapse
|
161
|
Blood-brain-barrier spheroids as an in vitro screening platform for brain-penetrating agents. Nat Commun 2017; 8:15623. [PMID: 28585535 PMCID: PMC5467173 DOI: 10.1038/ncomms15623] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/07/2017] [Indexed: 12/24/2022] Open
Abstract
Culture-based blood–brain barrier (BBB) models are crucial tools to enable rapid screening of brain-penetrating drugs. However, reproducibility of in vitro barrier properties and permeability remain as major challenges. Here, we report that self-assembling multicellular BBB spheroids display reproducible BBB features and functions. The spheroid core is comprised mainly of astrocytes, while brain endothelial cells and pericytes encase the surface, acting as a barrier that regulates transport of molecules. The spheroid surface exhibits high expression of tight junction proteins, VEGF-dependent permeability, efflux pump activity and receptor-mediated transcytosis of angiopep-2. In contrast, the transwell co-culture system displays comparatively low levels of BBB regulatory proteins, and is unable to discriminate between the transport of angiopep-2 and a control peptide. Finally, we have utilized the BBB spheroids to screen and identify BBB-penetrant cell-penetrating peptides (CPPs). This robust in vitro BBB model could serve as a valuable next-generation platform for expediting the development of CNS therapeutics. In vitro blood-brain barrier (BBB) models are crucial tools for screening brain-penetrating compounds. Here the authors develop a self-assembling BBB spheroid model with superior performance to the standard transwell BBB model, and use their platform to identify cell-penetrating peptides that can cross the BBB.
Collapse
|
162
|
Ziemba AM, Gilbert RJ. Biomaterials for Local, Controlled Drug Delivery to the Injured Spinal Cord. Front Pharmacol 2017; 8:245. [PMID: 28539887 PMCID: PMC5423911 DOI: 10.3389/fphar.2017.00245] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/19/2017] [Indexed: 11/13/2022] Open
Abstract
Affecting approximately 17,000 new people each year, spinal cord injury (SCI) is a devastating injury that leads to permanent paraplegia or tetraplegia. Current pharmacological approaches are limited in their ability to ameliorate this injury pathophysiology, as many are not delivered locally, for a sustained duration, or at the correct injury time point. With this review, we aim to communicate the importance of combinatorial biomaterial and pharmacological approaches that target certain aspects of the dynamically changing pathophysiology of SCI. After reviewing the pathophysiology timeline, we present experimental biomaterial approaches to provide local sustained doses of drug. In this review, we present studies using a variety of biomaterials, including hydrogels, particles, and fibers/conduits for drug delivery. Subsequently, we discuss how each may be manipulated to optimize drug release during a specific time frame following SCI. Developing polymer biomaterials that can effectively release drug to target specific aspects of SCI pathophysiology will result in more efficacious approaches leading to better regeneration and recovery following SCI.
Collapse
Affiliation(s)
| | - Ryan J. Gilbert
- Department of Biomedical Engineering and Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, TroyNY, USA
| |
Collapse
|
163
|
Casaca-Carreira J, Temel Y, Hescham SA, Jahanshahi A. Transependymal Cerebrospinal Fluid Flow: Opportunity for Drug Delivery? Mol Neurobiol 2017; 55:2780-2788. [PMID: 28455692 PMCID: PMC5842497 DOI: 10.1007/s12035-017-0501-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/07/2017] [Indexed: 12/23/2022]
Abstract
Drug delivery to the central nervous system (CNS) is complicated by the blood-brain barrier. As a result, many agents that are found to be potentially effective at their site of action cannot be sufficiently or effectively delivered to the CNS and therefore have been discarded and not developed further for clinical use, leaving many CNS diseases untreated. One way to overcome this obstacle is intracerebroventricular (ICV) delivery of the therapeutics directly to cerebrospinal fluid (CSF). Recent experimental and clinical findings reveal that CSF flows from the ventricles throughout the parenchyma towards the subarachnoid space also named minor CSF pathway, while earlier, it was suggested that only in pathological conditions such as hydrocephalus this form of CSF flow occurs. This transependymal flow of CSF provides a route to distribute ICV-infused drugs throughout the brain. More insight on transependymal CSF flow will direct more rational to ICV drug delivery and broaden its clinical indications in managing CNS diseases.
Collapse
Affiliation(s)
- João Casaca-Carreira
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- European Graduate School of Neuroscience (EURON), Maastricht, the Netherlands
| | - Yasin Temel
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
- European Graduate School of Neuroscience (EURON), Maastricht, the Netherlands
| | - Sarah-Anna Hescham
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
- European Graduate School of Neuroscience (EURON), Maastricht, the Netherlands
| | - Ali Jahanshahi
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.
- European Graduate School of Neuroscience (EURON), Maastricht, the Netherlands.
| |
Collapse
|
164
|
Yulug B, Hanoglu L, Kilic E. The neuroprotective effect of focused ultrasound: New perspectives on an old tool. Brain Res Bull 2017; 131:199-206. [PMID: 28458041 DOI: 10.1016/j.brainresbull.2017.04.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/24/2017] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Transcranial focused ultrasound (tFUS) is a novel technique that can noninvasively modulate the cortical function. Moreover, there are rapidly replicating evidence suggesting the role of tFUS for targeted neuroprotective drug delivery by increasing the permeability of the central nervous system barrier that results with increased neuroprotective activity. In contrast to the indirect neuroprotective effect, there is rare evidence suggesting the direct parenchymal neuroprotective effect of transcranial focused ultrasound (tFUS). In the light of these findings, we aimed to review the direct and indirect neuroprotective effect of FUS in various animal models of Stroke, Parkinson's Disease, Alzheimer's Disease and Major Depressive Disorder. METHODS A literary search was conducted, utilizing search terms "animal", "focused ultrasound", "neuroprotection", "Alzheimer's Disease", "Parkinson's Disease ", "Stroke", "Neurodegenerative disease" and "Major Depressive Disorder". Items were excluded if they failed to: (1) include patients, (2) editorials, and letters. RESULTS This mini-review article presents an up-to-date review of the neuroprotective effects of tFUS in animal studies and suggests the dual neurotherapeutic role of tFUS in various neurodegenerative diseases. CONCLUSION Future well-conducted human studies are emergently needed to assess the neuroprotective effects of FUS.
Collapse
Affiliation(s)
- Burak Yulug
- Department of Neurology, University of Istanbul-Medipol, Istanbul, Turkey; Regenerative and Restorative Medical Research Center, Experimental Neurology Laboratuary, University of Istanbul-Medipol, Istanbul, Turkey; Department of Physiology, University of Istanbul-Medipol, Istanbul, Turkey.
| | - Lutfu Hanoglu
- Department of Neurology, University of Istanbul-Medipol, Istanbul, Turkey; Regenerative and Restorative Medical Research Center, Experimental Neurology Laboratuary, University of Istanbul-Medipol, Istanbul, Turkey; Department of Physiology, University of Istanbul-Medipol, Istanbul, Turkey
| | - Ertugrul Kilic
- Regenerative and Restorative Medical Research Center, Experimental Neurology Laboratuary, University of Istanbul-Medipol, Istanbul, Turkey; Department of Physiology, University of Istanbul-Medipol, Istanbul, Turkey
| |
Collapse
|
165
|
Brzica H, Abdullahi W, Ibbotson K, Ronaldson PT. Role of Transporters in Central Nervous System Drug Delivery and Blood-Brain Barrier Protection: Relevance to Treatment of Stroke. J Cent Nerv Syst Dis 2017; 9:1179573517693802. [PMID: 28469523 PMCID: PMC5392046 DOI: 10.1177/1179573517693802] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/22/2017] [Indexed: 01/01/2023] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality in the United States. The only approved pharmacologic treatment for ischemic stroke is thrombolysis via recombinant tissue plasminogen activator (r-tPA). A short therapeutic window and serious adverse events (ie, hemorrhage, excitotoxicity) greatly limit r-tPA therapy, which indicates an essential need to develop novel stroke treatment paradigms. Transporters expressed at the blood-brain barrier (BBB) provide a significant opportunity to advance stroke therapy via central nervous system delivery of drugs that have neuroprotective properties. Examples of such transporters include organic anion–transporting polypeptides (Oatps) and organic cation transporters (Octs). In addition, multidrug resistance proteins (Mrps) are transporter targets in brain microvascular endothelial cells that can be exploited to preserve BBB integrity in the setting of stroke. Here, we review current knowledge on stroke pharmacotherapy and demonstrate how endogenous BBB transporters can be targeted for improvement of ischemic stroke treatment.
