Nanotherapeutic Strategies and New Pharmaceuticals: Part II -  - E-Book

Nanotherapeutic Strategies and New Pharmaceuticals: Part II E-Book

0,0
61,06 €

-100%
Sammeln Sie Punkte in unserem Gutscheinprogramm und kaufen Sie E-Books und Hörbücher mit bis zu 100% Rabatt.
Mehr erfahren.
Beschreibung

Advances in technology have enabled medicinal chemists to discover and formulate several highly specific, biocompatible, and non-toxic therapeutic agents for clinical applications. Nanotechnology has achieved significant progress in the last few decades and is crucial in every field of science and technology. Nanotechnology-based pharmaceuticals offer multifaceted and alternative methodologies in comparison to the limitations of many conventional clinical therapies. Expertise in designing and developing nanoformulations has helped in targeted drug delivery. Recently, the use of innovative therapeutic agents, particularly in nanomedicine, has accounted for a significant portion of the global pharmaceutical market and is predicted to continue to grow rapidly in the near future. Nanotherapeutic Strategies and New Pharmaceuticals is an accessible multi-part reference which informs the reader about several new techniques based on nanotechnology. The chapters explain relevant topics in detail. The book is designed to encourage and help undergraduate, graduate and post-graduate students in the field of nanotherapeutics, pharmaceuticals and bio-organic chemistry through the use of didactic language and simple illustrations. Part 2 of this book covers the potential of nanotherapeutics and natural therapies for treating neurological diseases, targeting ion channels, signal transduction therapy, gene therapy of single gene mutation diseases and for nanoformulations for special purposes such as wound healing and stimuli-responsive drug delivery. The book also features a chapter that summarizes the types of nanoparticles tailored for specific molecular targets that mediate different diseases. The book set serves as a textbook for students in pharmacology and medical biochemistry, as well as a quick reference for researchers on bio-organic chemistry, as well as general readers interested in nanomedicine.

Sie lesen das E-Book in den Legimi-Apps auf:

Android
iOS
von Legimi
zertifizierten E-Readern

Seitenzahl: 534

Bewertungen
0,0
0
0
0
0
0
Mehr Informationen
Mehr Informationen
Legimi prüft nicht, ob Rezensionen von Nutzern stammen, die den betreffenden Titel tatsächlich gekauft oder gelesen/gehört haben. Wir entfernen aber gefälschte Rezensionen.