Collapse
Affiliation(s)
- Hrvoje Brzica
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Wazir Abdullahi
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Kathryn Ibbotson
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
166
|
Ljubimova JY, Sun T, Mashouf L, Ljubimov AV, Israel LL, Ljubimov VA, Falahatian V, Holler E. Covalent nano delivery systems for selective imaging and treatment of brain tumors. Adv Drug Deliv Rev 2017; 113:177-200. [PMID: 28606739 PMCID: PMC5578712 DOI: 10.1016/j.addr.2017.06.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 06/07/2017] [Indexed: 02/06/2023]
Abstract
Nanomedicine is a rapidly evolving form of therapy that holds a great promise for superior drug delivery efficiency and therapeutic efficacy than conventional cancer treatment. In this review, we attempt to cover the benefits and the limitations of current nanomedicines with special attention to covalent nano conjugates for imaging and drug delivery in the brain. The improvement in brain tumor treatment remains dismal despite decades of efforts in drug development and patient care. One of the major obstacles in brain cancer treatment is the poor drug delivery efficiency owing to the unique blood-brain barrier (BBB) in the CNS. Although various anti-cancer agents are available to treat tumors outside of the CNS, the majority fails to cross the BBB. In this regard, nanomedicines have increasingly drawn attention due to their multi-functionality and versatility. Nano drugs can penetrate BBB and other biological barriers, and selectively accumulate in tumor cells, while concurrently decreasing systemic toxicity.
Collapse
Affiliation(s)
- Julia Y Ljubimova
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, USA.
| | - Tao Sun
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, USA
| | - Leila Mashouf
- Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Alexander V Ljubimov
- Department of Biomedical Sciences, Board of Governors Regenerative Medicine Institute, Los Angeles, CA 90048, USA
| | - Liron L Israel
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, USA
| | - Vladimir A Ljubimov
- Department of Neurosurgery and Brain Repair, University of South Florida, 2 Tampa General Circle, Tampa, FL 33606, USA
| | - Vida Falahatian
- Duke University School of Medicine, Department of Biostatistics and Bioinformatics, Clinical Research Training Program (CRTP), 2424 Erwin Road, Suite 1102, Hock Plaza Box 2721, Durham, NC 27710, USA
| | - Eggehard Holler
- Nanomedicine Research Center, Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., AHSP, Los Angeles, CA 90048, USA; Institut für Biophysik und Physikalische Biochemie, Universität Regensburg, D-93040 Regensburg, Germany
| |
Collapse
|
167
|
Venugopal I, Habib N, Linninger A. Intrathecal magnetic drug targeting for localized delivery of therapeutics in the CNS. Nanomedicine (Lond) 2017; 12:865-877. [PMID: 28339319 DOI: 10.2217/nnm-2016-0418] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The challenge in treating neurological diseases is not lack of drug potency, but ineffective targeting techniques. We propose a technique called intrathecal magnetic drug targeting (IT-MDT), in which intrathecally injected magnetic nanoparticles (MNPs) are targeted to specific sites using external magnets. MATERIALS & METHODS MRI and histology confirmed localization of MNPs via IT-MDT at target sites along the spine of Sprague-Dawley rats. RESULTS MRI results confirmed greater MNP localization when the duration of magnet application was extended. Histological analysis quantified MNP tissue uptake and provided insight into their route of transport into deeper tissue regions. CONCLUSION IT-MDT has potential for future use in neurological disease treatments. It can produce localized therapeutic effect, with decreased systemic toxicity.
Collapse
Affiliation(s)
- Indu Venugopal
- Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA
| | - Nazia Habib
- Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA
| | - Andreas Linninger
- Department of Bioengineering, University of Illinois at Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA
| |
Collapse
|
168
|
Gandomi N, Varshochian R, Atyabi F, Ghahremani MH, Sharifzadeh M, Amini M, Dinarvand R. Solid lipid nanoparticles surface modified with anti-Contactin-2 or anti-Neurofascin for brain-targeted delivery of medicines. Pharm Dev Technol 2017; 22:426-435. [DOI: 10.1080/10837450.2016.1226901] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Nargess Gandomi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Varshochian
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
169
|
The role of multidrug resistance protein (MRP-1) as an active efflux transporter on blood-brain barrier (BBB) permeability. Mol Divers 2017; 21:355-365. [PMID: 28050687 DOI: 10.1007/s11030-016-9715-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/16/2016] [Indexed: 01/30/2023]
Abstract
Drugs acting on central nervous system (CNS) may take longer duration to reach the market as these compounds have a higher attrition rate in clinical trials due to the complexity of the brain, side effects, and poor blood-brain barrier (BBB) permeability compared to non-CNS-acting compounds. The roles of active efflux transporters with BBB are still unclear. The aim of the present work was to develop a predictive model for BBB permeability that includes the MRP-1 transporter, which is considered as an active efflux transporter. A support vector machine model was developed for the classification of MRP-1 substrates and non-substrates, which was validated with an external data set and Y-randomization method. An artificial neural network model has been developed to evaluate the role of MRP-1 on BBB permeation. A total of nine descriptors were selected, which included molecular weight, topological polar surface area, ClogP, number of hydrogen bond donors, number of hydrogen bond acceptors, number of rotatable bonds, P-gp, BCRP, and MRP-1 substrate probabilities for model development. We identified 5 molecules that fulfilled all criteria required for passive permeation of BBB, but they all have a low logBB value, which suggested that the molecules were effluxed by the MRP-1 transporter.
Collapse
|
170
|
Wang D, Wu LP. Nanomaterials for delivery of nucleic acid to the central nervous system (CNS). MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:1039-1046. [PMID: 27772703 DOI: 10.1016/j.msec.2016.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/11/2016] [Accepted: 04/04/2016] [Indexed: 11/08/2022]
|
171
|
Abstract
The treatment of microbial infections has suffered greatly in this present century of pathogen dominance. Inspite of extensive research efforts and scientific advancements, the worldwide emergence of microbial tolerance continues to plague survivability. The innate property of microbe to resist any antibiotic due to evolution is the virtue of intrinsic resistance. However, the classical genetic mutations and extrachromosomal segments causing gene exchange attribute to acquired tolerance development. Rampant use of antimicrobials causes certain selection pressure which increases the resistance frequency. Genomic duplication, enzymatic site modification, target alteration, modulation in membrane permeability, and the efflux pump mechanism are the major contributors of multidrug resistance (MDR), specifically antibiotic tolerance development. MDRs will lead to clinical failures for treatment and pose health crisis. The molecular mechanisms of antimicrobial resistance are diverse as well as complex and still are exploited for new discoveries in order to prevent the surfacing of “superbugs.” Antimicrobial chemotherapy has diminished the threat of infectious diseases to some extent. To avoid the indiscriminate use of antibiotics, the new ones licensed for use have decreased with time. Additionally, in vitro assays and genomics for anti-infectives are novel approaches used in resolving the issues of microbial resistance. Proper use of drugs can keep it under check and minimize the risk of MDR spread.
Collapse
|
172
|
Mignani S, Bryszewska M, Zablocka M, Klajnert-Maculewicz B, Cladera J, Shcharbin D, Majoral JP. Can dendrimer based nanoparticles fight neurodegenerative diseases? Current situation versus other established approaches. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2016.09.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
173
|
Zheng Y, Chen X, Benet LZ. Reliability of In Vitro and In Vivo Methods for Predicting the Effect of P-Glycoprotein on the Delivery of Antidepressants to the Brain. Clin Pharmacokinet 2016; 55:143-67. [PMID: 26293617 DOI: 10.1007/s40262-015-0310-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
As the effect of P-glycoprotein (P-gp) transport on antidepressant delivery has been extensively evaluated using in vitro cellular and in vivo rodent models, an increasing number of publications have addressed the effect of P-gp in limiting brain penetration of antidepressants and causing treatment-resistant depression in current clinical therapies. However, contradictory results have been observed in different systems. It is of vital importance to understand the potential for drug interactions related to P-gp at the blood-brain barrier (BBB), and whether coadministration of a P-gp inhibitor together with an antidepressant is a good clinical strategy for dosing of patients with treatment-resistant depression. In this review, the complicated construction of the BBB, the transport mechanisms for compounds that cross the BBB, and the basic characteristics of antidepressants are illustrated. Further, the reliability of different systems related to antidepressant brain delivery, including in vitro bidirectional transport cell lines, in vivo Mdr1 knockout mice, and chemical inhibition studies in rodents are analyzed, supporting a low possibility that P-gp affects currently marketed antidepressants when these results are extrapolated to the human BBB. These findings can also be applied to other central nervous system drugs.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 533 Parnassus Avenue, Room U-68, San Francisco, CA, 94143-0912, USA
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xijing Chen
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 533 Parnassus Avenue, Room U-68, San Francisco, CA, 94143-0912, USA.
| |
Collapse
|
174
|
Ali T, Kim MJ, Rehman SU, Ahmad A, Kim MO. Anthocyanin-Loaded PEG-Gold Nanoparticles Enhanced the Neuroprotection of Anthocyanins in an Aβ 1-42 Mouse Model of Alzheimer's Disease. Mol Neurobiol 2016; 54:6490-6506. [PMID: 27730512 DOI: 10.1007/s12035-016-0136-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/14/2016] [Indexed: 12/31/2022]
Abstract
Nanomedicine is an emerging research area. In this study, we investigated the neuroprotective efficacy of anthocyanin-loaded polyethylene glycol-gold nanoparticles (PEG-AuNPs) for enhancing the neuroprotective efficacy of anthocyanins in an amyloid beta (Aβ)1-42 mouse model of Alzheimer's disease. We observed that both anthocyanin-loaded PEG-AuNPs and anthocyanins treatment (12 μg/g/day for 14 days) ameliorated memory impairments in the Aβ1-42-injected mice. However, the anthocyanin-loaded PEG-AuNPs were more effective than free anthocyanins. Anthocyanin-loaded PEG-AuNPs protected pre- and post-synaptic proteins from Aβ1-42-induced synaptic dysfunction. Interestingly, the anthocyanin-loaded PEG-AuNPs also regulated the p-PI3K/p-Akt/p-GSK3β pathway and, as a result, prevented the hyperphosphorylation of tau protein at serines 413 and 404 in the Aβ1-42-injected mice. Western blot results of cytochrome c, Bax/Bcl2, caspases and poly (ADP-ribose) polymerase-1 expression levels, and immunohistochemical Nissl and Fluoro-Jade B staining also indicated that the anthocyanin-loaded PEG-AuNPs inhibited apoptosis and neurodegeneration in the Aβ1-42-injected mice. Our results suggest that the conjugation of dietary polyphenolic compounds with gold nanoparticles, such as anthocyanin-loaded PEG-AuNPs, is a novel approach that may represent an important and promising nanomedicine strategy to prevent age-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Tahir Ali
- Division of Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Min Ju Kim
- Division of Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Shafiq Ur Rehman
- Division of Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Ashfaq Ahmad
- Division of Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Myeong Ok Kim
- Division of Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea.
| |
Collapse
|
175
|
Jeon WJ, Dean B, Scarr E, Gibbons A. The Role of Muscarinic Receptors in the Pathophysiology of Mood Disorders: A Potential Novel Treatment? Curr Neuropharmacol 2016; 13:739-49. [PMID: 26630954 PMCID: PMC4759313 DOI: 10.2174/1570159x13666150612230045] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 12/30/2014] [Accepted: 02/28/2015] [Indexed: 12/21/2022] Open
Abstract
The central cholinergic system has been implicated in the pathophysiology of mood disorders. An imbalance in central cholinergic neurotransmitter activity has been proposed to contribute to the manic and depressive episodes typical of these disorders. Neuropharmacological studies into the effects of cholinergic agonists and antagonists on mood state have provided considerable support for this hypothesis. Furthermore, recent clinical studies have shown that the pan-CHRM antagonist, scopolamine, produces rapid-acting antidepressant effects in individuals with either major depressive disorder (MDD) or bipolar disorder (BPD), such as bipolar depression, contrasting the delayed therapeutic response of conventional mood stabilisers and antidepressants. This review presents recent data from neuroimaging, post-mortem and genetic studies supporting the involvement of muscarinic cholinergic receptors (CHRMs), particularly CHRM2, in the pathophysiology of MDD and BPD. Thus, novel drugs that selectively target CHRMs with negligible effects in the peripheral nervous system might produce more rapid and robust clinical improvement in patients with BPD and MDD.
Collapse
Affiliation(s)
| | - Brian Dean
- Florey Institute for Neuroscience and Mental Health, 30 Royal Parade, Parkville, Victoria 3052, Australia.
| | | | | |
Collapse
|
176
|
Yunivita V, Dian S, Ganiem AR, Hayati E, Hanggono Achmad T, Purnama Dewi A, Teulen M, Meijerhof-Jager P, van Crevel R, Aarnoutse R, Ruslami R. Pharmacokinetics and safety/tolerability of higher oral and intravenous doses of rifampicin in adult tuberculous meningitis patients. Int J Antimicrob Agents 2016; 48:415-21. [PMID: 27526979 DOI: 10.1016/j.ijantimicag.2016.06.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/10/2016] [Accepted: 06/18/2016] [Indexed: 11/20/2022]
Abstract
High-dose intravenous (i.v.) rifampicin improved the outcome of tuberculous meningitis (TBM) in a previous study. Unfortunately, i.v. rifampicin is not available in many high-endemic settings. This study examined exposures to and safety of higher oral rifampicin doses compared with i.v. rifampicin. Thirty adult Indonesian TBM patients were randomised to rifampicin 750 mg (ca. 17 mg/kg) orally, 900 mg (ca. 20 mg/kg) orally or 600 mg (ca. 13 mg/kg, as used previously) i.v. over 1.5 h for 14 days, combined with other TB drugs. The pharmacokinetics of rifampicin was assessed in the critical phase of TBM treatment (≤3 days after treatment initiation) and at ≥9 days. In the first days of treatment, the geometric mean (range) plasma AUC0-24 values following rifampicin 750 mg orally, 900 mg orally and 600 mg i.v. were 131.4 (38.1-275.1), 164.8 (66.9-291.2) and 145.7 (77.7-430.2) mg⋅h/L, respectively; Cmax values were 14.3 (6.1-22.2), 16.2 (5.7-28.3) and 24.7 (13.9-37.8) mg/L. CSF concentrations correlated with plasma exposures. After ≥9 days, AUC0-24 values had decreased to 100.1, 101.2 and 94.9 mg⋅h/L. Transient grade 3 ALT increases (8/30 patients) and one grade 4 ALT increase occurred, not related to rifampicin exposure. Higher oral rifampicin doses resulted in approximately similar plasma AUC0-24 but lower plasma Cmax values compared with 600 mg i.v. over 1.5 h. Exposures to rifampicin varied substantially and decreased due to autoinduction. Liver function disturbances occurred in this severely ill population. Future studies should examine even higher rifampicin doses in TBM treatment.