Table of Contents
BENTHAM SCIENCE PUBLISHERS LTD.
End User License Agreement (for non-institutional, personal use)
Usage Rules:
Disclaimer:
Limitation of Liability:
General:
PREFACE
About the Editors
List of Contributors
Nanotherapeutics for Treatment of Neurological Disorders
Abstract
1. INTRODUCTION
2. TRANSPORTATION MECHANISM
2.1. Paracellular Transportation
2.2. Transcellular Passive Diffusion
2.2.1. Transporters
2.2.2. Transcytosis
3. NPs TYPES
3.1. Lipid-Based NPs
3.2. Polymeric NPs
3.3. Inorganic NPs
4. NEUROLOGICAL DISORDERS AND TREATMENT STRATEGIES
4.1. Parkinson’s Disease
4.2. Alzheimer’s Disease
4.3. Glioblastoma
4.4. Vascular Occlusion
5. BIOLOGICAL BARRIERS FOR DRUG DELIVERY
CONCLUDING REMARKS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Molecular Mechanism of Therapeutic Actions of Some Nanoparticles in Some Diseases
Abstract
1. INTRODUCTION
2. THE MOLECULAR MECHANISMS OF THERAPEUTIC ACTION OF NPS IN SOME DISEASES
2.1. Molecular Mechanism of Anticancer Activities of NPs
2.1.1. Molecular Mechanism of Anticancer Activity of AgNPs
2.1.1.1. Ultrastructural Destruction of Cancer Cells
2.1.1.2. ROS Production
2.1.1.3. DNA Damage
2.1.1.4. Inactivation of Proteins and Regulation of Signaling Pathways
2.1.1.5. Inhibition of Migration and Angiogenesis
2.1.2. Molecular Mechanism of Anticancer Activity of AuNPs
2.1.3. Molecular Mechanism of Anticancer Activity of Iron Oxide NPs (FeO-NPs)
2.1.4. Molecular Mechanism of Anticancer Activity of Titanium Dioxide NPs
2.1.5. Molecular Mechanism of Anticancer Activity of Cerium Oxide NPs
2.1.6. Molecular Mechanism of Anticancer Activity of Zinc Oxide NPs
2.1.7. Molecular Mechanism of Anticancer Activity of Copper Oxide NPs
2.2. Molecular Mechanism of Anti-diabetic Activities of NPs
2.3. Molecular Mechanism of Anti-bacterial Activities of NPs
2.3.1. Molecular Mechanism of Antibacterial Activity of ZnO-NPs
2.3.1.1. Generation of Reactive Oxygen Species (ROS)
2.3.1.2. Zinc Ions (Zn2+) Release
2.3.2. Molecular Mechanism of Antibacterial Activity of AgNPs
2.3.3. Molecular Mechanism of Antibacterial Activity of AuNPs
2.3.4. Molecular Mechanism of the Antibacterial Activity of TiO2-NPs
2.3.5. Molecular Mechanism of Antibacterial Activity of NO-NPs
2.4. Molecular Mechanism of Anti-fungal Activities of Metallic NPs
2.5. Molecular Mechanism of Anti-viral Activities of NPs
2.6. Molecular Mechanism of Anti-inflammatory Activities of NPs
2.6.1. Anti-inflammatory Activity of ZnO-NPs
2.6.1.1. Blocking the Production of Pro-inflammatory Cytokines
2.6.1.2. Inhibiting Mast Cell Proliferation
2.6.1.3. Suppressing LPS Induced COX-2 Expression
2.6.1.4. Suppressing the Expression of iNOS Gene
2.6.2. Anti-inflammatory Activity of AgNPs
2.6.2.1. Reducing VEGF Levels
2.6.2.2. Decreasing (HIF)-1α Expression
2.6.2.3. Preventing Mucin Hypersecretion
2.6.2.4. Suppressing Pro-inflammatory Cytokine Production
2.6.3. Anti-Inflammatory Activity of AuNPs
2.6.3.1. Reducing ROS Production (or by Oxygen Radical Scavenging)
2.6.3.2. Decreasing LPS Induced Cytokine Production
2.6.3.3. Modulating MAPK and PI3K Pathways in Kuppfer and Hepatic Stellate Cells
2.6.4. Anti-inflammatory Activity TiO2NPs
2.6.4.1. Reducing Platelet Numbers
2.6.4.2. Increasing Thrombin-antithrombin Levels
2.6.5. Anti-inflammatory Activity of SeNPs
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
Acknowledgment
REFERENCES
Nanotherapeutics for the Treatment of Voltage Gated Ion Channels
Abstract
1. INTRODUCTION
1.1. Background and History of Ion-Channels
1.2. Ligand-gated Ion Channels Convert Chemical Signals into Electrical Activities
1.3. Biological Role of Ion-Channels
1.4. Inhibition of the Sodium-Potassium Pump Causes Delay in the Resting Membrane Potential Decaying Process
2. GREEN TECHNOLOGIES FOR STANDARDIZATION
3. DEVELOPMENT AND APPLICATION OF POLYMERIC SYSTEM CONTAINING HERBAL EXTRACTS
3.1. Microspheres
3.2. Hydrogels
3.3. Phytosomes (Phyto-Phospholipid Complex)
3.4. Emulsions
3.4.1. Drug Delivering Systems in Emulsion
3.4.1.1. Self Microemulsifying Drug Delivery System (SMEDDS)
3.4.1.2. Self Nanoemulsifying Drug Delivery System (S-SNEDDS)
3.5. Nanoparticles
3.5.1. Solid Lipid Nanoparticles
3.6. Nanospheres And Nanocapsules
3.6.1. Gold and Silver Nanoparticles
3.6.2. Injectable Nanoparticles
3.6.3. Liposomes
3.6.3.1. Substances Enhancing the Activity of Liposomes
3.6.3.2. Examples of Drugs Loaded to Liposomes
3.6.3.3. Limitation of Herbal Extract Loaded Liposomes
3.6.3.4. Pegylated Liposomes
3.6.3.5. Effectiveness of Glutathione Coating on Pegylated Liposomes
3.6.3.6. Effect of Sito-g on Liposomal Activity
3.6.3.7. Thermosensitive and Non-thermosensitive Pegylated Liposomes
3.7. Niosomes
3.7.1. Effect of pH on Niosomes
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Stimuli Responsive Hydrogels Composites for Control Drug Delivery
Abstract
1. INTRODUCTION
2. ROLE OF HYDROGEL IN DRUG DELIVERY
2.1. Physically CL Methods for Hydrogel Synthesis
2.1.1. CL by Charge Interaction
2.1.2. Hydrophobic Interactions
2.1.3. Hydrogen Bonding Interaction
2.1.4. Stereo Complexation
2.1.5. CL via Crystallization
2.2. Chemically CL
2.2.1. Covalently CL
2.2.2. CL by Small Molecules
2.2.3. Polymer-polymer CL
3. STIMULI SENSITIVE HYDROGELS AND ITS ROLE IN CONTROL DRUG DELIVERY
3.1. Light Responsive Hydrogels
3.2. pH Sensitive Hydrogels
3.3. Thermo Responsive Hydrogels
3.4. Electric Signal Stimulated Hydrogels
3.5. Glucose Stimulated Hydrogels
4. MAGNETIC HYDROGELS NANO CT FOR CONTROL DRUG RELEASE
5. CHITOSAN BASED CT HYDROGELS
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGMENT
REFERENCES
Polymeric Nanofibers for Wound Dressing Applications
Abstract
1. INTRODUCTION
2. Anatomy of Skin
3. Wound
3.1. Classification of Wounds
3.2. Wound Healing
3.2.1. Stages of Wound Healing
3.2.1.1. Homeostasis Stage
3.2.1.2. Inflammation Stage
3.3.1.3. Proliferation Stage
3.2.1.4. Remodelling Stage
3.3. Wound Dressing
3.3.1. Classification of Wound Dressing
3.3.1.1. Passive Dressing
3.3.1.2. Interactive Dressing
3.3.1.3. Advanced Dressing
3.3.1.4. Smart Dressing
3.4. Significance of Wound Dressing
4. Antimicrobial Activity Imparting NFs
5. Fabrication of NFs
6. Strategies to Load Antimicrobial Drug on Electrospun NFs
6.1. Blending
6.2. Core-Sheath Fabrication
6.3. Encapsulation
6.4. Post-treatment
6.5. Attachment
7. Characterization Techniques
7.1. In-vitro Testing
7.2. In-vivo Testing
7.3. Assessment of Antibacterial Activity
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Nano-therapeutics of Flavonoids-loaded Polymeric Drug Delivery Systems
Abstract
1. INTRODUCTION
2. MEDICINAL PLANTS PHYTOCHEMICALS
2.1. Flavonoids and its Properties
3. RECENT APPROACHES OF DRUG DELIVERY SYSTEM FOR HERBAL BIOACTIVE SUBSTANCES
3.1. Delivery Systems for Enhanced Therapeutic Efficacy and Bioavailability of Flavonoids
3.2. NPs Based Delivery of Flavonoids
3.3. Polymeric Micelles-based Delivery of Flavonoids
3.4. Liposomal Based Delivery of Flavonoids
3.5. Self Microemulsifying, Micro-emulsions and Nano-emulsions Based Delivery of Flavonoids
3.6. Solid Lipid NPs (SLNS) Based Delivery of Flavonoids
3.7. Nanostructured Lipid Carriers (NLCS) Based Delivery of Flavonoids
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
ADHD Comorbid to Substance Use Disorder: A Review of Genetics, Neurobiology, Brain Circuitry, and Nanotherapeutics
ABSTRACT:
1. INTRODUCTION
1.1. ADHD, Substance Abuse, and Stimulant Therapy
1.2. Risk Factors for ADHD
1.2.1. Genetic Susceptibility to Comorbid Attention-Deficit/Hyperactivity Disorder and Heroin Addiction
1.2.2. Stress and Mood Reactivity in Addiction
1.2.3. Molecular Genetic Studies
1.2.4. Clinical Manifestation of ADHD with Heroin Addiction
1.3. Behavioral and Brain Functions: An Insight from Animal ADHD Models
1.3.1. Animal Models of ADHD
1.3.1.1. DopamineBased ADHD Models
1.3.1.2. Nor-Epinephrine Based ADHD Models
1.3.1.3. Serotonin Based ADHD Models
1.3.1.4. Glutamate Based ADHD Models
1.3.1.5. Psycho-Stimulants Based ADHD Models
1.3.1.6. 6-hyroxydopamine (6-OHDA) Lesioned ADHD Model
1.3.1.7. Dopamine transporter-knock out (DAT-KO) ADHD Model
1.3.1.8. Coloboma Mutant ADHD Model
1.3.1.9. Poor 5-CSRTT task performer ADHD Model
1.3.1.10. Anoxia in Neonatal Rats
1.3.1.11. The Insight Provided by Animal Models of ADHD
2. NANOTHERAPEUTICS FOR ADHD: CHALLENGES, OPPORTUNITIES & PROGRESS
2.1. Ginseng
2.2. Ginkgo Biloba
2.3. Gotu Kola
2.4. Guarana
2.5. Bacopa Monnier
2.6. Rhodiola
2.7. Other Herbal Treatments
CONCLUSION
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Sodium Channelopathies and Novel Viral/non-viral Vectors for their Gene Therapy
Abstract
1. INTRODUCTION
1.1. Chemogenetics Vs Optogenetics
2. GENE EDITING
3. TYPES OF DIFFERENT VECTOR SYSTEMS FOR GENE DELIVERY
3.1. Viral Delivery System
3.1.1. Retroviral Vector
3.1.2. Adenoviral Vector
3.1.3. Adeno-associated Vector
3.1.4. Adenoviral Hybrid Vectors
3.1.5. Herpes Simplex Vectors
3.1.6. POX Virus Vector
3.1.7. Lentiviruses Vector
3.1.8. Epstein- Barr Virus Vector
3.2. Non-Viral Delivery System
3.2.1. Physical Methods of Non-Viral Gene Delivery
3.2.1.1. Naked DNA
3.2.1.2. DNA Particle Bombardment by Gene GUN
3.2.1.3. Electroporation
3.2.1.4. Hydrodynamic
3.2.1.5. Ultrasound
3.2.1.6. Magnetofection
3.3. Chemical Non-Viral Delivery Systems
3.3.1. Lipid Polymer Hybrid
3.3.2. Cationic Liposomes
CONCLUSION
Future Perspectives
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Gene Therapy and Editing for the Treatment of Single-Gene Pain Disorders
Abstract
1. INTRODUCTION
2. THE FUNCTION OF ION CHANNELS IN NOCICEPTORS
3. TOXINS AND PHARMACEUTICALS AS TARGETS
4. TARGETING NAV CHANNELS: LOCAL ANESTHETICS STATE-DEPENDENT ACTION
5. MECHANISM OF 6 COUPLINGS (EC)
6. ION-SELECTIVITY OF VGSC
7. PAIN INSENSITIVITY IN CONGENITAL DISORDERS
8. HSAN TYPE IID RESULTING FROM MUTATIONS IN INACTIVATING SCN9A
9. THE GAIN OF FUNCTIONAL GENE VARIANT RELATED TO HERITABLE PAIN CONDITIONS
9.1. Inherited Erythromelalgia
9.2. The Paroxysmal Extreme Pain-disorder
9.3. Small-fiber Neuropathy
9.4. Familial Episodic Pain and TRAP1 Mutation
9.5. Hereditary-Insensitivity to Pain
9.6. Innate-pain Insensitivity and SCN9 Amutations Inactivating
9.7. SCN11A Mutation and Familial-episodic Pain-syndrome
10. BIOPHYSICS OF PAINLESS AND PAINFUL CHANNELOPATHIES
11. TREATMENTS FOR THE PAIN CAUSED BY SODIUM CHANNELOPATHIES
CONCLUSIONS
CONSENT FOR PUBLICATION
CONFLICT OF INTEREST
ACKNOWLEDGEMENTS
REFERENCES
Nanotherapeutic Strategies and
New Pharmaceuticals
(Part 2)
Edited by
Shahid Ali Khan
Department of Chemistry
University of Swabi
Swabi Anbar-23561
Khyber Pakhtunkhwa
Pakistan
Saad Salman
The University of Lahore
Islamabad Campus
Islamabad-44000
Pakistan
&
Youssef O. Al-Ghamdi
Department of Chemistry
College of Science Al-Zulfi
Majmaah University
Al-Majmaah 11952
Saudi Arabia

BENTHAM SCIENCE PUBLISHERS LTD.

End User License Agreement (for non-institutional, personal use)

This is an agreement between you and Bentham Science Publishers Ltd. Please read this License Agreement carefully before using the book/echapter/ejournal (“Work”). Your use of the Work constitutes your agreement to the terms and conditions set forth in this License Agreement. If you do not agree to these terms and conditions then you should not use the Work.

Bentham Science Publishers agrees to grant you a non-exclusive, non-transferable limited license to use the Work subject to and in accordance with the following terms and conditions. This License Agreement is for non-library, personal use only. For a library / institutional / multi user license in respect of the Work, please contact: [email protected].

Usage Rules:

All rights reserved: The Work is the subject of copyright and Bentham Science Publishers either owns the Work (and the copyright in it) or is licensed to distribute the Work. You shall not copy, reproduce, modify, remove, delete, augment, add to, publish, transmit, sell, resell, create derivative works from, or in any way exploit the Work or make the Work available for others to do any of the same, in any form or by any means, in whole or in part, in each case without the prior written permission of Bentham Science Publishers, unless stated otherwise in this License Agreement.You may download a copy of the Work on one occasion to one personal computer (including tablet, laptop, desktop, or other such devices). You may make one back-up copy of the Work to avoid losing it.The unauthorised use or distribution of copyrighted or other proprietary content is illegal and could subject you to liability for substantial money damages. You will be liable for any damage resulting from your misuse of the Work or any violation of this License Agreement, including any infringement by you of copyrights or proprietary rights.

Disclaimer:

Bentham Science Publishers does not guarantee that the information in the Work is error-free, or warrant that it will meet your requirements or that access to the Work will be uninterrupted or error-free. The Work is provided "as is" without warranty of any kind, either express or implied or statutory, including, without limitation, implied warranties of merchantability and fitness for a particular purpose. The entire risk as to the results and performance of the Work is assumed by you. No responsibility is assumed by Bentham Science Publishers, its staff, editors and/or authors for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products instruction, advertisements or ideas contained in the Work.