Collapse
Affiliation(s)
- Vycke Yunivita
- Department of Pharmacology and Therapy, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Sofiati Dian
- Department of Neurology, Faculty of Medicine, Hasan Sadikin Hospital, Universitas Padjadjaran, Bandung, Indonesia
| | - Ahmad Rizal Ganiem
- Department of Neurology, Faculty of Medicine, Hasan Sadikin Hospital, Universitas Padjadjaran, Bandung, Indonesia
| | - Ela Hayati
- Department of Neurology, Faculty of Medicine, Hasan Sadikin Hospital, Universitas Padjadjaran, Bandung, Indonesia
| | - Tri Hanggono Achmad
- Department of Biochemistry, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Atu Purnama Dewi
- Department of Pharmacology and Therapy, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Marga Teulen
- Department of Pharmacy, Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen 65625 GA, The Netherlands
| | - Petra Meijerhof-Jager
- Department of Pharmacy, Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen 65625 GA, The Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob Aarnoutse
- Department of Pharmacy, Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen 65625 GA, The Netherlands.
| | - Rovina Ruslami
- Department of Pharmacology and Therapy, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
177
|
Draffehn S, Eichhorst J, Wiesner B, Kumke MU. Insight into the Modification of Polymeric Micellar and Liposomal Nanocarriers by Fluorescein-Labeled Lipids and Uptake-Mediating Lipopeptides. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:6928-6939. [PMID: 27295095 DOI: 10.1021/acs.langmuir.6b01487] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Encapsulation of diagnostic and therapeutic compounds in transporters improves their delivery to the point of need. An even more efficient treatment of diseases can be achieved using carriers with targeting or protecting moieties. In the present work, we investigated micellar and liposomal nanocarriers modified with fluorescein, peptides, and polymers that are covalently bound to fatty acids or phospholipids to ensure a self-driven incorporation into the micelles or liposomes. First, we characterized the photophysics of the fluorescent probes in the absence and in the presence of nanocarriers. Changes in the fluorescence decay time, quantum yield, and intensity of a fluorescein-labeled fatty acid (fluorescein-labeled palmitic acid [fPA]) and a fluorescein-labeled lipopeptide (P2fA2) were found. By exploiting these changes, we investigated a lipopeptide (P2A2 as an uptake-mediating unit) in combination with different nanocarriers (micelles and liposomes) and determined the corresponding association constant Kass values, which were found to be very high. In addition, the mobility of fPA was exploited using fluorescence correlation spectroscopy (FCS) and fluorescence depolarization (FD) experiments to characterize the nanocarriers. Cellular uptake experiments with mouse brain endothelial cells provided information on the uptake behavior of liposomes modified by uptake-mediating P2A2 and revealed differences in the uptake behavior between pH-sensitive and pH-insensitive liposomes.
Collapse
Affiliation(s)
- Sören Draffehn
- Department of Physical Chemistry, University of Potsdam , Karl-Liebknecht-Str. 24-25, 14476 Potsdam-Golm, Germany
| | - Jenny Eichhorst
- Department of Cellular Imaging, Leibniz Institut für Molekulare Pharmakologie (FMP) , Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - Burkhard Wiesner
- Department of Cellular Imaging, Leibniz Institut für Molekulare Pharmakologie (FMP) , Robert-Roessle-Str. 10, 13125 Berlin, Germany
| | - Michael U Kumke
- Department of Physical Chemistry, University of Potsdam , Karl-Liebknecht-Str. 24-25, 14476 Potsdam-Golm, Germany
| |
Collapse
|
178
|
Jakki SL, Ramesh YV, Gowthamarajan K, Senthil V, Jain K, Sood S, Pathak D. Novel anionic polymer as a carrier for CNS delivery of anti-Alzheimer drug. Drug Deliv 2016; 23:3471-3479. [DOI: 10.1080/10717544.2016.1196767] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Satya Lavanya Jakki
- Department of Pharmaceutics, JSS College of Pharmacy, Off Campus J.S.S. University, Mysore, Udhagamandalam, Tamil Nadu, India,
| | - Yasam Venkata Ramesh
- Department of Pharmaceutics, JSS College of Pharmacy, Off Campus J.S.S. University, Mysore, Udhagamandalam, Tamil Nadu, India,
| | - K. Gowthamarajan
- Department of Pharmaceutics, JSS College of Pharmacy, Off Campus J.S.S. University, Mysore, Udhagamandalam, Tamil Nadu, India,
| | - V. Senthil
- Department of Pharmaceutics, JSS College of Pharmacy, Off Campus J.S.S. University, Mysore, Udhagamandalam, Tamil Nadu, India,
| | - Kunal Jain
- Scientific Writer, Novartis Healthcare Pvt. Ltd, Hyderabad, Andhra Pradesh, India,
| | - Sumeet Sood
- Scientific Writer, Indegene Lifesystems Pvt. Ltd, Bengaluru, Karnataka, India, and
| | - Deepa Pathak
- Head (R&D), United Biotech Pvt. Ltd, Baddi, Himachal Pradesh, India
| |
Collapse
|
179
|
Aparicio-Blanco J, Martín-Sabroso C, Torres-Suárez AI. In vitro screening of nanomedicines through the blood brain barrier: A critical review. Biomaterials 2016; 103:229-255. [PMID: 27392291 DOI: 10.1016/j.biomaterials.2016.06.051] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier accounts for the high attrition rate of the treatments of most brain disorders, which therefore remain one of the greatest health-care challenges of the twenty first century. Against this background of hindrance to brain delivery, nanomedicine takes advantage of the assembly at the nanoscale of available biomaterials to provide a delivery platform with potential to raising brain levels of either imaging or therapeutic agents. Nevertheless, to prevent later failure due to ineffective drug levels at the target site, researchers have been endeavoring to develop a battery of in vitro screening procedures that can predict earlier in the drug discovery process the ability of these cutting-edge drug delivery platforms to cross the blood-brain barrier for biomedical purposes. This review provides an in-depth analysis of the currently available in vitro blood-brain barrier models (both cell-based and non-cell-based) with the focus on their suitability for understanding the biological brain distribution of forthcoming nanomedicines. The relationship between experimental factors and underlying physiological assumptions that would ultimately lead to a more predictive capacity of their in vivo performance, and those methods already assayed for the evaluation of the brain distribution of nanomedicines are comprehensively discussed.
Collapse
Affiliation(s)
- Juan Aparicio-Blanco
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain
| | - Cristina Martín-Sabroso
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain
| | - Ana-Isabel Torres-Suárez
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain; University Institute of Industrial Pharmacy, Complutense University, 28040, Madrid, Spain.
| |
Collapse
|
180
|
Rodriguez M, Kaushik A, Lapierre J, Dever SM, El-Hage N, Nair M. Electro-Magnetic Nano-Particle Bound Beclin1 siRNA Crosses the Blood-Brain Barrier to Attenuate the Inflammatory Effects of HIV-1 Infection in Vitro. J Neuroimmune Pharmacol 2016; 12:120-132. [PMID: 27287620 DOI: 10.1007/s11481-016-9688-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 05/31/2016] [Indexed: 10/21/2022]
Abstract
The purpose of this study was to evaluate a novel drug delivery system comprised of ferric-cobalt electro-magnetic nano-material (CoFe2O4@ BaTiO3; MENP) bound to siRNA targeting Beclin1 (MENP-siBeclin1) to cross the blood-brain barrier (BBB) and attenuate the neurotoxic effects of HIV-1 infection in the central nervous system following on-demand release of siRNA using an in vitro primary human BBB model. Beclin1 is a key protein in the regulation of the autophagy pathway and we have recently demonstrated the importance of Beclin1 in regulating viral replication and viral-induced inflammation in HIV-1-infected microglia. The MENP-siBeclin1 nano-formulation did not compromise the physiological function or integrity of the BBB model. Furthermore, the in vitro BBB data revealed that MENP-siBeclin1 could efficiently attenuate viral replication and viral-induced inflammation, likely due to STAT1/ NF-κB signaling pathways. MENP-siBeclin1 also silenced Beclin1 protein expression in HIV-1-infected microglial cells within the model system. In addition, the cytotoxic effects of direct treatment with siBeclin1 and MENP alone or in nano-formulation on primary human neuronal cells showed a minimal amount of cell death. Overall, the data shows that the nano-formulation can silence the BECN1 gene as an effective mechanism to attenuate HIV-1 replication and viral-induced inflammation in the context of the BBB.
Collapse
Affiliation(s)
- Myosotys Rodriguez
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ajeet Kaushik
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Jessica Lapierre
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Seth M Dever
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Nazira El-Hage
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - Madhavan Nair
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| |
Collapse
|
181
|
Gao H. Perspectives on Dual Targeting Delivery Systems for Brain Tumors. J Neuroimmune Pharmacol 2016; 12:6-16. [PMID: 27270720 DOI: 10.1007/s11481-016-9687-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/31/2016] [Indexed: 12/30/2022]
Abstract
Brain tumor remains one of the most serious threats to human beings. Different from peripheral tumors, drug delivery to brain tumor is largely restricted by the blood brain barrier (BBB). To fully conquer this barrier and specifically deliver drugs to brain tumor, dual targeting delivery systems were explored, which are functionalized with two active targeting ligands: one to the BBB and the other to the brain tumor. The development of dual targeting delivery system is still in its early stage, and attentions need to be paid to issues and concerns that remain unresolved in future studies.
Collapse
Affiliation(s)
- Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu, China.
| |
Collapse
|
182
|
Govender T, Choonara YE, Kumar P, Bijukumar D, du Toit LC, Modi G, Naidoo D, Pillay V. Implantable and transdermal polymeric drug delivery technologies for the treatment of central nervous system disorders. Pharm Dev Technol 2016; 22:476-486. [PMID: 27268737 DOI: 10.1080/10837450.2016.1189937] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The complexity of the brain and the membranous blood-brain barrier (BBB) has proved to be a significant limitation to the systemic delivery of pharmaceuticals to the brain rendering them sub-therapeutic and ineffective in the treatment of neurological diseases. Apart from this, lack of innovation in product development to counteract the problem is also a major contributing factor to a poor therapeutic outcome. Various innovative strategies show potential in treating some of the neurological disorders; however, drug delivery remains the most popular. To attain therapeutic drug levels in the central nervous system, large, intolerable systemic doses are generally administered. The major factors responsible for the success maintenance therapy of neurological diseases included controlled and sustained release of neurotherapeutics, reduced frequency of administration, higher bioavailability, and patient compliances. Conventional oral or injectable formulations cannot satisfy all the requirements in many circumstances. This article reviews the therapeutic implantable polymeric and transdermal devices employed in an attempt to effectively achieve therapeutic quantities of drug across the BBB over a prolonged period, to improve patient disease prognosis.
Collapse
Affiliation(s)
- Thiresen Govender
- a Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Yahya E Choonara
- a Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Pradeep Kumar
- a Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Divya Bijukumar
- a Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Lisa C du Toit
- a Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Girish Modi
- b Division of Neurosciences, Department of Neurology, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Dinesh Naidoo
- c Division of Neurosciences, Department of Neurosurgery, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Viness Pillay
- a Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| |
Collapse
|
183
|
Adabi M, Naghibzadeh M, Adabi M, Zarrinfard MA, Esnaashari SS, Seifalian AM, Faridi-Majidi R, Tanimowo Aiyelabegan H, Ghanbari H. Biocompatibility and nanostructured materials: applications in nanomedicine. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:833-842. [DOI: 10.1080/21691401.2016.1178134] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Naghibzadeh
- Department of Nanotechnology, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohsen Adabi
- Department of Metallurgy and Materials Engineering, Faculty of Engineering, Roudehen Branch, Islamic Azad University, Roudehen, Tehran, Iran
| | - Mohammad Ali Zarrinfard
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Sara Esnaashari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alexander M. Seifalian
- Centre for Nanotechnology and Regenerative Medicine, University College London, London, United Kingdom
| | - Reza Faridi-Majidi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hammed Tanimowo Aiyelabegan
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
184
|
Huang L, Deng M, He Y, Lu S, Ma R, Fang Y. β-asarone and levodopa co-administration increase striatal dopamine level in 6-hydroxydopamine induced rats by modulating P-glycoprotein and tight junction proteins at the blood-brain barrier and promoting levodopa into the brain. Clin Exp Pharmacol Physiol 2016; 43:634-43. [PMID: 26991136 DOI: 10.1111/1440-1681.12570] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Liping Huang
- Hainan Medical University; Haikou China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Minzhen Deng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Yuping He
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Shiyao Lu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Ruanxin Ma
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Yongqi Fang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| |
Collapse
|
185
|
Hernando S, Gartziandia O, Herran E, Pedraz JL, Igartua M, Hernandez RM. Advances in nanomedicine for the treatment of Alzheimer’s and Parkinson’s diseases. Nanomedicine (Lond) 2016; 11:1267-85. [DOI: 10.2217/nnm-2016-0019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Alzheimer‘s disease and Parkinson’s disease are the most common neurodegenerative diseases worldwide. Despite all the efforts made by the scientific community, current available treatments have limited effectiveness, without halting the progression of the disease. That is why, new molecules such as growth factors, antioxidants and metal chelators have been raised as new therapeutical approaches. However, these molecules have difficulties to cross the blood–brain barrier limiting its therapeutic effect. The development of nanometric drug delivery systems may permit a targeted and sustained release of old and new treatments offering a novel strategy to treat these neurodegenerative disorders. This review summarized the main investigated drug delivery systems as promising approaches to treat Alzheimer‘s disease and Parkinson’s disease.
Collapse
Affiliation(s)
- Sara Hernando
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Oihane Gartziandia
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Enara Herran
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| |
Collapse
|
186
|
Loureiro JA, Gomes B, Fricker G, Coelho MAN, Rocha S, Pereira MC. Cellular uptake of PLGA nanoparticles targeted with anti-amyloid and anti-transferrin receptor antibodies for Alzheimer's disease treatment. Colloids Surf B Biointerfaces 2016; 145:8-13. [PMID: 27131092 DOI: 10.1016/j.colsurfb.2016.04.041] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 04/12/2016] [Accepted: 04/19/2016] [Indexed: 12/27/2022]
Abstract
During the last few decades, relevant efforts have been reported to design nanocarriers for drug transport through the blood brain barrier (BBB). New drugs, such as peptide iAβ5, capable to inhibit the aggregates associated with Alzheimeŕs disease (AD) are being tested but the most frequent drawback is to reach the brain in the desired concentrations due to the low BBB permeability-surface area. Our approach, as a proof of concept to improve drug transport through the BBB, is based on poly(lactic-co-glycolic acid) (PLGA) nanoparticles with surface functionalized with anti-transferrin receptor monoclonal antibody (OX26) and anti-Aβ (DE2B4) to deliver encapsulated iAβ5 into the brain. Porcine brain capillary endothelial cells (PBCECs) were used as a BBB model to evaluate the system efficacy and toxicity. The uptake of immune nanoparticles with a controlled delivery of the peptide iAβ5 was substantially increased compared to the nanoparticles (NPs) without monoclonal antibody functionalization.