Limitation of Liability:

In no event will Bentham Science Publishers, its staff, editors and/or authors, be liable for any damages, including, without limitation, special, incidental and/or consequential damages and/or damages for lost data and/or profits arising out of (whether directly or indirectly) the use or inability to use the Work. The entire liability of Bentham Science Publishers shall be limited to the amount actually paid by you for the Work.

General:

Any dispute or claim arising out of or in connection with this License Agreement or the Work (including non-contractual disputes or claims) will be governed by and construed in accordance with the laws of Singapore. Each party agrees that the courts of the state of Singapore shall have exclusive jurisdiction to settle any dispute or claim arising out of or in connection with this License Agreement or the Work (including non-contractual disputes or claims).Your rights under this License Agreement will automatically terminate without notice and without the need for a court order if at any point you breach any terms of this License Agreement. In no event will any delay or failure by Bentham Science Publishers in enforcing your compliance with this License Agreement constitute a waiver of any of its rights.You acknowledge that you have read this License Agreement, and agree to be bound by its terms and conditions. To the extent that any other terms and conditions presented on any website of Bentham Science Publishers conflict with, or are inconsistent with, the terms and conditions set out in this License Agreement, you acknowledge that the terms and conditions set out in this License Agreement shall prevail.

Bentham Science Publishers Pte. Ltd. 80 Robinson Road #02-00 Singapore 068898 Singapore Email: [email protected]

PREFACE

Dr. Shahid Ali Khan

The field of nanotechnology evolved as a discipline and is not a mere specialization. It requires multiple fields such as engineering, physics, chemistry, mathematics, medicine, and pharmacy to be integrated. This book provides a quick review of the practical aspects of these diverse arenas. Advances in nanotechnology have increased the feasibility to tailor the functional modalities that assist in targeting selective biological barriers for drug delivery and other biomedical applications. This book provides knowledge to the students interested in nanotechnology research. This book paves a path between pharmacy and nanoscience while striking an equilibrium between approachability and depth.

All the editors of this book have research collaborations with various local and international universities in the field of nanoscience. They had supervised M. Phil and Ph.D. students in the field of nanoscience and pharmaceutical technology. They had published manuscripts on nanotechnology in Advanced Drug Delivery Reviews and other prestigious journals.

Phoebe’s chapter discusses the role of the blood-brain barrier comprising of a highly selective semipermeable border of epithelial cells that shield the brain from substances that impede the transportation of drug delivery used to treat various neurological disorders. Bello et al. reported the molecular mechanisms underlying silver, gold, Iron Oxide, Titanium Dioxide, Cerium Oxide, Zinc Oxide, Nitric oxide-releasing nanoparticles’ therapeutic action in cancer, diabetes, bacterial, fungal, viral and inflammatory diseases. The mechanisms of anticancer activity of the nanoparticles ranging from ultra-structure disruption, generation of reactive oxygen species (ROS), induction of DNA damage, inactivation of proteins that regulate signaling pathways, inhibition of migration and angiogenesis as well as induction of apoptosis are debated. Fahad et al. described the delivery of plant-derived nanoparticles comprised of nano-hydrogels, emulsions, and liposomes to targeted sites for disorders of voltage-gated channels. Zubair et al. explained the synthesis techniques pertaining to polymeric injectable hydrogels to reach safely to the targeted site. Abid et al. elucidated the antimicrobial drug-loaded polymeric nanofibers for wound dressing. The technology such as electrospinning and characterization of nanofibers for the drug release, shape, surface quality, ability to endure mechanical shocks, antimicrobial activity, and in vivo wound healing effectiveness are also discussed. Fazle et al. discussed the techniques regarding the synthesis, characterization, and biosafety of flavonoid-loaded polymeric nanoparticles, liposomes, matrix systems, and micro-emulsions by ameliorating their pharmacological activity and reducing the side effects. Jawaria and Fahad et al., in the subsequent two chapters, had described the molecular mechanisms underlying the channelopathies caused by various genetic or acquired factors. Different neurological diseases such as migraine, epilepsy, small fiber neuropathy, paroxysmal pain disorder, dravet syndrome, and congenital insensitivity to pain are explained, and their gene therapy and editing are discussed.

The chapters of this book are written by scientists and researchers of specialized fields and overtly different scientific backgrounds, but everything boiled down to one common goal – Nanoscience. The tremendous consequence of the combined effort led to this book, “Nanotherapeutic strategies and new pharmaceuticals Part 2.”

Shahid Ali Khan Department of Chemistry University of Swabi Swabi Anbar-23561 Khyber Pakhtunkhwa PakistanSaad Salman The University of Lahore Islamabad Campus Islamabad-44000 PakistanYoussef O. Al-Ghamdi Department of Chemistry College of Science Al-Zulfi, Majmaah University,

About the Editors

Shahid Ali Khan

Shahid Ali Khan (Editor)

Shahid Ali Khan did his master of science (MSC) in 2010 in organic chemistry from Kohat University of Science and Technology, and Master of Philosophy (MPhil) in Natural Product and Medicinal Chemistry in 2014 from ICCBS Karachi and PhD in Nanocatalysis in 2017 from King Abdulaziz University, Saudi Arabia. Dr. Khan got the best PhD thesis award in natural sciences, awarded by King Abdulaziz University. Dr. Khan joined the University of Swabi as Assistant Professor in Feb. 2018 and still working in the same University. During his stay at the University of Swabi, Dr. Khan has supervised many MPhil, MSC and BS students. Currently, many students of Dr. Khan's research group are involved in the synthesis and designing of nanocatalyst and hydrogels for various technological applications. . Moreover, Dr. Khan is working on multi-dimensional projects including Natural product chemistry, medicinal chemistry, Nanocatalysis and self-assembled hydrogels for diverse applications. Dr. Khan is also involved in the designing and synthesis of thin films as solid support for the stabilization of zero-valent metal nanoparticles. Dr. Khan is teaching various courses in Organic Chemistry, Physical chemistry, and spectroscopy to BS, MS and PhD students.

In brief, Dr. Khan has published 62 publications with a cumulative impact factor of 260 and published 5 book chapters in various international journals of high repute.

Dr. Khan edited the book entitled “Nanotherapeutics strategies and new pharmaceuticals Part 1.

Saad Salman (Co-Editor)

Saad Salman is a Lecturer at the Department of Pharmacy, The University of Lahore (Islamabad Campus), teaching Undergraduate and Postgraduate students. He is a Certified Health Researcher from NIH, USA. He has over twenty-one International publications, two International books, two book chapters, and thirty abstracts published. He is the founder and CEO of Research and Publication Inc. provides solutions to research students. He had received numerous awards at various conferences. He is an active member of the American Society of Pharmacology and Experimental Therapeutics (ASPET), the American Pharmacist Association, and American Society of Consultant Pharmacists.