Collapse
Affiliation(s)
- Joana A Loureiro
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Bárbara Gomes
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Gert Fricker
- Institute for Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, 69120 Heidelberg, Germany
| | - Manuel A N Coelho
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Sandra Rocha
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg SE-41296, Sweden
| | - Maria Carmo Pereira
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal.
| |
Collapse
|
187
|
Hänggi D, Etminan N, Steiger HJ, Johnson M, Peet MM, Tice T, Burton K, Hudson B, Turner M, Stella A, Heshmati P, Davis C, Faleck HJ, Macdonald RL. A Site-Specific, Sustained-Release Drug Delivery System for Aneurysmal Subarachnoid Hemorrhage. Neurotherapeutics 2016; 13:439-49. [PMID: 26935204 PMCID: PMC4824023 DOI: 10.1007/s13311-016-0424-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Nimodipine is the only drug approved for use by the Food and Drug Administration for improving outcome after aneurysmal subarachnoid hemorrhage (SAH). It has less than optimal efficacy, causes dose-limiting hypotension in a substantial proportion of patients, and is administered enterally 6 times daily. We describe development of site-specific, sustained-release nimodipine microparticles that can be delivered once directly into the subarachnoid space or cerebral ventricles for potential improvement in outcome of patients with aneurysmal SAH. Eight injectable microparticle formulations of nimodipine in poly(DL-lactide-co-glycolide) (PLGA) polymers of varying composition were tested in vitro, and 1 was advanced into preclinical studies and clinical application. Intracisternal or intraventricular injection of nimodipine-PLGA microparticles in rats and beagles demonstrated dose-dependent, sustained concentrations of nimodipine in plasma and cerebrospinal fluid for up to 29 days with minimal toxicity in the brain or systemic tissues at doses <2 mg in rats and 51 mg in beagles, which would be equivalent of up to 612-1200 mg in humans, based on scaling relative to cerebrospinal fluid volumes. Efficacy was tested in the double-hemorrhage dog model of SAH. Nimodipine-PLGA microparticles significantly attenuated angiographic vasospasm. This therapeutic approach shows promise for improving outcome after SAH and may have broader applicability for similar diseases that are confined to body cavities or spaces, are self-limited, and lack effective treatments.
Collapse
Affiliation(s)
- Daniel Hänggi
- Department of Neurosurgery, University Medical Center Mannheim, Ruprecht-Karls-University Heidelberg, Germany, Mannheim, Germany.
| | - Nima Etminan
- Department of Neurosurgery, University Medical Center Mannheim, Ruprecht-Karls-University Heidelberg, Germany, Mannheim, Germany
| | - Hans Jakob Steiger
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | | | | - Tom Tice
- Evonik Industries, Birmingham, AL, USA
| | | | | | | | | | | | | | | | - R Loch Macdonald
- Edge Therapeutics, Inc., Berkeley Heights, NJ, USA
- Division of Neurosurgery, St. Michael's Hospital, Labatt Family Centre of Excellence in Brain Injury and Trauma Research, Keenan Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
188
|
On NH, Yathindranath V, Sun Z, Miller DW. Pathways for Drug Delivery to the Central Nervous System. Drug Deliv 2016. [DOI: 10.1002/9781118833322.ch16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
189
|
Engelhardt L, Röhm M, Mavoungou C, Schindowski K, Schafmeister A, Simon U. First Steps to Develop and Validate a CFPD Model in Order to Support the Design of Nose-to-Brain Delivered Biopharmaceuticals. Pharm Res 2016; 33:1337-50. [PMID: 26887679 DOI: 10.1007/s11095-016-1875-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 02/08/2016] [Indexed: 11/24/2022]
Abstract
PURPOSE Aerosol particle deposition in the human nasal cavity is of high interest in particular for intranasal central nervous system (CNS) drug delivery via the olfactory cleft. The objective of this study was the development and comparison of a numerical and experimental model to investigate various parameters for olfactory particle deposition within the complex anatomical nasal geometry. METHODS Based on a standardized nasal cavity, a computational fluid and particle dynamics (CFPD) model was developed that enables the variation and optimization of different parameters, which were validated by in vitro experiments using a constructed rapid-prototyped human nose model. RESULTS For various flow rates (5 to 40 l/min) and particle sizes (1 to 10 μm), the airflow velocities, the calculated particle airflow patterns and the particle deposition correlated very well with the experiment. Particle deposition was investigated numerically by varying particle sizes at constant flow rate and vice versa assuming the particle size distribution of the used nebulizer. CONCLUSIONS The developed CFPD model could be directly translated to the in vitro results. Hence, it can be applied for parameter screening and will contribute to the improvement of aerosol particle deposition at the olfactory cleft for CNS drug delivery in particular for biopharmaceuticals.
Collapse
Affiliation(s)
- Lucas Engelhardt
- Scientific Computing Centre Ulm, Ulm University, Helmholtzstraße 20, 89081, Ulm, Germany
| | - Martina Röhm
- Institute of Applied Biotechnology, Biberach University of Applied Sciences, Hubertus-Liebrecht-Strasse 35, 88400, Biberach, Germany. .,Faculty of Medicine, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| | - Chrystelle Mavoungou
- Institute of Applied Biotechnology, Biberach University of Applied Sciences, Hubertus-Liebrecht-Strasse 35, 88400, Biberach, Germany
| | - Katharina Schindowski
- Institute of Applied Biotechnology, Biberach University of Applied Sciences, Hubertus-Liebrecht-Strasse 35, 88400, Biberach, Germany
| | - Annette Schafmeister
- Institute of Applied Biotechnology, Biberach University of Applied Sciences, Hubertus-Liebrecht-Strasse 35, 88400, Biberach, Germany
| | - Ulrich Simon
- Scientific Computing Centre Ulm, Ulm University, Helmholtzstraße 20, 89081, Ulm, Germany
| |
Collapse
|
190
|
Nanocarriers for the treatment of glioblastoma multiforme: Current state-of-the-art. J Control Release 2016; 227:23-37. [PMID: 26892752 DOI: 10.1016/j.jconrel.2016.02.026] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/12/2016] [Accepted: 02/13/2016] [Indexed: 01/26/2023]
Abstract
Glioblastoma multiforme, a grade IV glioma, is the most frequently occurring and invasive primary tumor of the central nervous system, which causes about 4% of cancer-associated-deaths, making it one of the most fatal cancers. With present treatments, using state-of-the-art technologies, the median survival is about 14 months and 2 year survival rate is merely 3-5%. Hence, novel therapeutic approaches are urgently necessary. However, most drug molecules are not able to cross the blood-brain barrier, which is one of the major difficulties in glioblastoma treatment. This review describes the features of blood-brain barrier, and its anatomical changes with different stages of tumor growth. Moreover, various strategies to improve brain drug delivery i.e. tight junction opening, chemical modification of the drug, efflux transporter inhibition, convection-enhanced delivery, craniotomy-based drug delivery and drug delivery nanosystems are discussed. Nanocarriers are one of the highly potential drug transport systems that have gained huge research focus over the last few decades for site specific drug delivery, including drug delivery to the brain. Properly designed nanocolloids are capable to cross the blood-brain barrier and specifically deliver the drug in the brain tumor tissue. They can carry both hydrophilic and hydrophobic drugs, protect them from degradation, release the drug for sustained period, significantly improve the plasma circulation half-life and reduce toxic effects. Among various nanocarriers, liposomes, polymeric nanoparticles and lipid nanocapsules are the most widely studied, and are discussed in this review. For each type of nanocarrier, a general discussion describing their composition, characteristics, types and various uses is followed by their specific application to glioblastoma treatment. Moreover, some of the main challenges regarding toxicity and standardized evaluation techniques are narrated in brief.