Youssef O. Al-Ghamdi (Co-Editor)

Youssef O. Al-Ghamdi received a bachelor’s degree in Chemistry from Umm AL-Qura University, Makkah, Saudi Arabia, in 2008. Dr. Alghamdi received his master’s degree in Organic Chemistry from King Khalid University, Abha, Saudi Arabia, in 2012, and the Ph.D. degree in Organic Chemistry, Polymers King Abdualaziz University, Jeddah, Saudi Arabia in 2018. He joined the College of Science at Al-Zulfi branch, Majmaah University, as the head of the chemistry department. Dr. Alghamdi heading several units such as Planning and Development, and Unit of Scientific Affairs. He is a member of the College Council, College of Science, Al-Zulfi, Majmaah University and a member of Labs and Facilities. Dr. Youssef O. Al-Ghamdi is involved in teaching various courses in Organic Chemistry, to BS and he has published 20 publications.

List of Contributors

Abid Mehmood YousafDepartment of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, PakistanAmna NisarDepartment of Pharmacy, University of Peshawar, Peshawar, 25120, Khyber Pakhtunkhwa, PakistanBello Aminu BelloDepartment of Biochemistry, Federal University Dutse, P.M.B.7156 Dutse, Jigawa State, NigeriaFahad Hassan ShahDepartment of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju 32588, Republic of KoreaFahimUllah KhanDepartment of Pharmacy, City University of Science & Information Technology Peshawar, Peshawar, 25000, Khyber Pakhtunkhwa, PakistanFakhar Ud DinDepartment of Pharmacy, Quaid-i-Azam University, Islamabad 45320, PakistanFakhra SibtainThe University of Lahore, Islamabad Campus, Islamabad-44000, PakistanFatima Sulaiman AbdullahiDepartment of Food Science and Technology, Federal University Dutsin-MA, P.M.B. 5001, Katsina State , NigeriaFazle RabbiDepartment of Pharmacy, Abasyn University Peshawar, Peshawar, 25000, Khyber Pakhtunkhwa, PakistanHassan A. HemegDepartment of Medical Laboratories Technology, Faculty of Applied Medical Sciences, Taibah University, Medina, 30001, Kingdom of Saudi ArabiaIbrahim Khalil AdamDepartment of Biochemistry, Federal University Dutse, P.M.B.7156 Dutse, Jigawa State, NigeriaIkram Ullah KhanDepartment of Pharmaceutics, Government College University, Faisalabad-38000, PakistanImad AhmadDepartment of Pharmacy, Abasyn University Peshawar, Peshawar, 25000, Khyber Pakhtunkhwa, PakistanJawaria IdreesDepartment of Biochemistry, Lady Reading Hospital, Peshawar, KPK, 25000, PakistanLaiba WasimThe University of Lahore, Islamabad Campus, Islamabad-44000, PakistanLuqman ShahDepartment of Biochemistry, Hazara University Mansehra, Mansehra, Khyber Pakhtunkhwa, PakistanMaham ChaudryUniversity of Lahore, Islamabad Campus, Islamabad-44000, PakistanMehreen BashirDepartment of Pharmaceutics, Government College University, Faisalabad-38000, PakistanMuhammad Usman GhoriDepartment of Pharmacy, School of Applied Science, University of Huddersfield, Huddersfield, HD1 3DH, United KingdomMuniba TariqThe University of Lahore Islamabad Campus, Islamabad-44000, PakistanNoureen KhanDepartment of Chemistry, Sardar Bahadur Khan Women’s University, Quetta, PakistanPhoebe WilsonScience Advisory Board, 500 Women Scientists, United StatesQurrat ul Ain YousafiDepartment of Neurosurgery, District Headquarters Hospital, Rawalpindi, 46000, PakistanSaad SalmanDepartment of Pharmaceutics, Government College University, Faisalabad-38000, Pakistan The University of Lahore Islamabad Campus, Islamabad-44000, PakistanSajid AsgharDepartment of Pharmaceutics, Government College University, Faisalabad-38000, PakistanSani S. UsmanDepartment of Biological Sciences Gombe, Federal University Kashere, P.M.B 0182 Kashere, Gombe, NigeriaShahid Ali KhanDepartment of Chemistry, University of Swabi, Khyber Pakhtunkhwa, 23561, PakistanShanza NageenThe University of Lahore, Islamabad Campus, Islamabad-44000, PakistanSifeng Lucy ChenDepartment of Life Sciences, Imperial College London, London, United KingdomSong Ja KimDepartment of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju 32588, Republic of KoreaSyed A. A. RizviHampton School of Pharmacy, Hampton University, Virginia, 23669, United StatesSyed Haroon KhalidDepartment of Pharmaceutics, Government College University, Faisalabad-38000, PakistanSyed Muhammad Mukarram ShahDepartment of Pharmacy, University of Swabi, Anbar-23561, Khyber Pakhtunkhwa, PakistanTalib HussainDepartment of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, PakistanWaseeq Ur RehmanDepartment of Chemistry, Govt. Postgraduate College, Nowshera 24100, Khyber Pakhtunkhwa, PakistanYahaya Saidu GwarzoDepartment of Biochemistry, Federal University Dutse, P.M.B.7156 Dutse, Jigawa State, NigeriaYasir AnwarDepartment of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Kingdom of Saudi ArabiaYasser ShahzadDepartment of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, PakistanYoussef O. Al-GhamdiDepartment of Chemistry, College of Science Al-zulfi, Majmaah University, Al-Majmaah, 11952, Saudi ArabiaZubair AhmadDepartment of Chemistry, University of Swabi, Anbar-23561, Khyber Pakhtunkhwa, PakistanZunaira AfzalDepartment of Pharmaceutics, Government College University, Faisalabad-38000, Pakistan

Nanotherapeutics for Treatment of Neurological Disorders

Phoebe Wilson1,*
1 Science Advisory Board, 500 Women Scientists, United States

Abstract

Our brains are undisputedly regarded as one of the most complex biological structures, therefore it is not surprising that there are challenges associated with the transportation of therapeutic agents across this organ. This may be attributed in large part to the blood-brain barrier (BBB), which maintains a very stable environment in order to sustain normal brain function. The blood-brain barrier is comprised of a highly selective semipermeable border of epithelial cells that shield the brain from unwelcome and invasive substances. It is so effective, however, that it impedes the transportation of drug delivery used to treat various neurological and cerebrovascular disorders, such as Parkinson’s Disease (PD), Alzheimer’s Disease (AD), stroke, and gliomas (tumors in the brain and spinal cord). Consequently, many central nervous system disorders are undertreated. Significant advances in nanotechnology have increased the feasibility for biomedical applications to the brain, as nanopharmaceuticals may be tailored with functional modalities that assist to target selective brain tissue.

Keywords: Blood-brain barrier, Nanomedicine, Nanotherapeutics, Neurological disorders, Neurovascular, Targeted drug delivery.
*Corresponding author Phoebe Wilson: Science Advisory Board & 500 Women Scientists, USA; Email:[email protected]

1. INTRODUCTION

There is ample evidence to suggest that neurological disorders exist as one of the greatest threats to public health, with recent studies crediting them as the second leading cause of deaths [1]. The increase in patients diagnosed with disorders is contested by the growing demand for effective treatments, which are met with their own obstacles. One of the largest challenges for delivering therapeutic molecules is their inability to breach the blood-brain barrier (BBB). The BBB is formed as astrocytes wrap their “feet” around capillaries in the brain. The tight junctions situated between epithelial cells in the capillary wall, accompanied by the covering comprised of foot-like extensions of the astrocytes, form a barrier

that regulates the passage of most ions and molecules between the blood and the brain tissue.

If they were to traverse the brain freely, ions such as sodium (Na+) and potassium (K+) could hinder the transmission of nerve impulses. Water, glucose, oxygen, carbon dioxide and small, lipid-soluble molecules are able to diffuse across the barrier with ease. Delivery drugs need to be constructed with optimal lipid solubility in mind, however, this is not a simple feat. By increasing the lipophilicity of the drug through chemical modification, there is a potential risk for decreased systematic solubility, and so the desired pharmacokinetic result may not be obtained.

The introduction of nanotechnology and nanoscience has served as the driving force for developing new strategies to treat neurological conditions. Their ability to penetrate the blood-brain barrier is in large part due to their malleable nature, as they can be modelled into different morphological structures in order to reach their constituent targets. The size of these NPs often resembles biomolecules, which plays a key role in drug targeting. The basis for nanotechnological drug delivery calls for the use of a nanoscopic scale (or nanoscale) and a therapeutic agent, which serve to function as the nanocarrier and the ‘consignment’, respectively [2]. Both systems’ properties are contingent upon whether the drug absorbs into or conjugates to the external surface of the nanoparticle, or instead is enclosed within [2]. These modifications help to supplement blood-brain barrier entry and disease-targeting efficiency [3].

2. TRANSPORTATION MECHANISM

Although the blood-brain barrier provides an impermeable border to particular solutes, brain capillary endothelial cells are able to assist the transcapillary exchange of others. Vital substances such as glucose are able to pass through the barrier in order to facilitate the generation of adenosine triphosphate (ATP) and neurotransmitters. Molecules are able to enter the brain tissue in a paracellular manner, by means of passive diffusion or via solute carriers and vesicular transport, as exhibited in Fig. (1) [4].

2.1. Paracellular Transportation

Paracellular transport is characterized by the transfer of substances between adjacent epithelial cells [5]. Smaller, hydrophilic materials can passively penetrate the blood-brain barrier through paracellular pathways, while large molecules are restricted due to the tight junctions that are present [6]. For that reason, the majority of peptides, proteins, and other macromolecules are inhibited from traversing through. Consequently, many problems have arisen from synthesizing these molecules for oral absorption and delivery [5, 6].

Fig. (1)) Transport of substances from blood to brain via several routes; paracellular (a), transcellular (b), transport proteins (c), efflux pumps (d), receptor-mediated transcytosis (e), adsorptive transcytosis (f) and cell-mediated transcytosis (g).

2.2. Transcellular Passive Diffusion

Transcellular passive enables the interaction between small, hydrophobic molecules and the endothelium of the blood-brain barrier [4, 7]. Drug molecules are able to passively diffuse into the cellular membrane via transcellular diffusion. Unfortunately, not all small-scale and hydrophobic molecules are able to diffuse across the endothelial layer, thus prompting further research [8].

2.2.1. Transporters

Transporters are able to assist drug molecules throughout the course of receptor-mediated transcellular crossing [6], as they would otherwise be unable to progress through the blood-brain barrier [4]. In recent years, transporters have aided the bioavailability of nanotherapeutics (NTs) via both oral and non-oral distribution and together with NPs, can vastly improve the overall efficacy of drug delivery [9].

2.2.2. Transcytosis

Transcytosis is most commonly observed in epithelial cells and involves the transportation of macromolecules across the interior of the cell. Unlike other transport mechanisms, transcytosis facilitates larger molecules that ordinarily struggle to cross the blood-brain barrier [4]. The system is broken down into multiple steps: endocytosis, vesicular transferral, and exocytosis, and is able to operate in an adsorptive-mediated or receptor-mediated manner 10. Adsorptive-mediated transcytosis is present for positively-charged ions and macromolecules located on the endothelial cells’ periphery, whereas receptor-mediated transcytosis relies on one frequent pathway for transportation 4. Both methods have been utilised as a means for the successful delivery of NPs across the blood-brain barrier [4, 6].

3. NPs TYPES

There are several variations of NPs types, each of which boast their own unique pharmacodynamic characteristics. General guidelines recommend that NPs be non-toxic and biocompatible, while maintaining a diameter less than 100 nanometers (unless it is involved in cell-mediated transport) [11]. Any larger size may compromise the efficacy of the NPs due to the restricted nature of the extracellular space [4, 10]. Additionally, NTs should maintain limited aggregation or dissociation and prompt minimal drug alterations such as biochemical degradation. This section provides an overview of contrasting NPs types (Fig. 2-B), and inorganic NPs (Fig. 2-C).

Fig. (2)) Schematic illustration of various polymeric nanoparticles, inorganic nanoparticles, and lipid-based nanoparticles.