Collapse
|
191
|
Gao Z, Ma T, Zhao E, Docter D, Yang W, Stauber RH, Gao M. Small is Smarter: Nano MRI Contrast Agents - Advantages and Recent Achievements. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:556-76. [PMID: 26680328 DOI: 10.1002/smll.201502309] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 09/22/2015] [Indexed: 05/23/2023]
Abstract
Many challenges for advanced sensitive and noninvasive clinical diagnostic imaging remain unmatched. In particular, the great potential of magnetic nano-probes is intensively discussed to further improve the performance of magnetic resonance imaging (MRI), especially for cancer diagnosis. Based on recent achievements, here the concepts of magnetic nanoparticle-based MRI contrast agents and tumor-specific imaging probes are critically summarized. Advances in their synthesis, biocompatible chemical and biofunctional surface modifications, and current strategies for further developing them into multimodality imaging probes are discussed. In addition, how engineered versus unintended surface coatings such as protein coronas affect the biocompatibility and performance of MRI nano-probes is also considered. To stimulate progress in the field, future strategies and relevant challenges that still need to be resolved in the field conclude this review.
Collapse
Affiliation(s)
- Zhenyu Gao
- College of Chemistry, Jilin University, Changchun, 130012, China
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China
| | - Tiancong Ma
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China
| | - Enyu Zhao
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China
| | - Dominic Docter
- Department of Nanobiomedicine, ENT/University Medical Center of Mainz, Langenbeckstr. 1, 55101, Mainz, Germany
| | - Wensheng Yang
- College of Chemistry, Jilin University, Changchun, 130012, China
| | - Roland H Stauber
- Department of Nanobiomedicine, ENT/University Medical Center of Mainz, Langenbeckstr. 1, 55101, Mainz, Germany
| | - Mingyuan Gao
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China
| |
Collapse
|
192
|
Jenkins SI, Weinberg D, al-Shakli AF, Fernandes AR, Yiu HH, Telling ND, Roach P, Chari DM. ‘Stealth’ nanoparticles evade neural immune cells but also evade major brain cell populations: Implications for PEG-based neurotherapeutics. J Control Release 2016; 224:136-145. [DOI: 10.1016/j.jconrel.2016.01.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/18/2022]
|
193
|
Berg C. Quantitative analysis of nanoparticle transport through in vitro blood-brain barrier models. Tissue Barriers 2016; 4:e1143545. [PMID: 27141425 PMCID: PMC4836482 DOI: 10.1080/21688370.2016.1143545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/08/2016] [Accepted: 01/12/2016] [Indexed: 01/11/2023] Open
Abstract
Nanoparticle transport through the blood-brain barrier has received much attention of late, both from the point of view of nano-enabled drug delivery, as well as due to concerns about unintended exposure of nanomaterials to humans and other organisms. In vitro models play a lead role in efforts to understand the extent of transport through the blood-brain barrier, but unique features of the nanoscale challenge their direct adaptation. Here we highlight some of the differences compared to molecular species when utilizing in vitro blood-brain barrier models for nanoparticle studies. Issues that may arise with transwell systems are discussed, together with some potential alternative methodologies. We also briefly review the biomolecular corona concept and its importance for how nanoparticles interact with the blood-brain barrier. We end with considering future directions, including indirect effects and application of shear and fluidics-technologies.
Collapse
Affiliation(s)
- Christoffer Berg
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen ; Groningen, The Netherlands
| |
Collapse
|
194
|
Mandal S, Mandal SD, Chuttani K, Sawant KK, Subudhi BB. Design and evaluation of mucoadhesive microemulsion for neuroprotective effect of ibuprofen following intranasal route in the MPTP mice model. Drug Dev Ind Pharm 2016; 42:1340-50. [PMID: 26710671 DOI: 10.3109/03639045.2015.1135936] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The present study is to investigate the neuroprotective effect of ibuprofen by intranasal administration of mucoadhesive microemulsion (MMEI) against inflammation-mediated by dopaminergic neurodegeneration in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson's disease (PD). METHODS Ibuprofen-loaded polycarbophil-based MMEI was developed by using response surface methodology (RSM). Ibuprofen with dose of 2.86 mg/kg/day was administered intranasally to male C57BL/6 mice for two consecutive weeks which were pre-treated with four intraperitoneal injections of MPTP (20 mg/kg of body weight) at 2 h intervals. Immunohistochemistry was performed. RESULTS Optimal MMEI was stable and non-ciliotoxic with 66.29 ± 4.15 nm as average globule size and -20.9 ± 3.98 mV as zeta potential. PDI value and transmission electron microscopy result showed the narrow globule size distribution of MMEI. The result showed that all three independent variables had a significant effect (p < 0.05) on the responses. Rota-rod and open-field test findings revealed the significant improvement in motor performance and gross behavioral activity of the mice. The results from in vivo study and immunohistochemistry showed that nasal administration of Ibuprofen significantly reduced the MPTP-mediated dopamine depletion. Furthermore TH neurons count in the substantia nigra and the density of striatal dopaminergic nerve terminals were found to be significant higher for ibuprofen treated groups. CONCLUSION Findings of the investigation revealed that Ibuprofen through developed MMEI was shown to protect neurons against MPTP-induced injury in the Substantia nigra pars compacta (SNpc) and striatum and hence, could be a promising approach for brain targeting of Ibuprofen through intranasal route to treat PD.
Collapse
Affiliation(s)
- Surjyanarayan Mandal
- a School of Pharmaceutical Sciences , Siksha 'O' Anusandhan University , Khandagiri Square , Bhubaneswar , Orissa , India
| | - Snigdha Das Mandal
- b Department of Pharmacology , Parul Institute of Pharmacy and Research , Vadodara , Gujarat , India
| | - Krishna Chuttani
- c Division of Cyclotron & Radiopharmaceutical Sciences , Institute of Nuclear Medicine and Allied Sciences (INMAS) , DRDO , Delhi , India
| | - Krutika K Sawant
- d Department of Pharmaceutics , MS University , TIFAC Core , Vadodara , India
| | - Bharat Bhushan Subudhi
- a School of Pharmaceutical Sciences , Siksha 'O' Anusandhan University , Khandagiri Square , Bhubaneswar , Orissa , India
| |
Collapse
|
195
|
Pang Y, Lin S, Wright C, Shen J, Carter K, Bhatt A, Fan LW. Intranasal insulin protects against substantia nigra dopaminergic neuronal loss and alleviates motor deficits induced by 6-OHDA in rats. Neuroscience 2016; 318:157-65. [PMID: 26777890 DOI: 10.1016/j.neuroscience.2016.01.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/22/2015] [Accepted: 01/08/2016] [Indexed: 01/04/2023]
Abstract
Protection of substantia nigra (SN) dopaminergic (DA) neurons by neurotrophic factors (NTFs) is one of the promising strategies in Parkinson's disease (PD) therapy. A major clinical challenge for NTF-based therapy is that NTFs need to be delivered into the brain via invasive means, which often shows limited delivery efficiency. The nose to brain pathway is a non-invasive brain drug delivery approach developed in recent years. Of particular interest is the finding that intranasal insulin improves cognitive functions in Alzheimer's patients. In vitro, insulin has been shown to protect neurons against various insults. Therefore, the current study was designed to test whether intranasal insulin could afford neuroprotection in the 6-hydroxydopamine (6-OHDA)-based rat PD model. 6-OHDA was injected into the right side of striatum to induce a progressive DA neuronal lesion in the ipsilateral SN pars compact (SNc). Recombinant human insulin was applied intranasally to rats starting from 24h post lesion, once per day, for 2 weeks. A battery of motor behavioral tests was conducted on day 8 and 15. The number of DA neurons in the SNc was estimated by stereological counting. Our results showed that 6-OHDA injection led to significant motor deficits and 53% of DA neuron loss in the ipsilateral side of injection. Treatment with insulin significantly ameliorated 6-OHDA-induced motor impairments, as shown by improved locomotor activity, tapered/ledged beam-walking performance, vibrissa-elicited forelimb-placing, initial steps, as well as methamphetamine-induced rotational behavior. Consistent with behavioral improvements, insulin treatment provided a potent protection of DA neurons in the SNc against 6-OHDA neurotoxicity, as shown by a 74.8% increase in tyrosine hydroxylase (TH)-positive neurons compared to the vehicle group. Intranasal insulin treatment did not affect body weight and blood glucose levels. In conclusion, our study showed that intranasal insulin provided strong neuroprotection in the 6-OHDA rat PD model, suggesting that insulin signaling may be a novel therapeutic target in broad neurodegenerative disorders.