3.1. Lipid-Based NPs

Lipid-based NPs are incredibly stable drug carriers that possess minimal toxicity when administered in vivo [12]. For that reason, they are regarded as one of the most promising candidates for successful drug delivery. Two of the most common types are solid lipid NPs (SLNs) and liposomes. Solid lipid NPs are composed of a solid lipid core that sustains a solid form both at room temperature and human body temperature [12]. Additionally, they have the capacity to absorb and disperse drugs, making them highly biocompatible and increasing their overall drug trapping capabilities [13]. Overall, SLNs provide many advantages, such as the ability to issue controlled drug release over a prolonged time period [14] (i.e. several weeks) as a result of the increased mass transfer resistance provided by the lipid’s solid structure [12, 13]. A recent application of solid lipid NPs uncovered its ability to enter a brain tumor when delivering resveratrol, a drug used to treat cancer [4, 15].

Liposomes are comprised of spherical sacs that encompass phospholipid molecules. Cholesterol is also commonly included in the fabrication of liposomes, as it has been demonstrated to contribute to the NPs’ stability in-vivo [16]. Liposomes range in size and lamellae count, and therefore classified under varying subcategories. Small unilamellar vesicles (SUVs) span up to 100 nanometers with one lipid bilayer, large unilamellar vesicles (LUVs) exceed 100 nanometers with one lipid bilayer, and multilamellar vesicles (MLVs) frequently surpass 500 nanometers in diameter and involve multiple concentric bilayers (Fig 3).

Fig. (3)) Classification of liposomes based on the lamellarity: Multilamellar Vesicles (MLV) are composed of many lipid bilayers and ranges from 1-5 µm in size. Large Unilamellar Vesicles (LUV) are in the size range of 100-250 nm with single lipid bilayer. Small Unilamellar Vesicles (SUV) consist of a single phospholipid bilayer surrounding the aqueous phase with a size range of 20-100 nm.

Liposomal delivery of anticancer medication was the first nanotherapeutic method to be approved for cancer treatment by the Food and Drug Administration (FDA) [14]. Through modification of the liposome surface, it has demonstrated extreme effectiveness in breaching the blood-brain barrier. This is attributable to its likeness to the lipid bilayer of the endothelial cell membrane itself [17]. Liposomes ordinarily adopt either receptor-mediated transcytosis (RMT), or adsorptive-mediated transcytosis (AMT) to transport drug delivery across the blood-brain barrier.

3.2. Polymeric NPs

Polymeric NPs are subdivided into polymeric micelles, dendrimers, nanocapsules, and nanospheres. Micelles make up a collection of amphiphilic molecules that are dispersed in an aqueous solution [18]. Research has found micelles to be successful in crossing the blood-brain barrier in human astrocyte cell culture when conjugated to transcriptional activators peptides [19]. This same peptide has helped to facilitate selective brain penetration through adsorptive-mediated transcytosis (AMT), helping to concentrate the NPs within the astrocyte and around the neuron’s nucleus [19, 20]. Further research has affirmed the benefits of micelles usage, including the enhanced transportation of a drug into functional tissues including the caudate putamen, hippocampus, substantia nigra, and cortical layer of the brain [21].

Dendrimers are unique in that they consist of three domains: the core, radially concentric interior shells, and a multivalent exterior. Drug molecules are able to attach to the surface covalently to form dendrimer prodrugs or can become encapsulated internally through supramolecular formation [22]. Dendrimers are highly beneficial for transporting drugs on account of their low toxicity, high loading capabilities, and water solubility [23], which makes them easily malleable in response to their environment. Additionally, they have been adopted as a favourable method for penetrating cells within the neurovascular unit (NVU) through both neurosurgical and intravenous administration [24]. The composition of a polymeric nanosphere is made up of a closely-packed polymer matrix that aids the adsorption and binding of drugs. In comparison, polymeric nanocapsules take on a polymeric shell composition (although the capsule may also consist of lipids) [14]. Both NPs types are currently under investigation for their drug delivery abilities.

3.3. Inorganic NPs

Inorganic NPs embody a wide variety of substances, several of which can be utilised for neurological drug delivery. These include gold NPs (AuNPS), fluorescent nanodiamonds (FNDs), magnetic NPs (MNPs) ceramic NPs, and upconversion NPs (UCNPs). Gold NPs are nanoscopic in size with a sizable surface area to mass ratio. These features, combined with their proficient functionalisation, make them incredibly valuable carriers for drug delivery [25]. The same may be said for fluorescent nanodiamonds, which can enhance drug precision and retention through conjugation to various drugs and high levels of biocompatibility [26]. Magnetic NPs also denote significant benefits, as their magnetic properties enable them to be utilised as magnetic resonance imaging (MRI) contrast agents as well as carriers for nanotherapeutic drugs. This is due to their magnetic core that consists of two material forms: paramagnetic and super-paramagnetic properties. The latter material is beneficial in that does not manifest any magnetic properties beyond the external magnetic field, therefore it is highly functional for biomedical applications [27]. In contrast, ceramic NPs maintain a small, porous structure, which helps to aid water solubility. These NPs are often appointed for anticancer treatment due to their stability in biological environments and high molecular weight compounds [28].

Upconversion NPs constitute a selective inorganic substance and can serve as a neuroprotective unit for drug transportation. Their distinct features set them apart from other inorganic NPs, as they possess magnetic resonance imaging and upconversion luminescence (UCL) imaging features. The development of nanoprobes, synthesized to traverse the blood-brain barrier, has greatly assisted upconversion NPs in anticancer treatment. One application of this was a study whereby glioblastoma-affected mice were observed following intravenous nanotherapeutic treatment. These nanoprobes were successful in crossing the blood-brain barrier via receptor-mediated transcytosis [29]. The results obtained strongly indicated that the targeting efficacy of the nanoprobes surpassed that of single-mode imaging agents currently implemented in clinical practice [29]. Furthermore, the likelihood of incorporating upconversionnanoprobes for tumor radiotherapy and has been regarded as a strong possibility for future applications [30].

4. NEUROLOGICAL DISORDERS AND TREATMENT STRATEGIES

4.1. Parkinson’s Disease

Parkinson’s Disease is characterised by the progressive loss of dopaminergic neurons, which leads to slow movement (bradykinesia), tremors, and stiffness. Dopamine plays a crucial role in coordination and movement; therefore, a deficiency presents adverse neuronal effects. An additional marker of Parkinson’s is an increase in Lewy bodies, although little is known about their processes. Currently, there is no cure for Parkinson’s disease, but the initial stages may be treated with levodopa (L-DOPA), the precursor for dopamine. If the delivery of levodopa is untargeted however, the peripheral system may be compromised, generating dyskinesia and harmful cardiovascular effects [31]. For that reason, encapsulating neurotransmitters used for Parkinson’s treatment would guarantee an appropriate delivery system and allow for blood-brain barrier diffusion.