Collapse
Affiliation(s)
- Y Pang
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States.
| | - S Lin
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - C Wright
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - J Shen
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - K Carter
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - A Bhatt
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| | - L-W Fan
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, United States
| |
Collapse
|
196
|
Shah S. The nanomaterial toolkit for neuroengineering. NANO CONVERGENCE 2016; 3:25. [PMID: 28191435 PMCID: PMC5271150 DOI: 10.1186/s40580-016-0086-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 09/29/2016] [Indexed: 05/03/2023]
Abstract
There is a growing interest in developing effective tools to better probe the central nervous system (CNS), to understand how it works and to treat neural diseases, injuries and cancer. The intrinsic complexity of the CNS has made this a challenging task for decades. Yet, with the extraordinary recent advances in nanotechnology and nanoscience, there is a general consensus on the immense value and potential of nanoscale tools for engineering neural systems. In this review, an overview of specialized nanomaterials which have proven to be the most effective tools in neuroscience is provided. After a brief background on the prominent challenges in the field, a variety of organic and inorganic-based nanomaterials are described, with particular emphasis on the distinctive properties that make them versatile and highly suitable in the context of the CNS. Building on this robust nano-inspired foundation, the rational design and application of nanomaterials can enable the generation of new methodologies to greatly advance the neuroscience frontier.
Collapse
Affiliation(s)
- Shreyas Shah
- Physiological Communications Research Group, Nokia Bell Labs, 600 Mountain Avenue, Murray Hill, NJ 07974 USA
| |
Collapse
|
197
|
Gartziandia O, Egusquiaguirre SP, Bianco J, Pedraz JL, Igartua M, Hernandez RM, Préat V, Beloqui A. Nanoparticle transport across in vitro olfactory cell monolayers. Int J Pharm 2015; 499:81-89. [PMID: 26721725 DOI: 10.1016/j.ijpharm.2015.12.046] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/16/2015] [Accepted: 12/16/2015] [Indexed: 11/29/2022]
Abstract
Drug access to the CNS is hindered by the presence of the blood-brain barrier (BBB), and the intranasal route has risen as a non-invasive route to transport drugs directly from nose-to-brain avoiding the BBB. In addition, nanoparticles (NPs) have been described as efficient shuttles for direct nose-to-brain delivery of drugs. Nevertheless, there are few studies describing NP nose-to-brain transport. Thus, the aim of this work was (i) to develop, characterize and validate in vitro olfactory cell monolayers and (ii) to study the transport of polymeric- and lipid-based NPs across these monolayers in order to estimate NP access into the brain using cell penetrating peptide (CPPs) moieties: Tat and Penetratin (Pen). All tested poly(d,l-lactide-co-glycolide) (PLGA) and nanostructured lipid carrier (NLC) formulations were stable in transport buffer and biocompatible with the olfactory mucosa cells. Nevertheless, 0.7% of PLGA NPs was able to cross the olfactory cell monolayers, whereas 8% and 22% of NLC and chitosan-coated NLC (CS-NLC) were transported across them, respectively. Moreover, the incorporation of CPPs to NLC surface significantly increased their transport, reaching 46% of transported NPs. We conclude that CPP-CS-NLC represent a promising brain shuttle via nose-to-brain for drug delivery.
Collapse
Affiliation(s)
- Oihane Gartziandia
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain; Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Susana Patricia Egusquiaguirre
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - John Bianco
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium; Integrated Center for Cell Therapy and Regenerative Medicine, International Clinical Research Center (FNUSA-ICRC), St. Anne's University Hospital Brno, Pekařská 53, Brno 656 91, Czech Republic
| | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Véronique Préat
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium.
| | - Ana Beloqui
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium.
| |
Collapse
|
198
|
Intravenous Administration of Cilostazol Nanoparticles Ameliorates Acute Ischemic Stroke in a Cerebral Ischemia/Reperfusion-Induced Injury Model. Int J Mol Sci 2015; 16:29329-44. [PMID: 26690139 PMCID: PMC4691110 DOI: 10.3390/ijms161226166] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/30/2015] [Accepted: 12/03/2015] [Indexed: 11/16/2022] Open
Abstract
It was reported that cilostazol (CLZ) suppressed disruption of the microvasculature in ischemic areas. In this study, we have designed novel injection formulations containing CLZ nanoparticles using 0.5% methylcellulose, 0.2% docusate sodium salt, and mill methods (CLZnano dispersion; particle size 81 ± 59 nm, mean ± S.D.), and investigated their toxicity and usefulness in a cerebral ischemia/reperfusion-induced injury model (MCAO/reperfusion mice). The pharmacokinetics of injections of CLZnano dispersions is similar to that of CLZ solutions prepared with 2-hydroxypropyl-β-cyclodextrin, and no changes in the rate of hemolysis of rabbit red blood cells, a model of cell injury, were observed with CLZnano dispersions. In addition, the intravenous injection of 0.6 mg/kg CLZnano dispersions does not affect the blood pressure and blood flow, and the 0.6 mg/kg CLZnano dispersions ameliorate neurological deficits and ischemic stroke in MCAO/reperfusion mice. It is possible that the CLZnano dispersions will provide effective therapy for ischemic stroke patients, and that injection preparations of lipophilic drugs containing drug nanoparticles expand their therapeutic usage.
Collapse
|
199
|
Tibbitts J, Canter D, Graff R, Smith A, Khawli LA. Key factors influencing ADME properties of therapeutic proteins: A need for ADME characterization in drug discovery and development. MAbs 2015; 8:229-45. [PMID: 26636901 PMCID: PMC4966629 DOI: 10.1080/19420862.2015.1115937] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein therapeutics represent a diverse array of biologics including antibodies, fusion proteins, and therapeutic replacement enzymes. Since their inception, they have revolutionized the treatment of a wide range of diseases including respiratory, vascular, autoimmune, inflammatory, infectious, and neurodegenerative diseases, as well as cancer. While in vivo pharmacokinetic, pharmacodynamic, and efficacy studies are routinely carried out for protein therapeutics, studies that identify key factors governing their absorption, distribution, metabolism, and excretion (ADME) properties have not been fully investigated. Thorough characterization and in-depth study of their ADME properties are critical in order to support drug discovery and development processes for the production of safer and more effective biotherapeutics. In this review, we discuss the main factors affecting the ADME characteristics of these large macromolecular therapies. We also give an overview of the current tools, technologies, and approaches available to investigate key factors that influence the ADME of recombinant biotherapeutic drugs, and demonstrate how ADME studies will facilitate their future development.
Collapse
|
200
|
Piotrowski M, Jantas D, Szczepanowicz K, Łukasiewicz S, Lasoń W, Warszyński P. Polyelectrolyte-coated nanocapsules containing undecylenic acid: Synthesis, biocompatibility and neuroprotective properties. Colloids Surf B Biointerfaces 2015; 135:8-17. [DOI: 10.1016/j.colsurfb.2015.07.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 05/28/2015] [Accepted: 07/13/2015] [Indexed: 02/08/2023]
|