4.2. Alzheimer’s Disease

Alzheimer’s Disease is the most prevalent form of dementia (roughly 70% of cases) [32], signalised by memory loss and a difficulty in executing familiar tasks. Pathologically, the aggregation of insoluble amyloid-beta (Aβ) peptide deposits and neurofibrillary tangles incite neuroinflammation [33], which results in widespread brain atrophy. While no cure currently exists for Alzheimer’s, many therapeutic approaches and clinically approved drugs exist to mitigate its progression. Amyloid-beta and tau proteins within the cerebrospinal fluid (CSF) have been utilised diagnostically as they are considered to be significant biochemical markers [3]. The inhibition of amyloid-beta plaque and tau neurofibrillary tangle formation has been the focal point for recent remedial approaches. Transition metals exist as a foundation for amyloid-beta aggregation; therefore, it has been speculated that chelating agents could bind to these select metals [34]. Chelating agents are chemical compounds that help to lower blood and tissue levels of heavier metals (national institute of diabetes), and so could be seriously regarded for Alzheimer’s treatment [34]. Unfortunately, due to the blood-brain barrier’s selective permeability, the chelators’ potential is severely diminished. This is where NTs are advantageous, as they may be conjugated to iron chelators [34]. One NPs prototype, Nano-N2PY, was synthesized to impede Aβ aggregate formation, thus protecting the human brain from neurotoxicity.

4.3. Glioblastoma

Among the primary brain tumors, glioblastomas are regarded as the most common and most aggressive. It is accompanied by a grim prognosis, with only 25% of affected individuals possessing a 2-year survival rate following treatment [35]. Additionally, clinical treatment is inhibited by the formation of the blood-brain tumor barrier (BBTB). This structure, combined with blood-brain barrier, produces an additional hindrance for drug delivery to the glioblastoma cells, thus requiring newer drug development strategies so as to aid delivery to the tumor site. Extensive research has exhibited that the blood-brain tumor barrier may be breached through the utilization of non-toxic NPs that are no larger than 11.7-11.9 nm in diameter and possess prolonged blood half-lives [36]. Further studies have affirmed the correlation between glioblastoma targeting efficacy and nanotherapeutic treatment, such as conjugated liposomes binding to glioblastoma multiforme tissue which resulted in reduced tumor volume [37]. Additionally, magnetic resonance imaging (MRI) assessment showed that the dual-functionalized liposomes exerted significant tumor suppressive effects on glioblastoma cells, resulting in the reduced size of select tumor regions [20]. A selection of nanomaterials has displayed a range of favourable effects, including liposomes, polymeric micelles, and iron oxide NPs (IONP) 38. Overall, the nanomaterials have exhibited enhanced permeability and retention (EPR) through the implementation of positive targeting, which in turn evokes retention of the NPs within the tumors [38]. To supplement EPR, active targeting is administered in order to enhance drug delivery to tumor tissues.

4.4. Vascular Occlusion

Cardiovascular disease continues to claim one of the leading causes of death worldwide, despite being one of the most thoroughly researched and progressive. Various methods such as cellular therapy have been introduced to mitigate long-term effects, however, poor cell retention has posed as a severe hindrance [39]. A solution to this was the introduction of superparamagnetic iron oxide NPs, which demonstrated no adverse effects on cell viability and differentiation, nor functional capacity [39]. Further practical nanotherapeutic methods have included mechanically-activated biomimetic drug carriers, a synthetic implementation through which biochemical processes may be imitated. These drug carriers target sites for vascular occlusion, whereby a blockage in the blood vessel occurs. A vital component and determining factor for vascular pathophysiology is shear stress, a frictional force that is generated by blood flow. High levels of shear stress assist in promoting vasodilation, anticoagulation, and endothelial cell survival 40, whereas low levels correlate to high prothrombotic and inflammatory [41]. Our bodies are able to regulate shear stress levels ordinarily, however, they are compromised in the presence of arterial vascular diseases and hemodynamic conditions [41].

5. BIOLOGICAL BARRIERS FOR DRUG DELIVERY

Although NPs are able to distribute therapeutic agents in a safe and effective manner, they still come with their setbacks (Table 1). In spite of these barriers, however, great opportunities still remain for drug targeting by NTs. The vascular arrangement of tumors is incredibly heterogenous when it comes to dispersal, with certain areas being more permeable than others [44]. However, poor perfusion is a also a common occurrence in tumors. When lymphatic drainage in tumors is compromised, interstitial fluid pressure (IFP) is augmented [45], thus establishing another obstacle for drug delivery. Increased IFP is a leading source for the restriction of extravasation and transvascular macromolecule transport, while additionally impeding molecule transportation within the interstitial tumor space [46]. By increasing the cell density of the tumor, drug transportation may be affected [44]. For that reason, the targeted tissue area requires thorough investigation in order to ascertain permeability prior to administering nanotherapeutic drug delivery. Another challenge that NPs are faced with is their susceptibility to aggregate during formulation, storage, and application. This is due to their considerable surface area to volume ratio and may be resultant of external factors or simply their own inherent chemical properties. There are several proceedings through which this issue can be mitigated, however, including lyophilisation (or ‘freeze-drying’). This stabilises the NPs for a short period of time, after which they are able to assume their original structure for administration [47]. It does not stop there, unfortunately, as the NPs are still prone to aggregating, which interferes with their ability to redisperse when placed into an aqueous solution. For that reason, a non-reducing saccharide known as trehalose can be utilized, as it is able to undertake a cryoprotectant role 43. Trehalose has the ability to lower NP aggregation and increase their rate of separation, while placing no effect on their size, morphology, or yield [43]. Small NPs that were treated with trehalose went on to demonstrate more efficient brain tissue penetration than NPs who had not received cryoprotection [43].

Table 1A summary of the main challenges’ NPs faces in vivo during drug delivery, accompanied by the relevant surface property modifications that can be applied to overcome these obstacles.ChallengeModificationReferencesDifficulty in delivering therapeutic agents into solid tumors due to in adequate perfusion.Target tumor areas with more permeability.[42]NPs are susceptible to aggregation throughout the course of design, storage, and application.Modification viastabilising coatings.[43]Impaired lymphatic drainage in tumors, resulting in interstitial fluid pressure (IFP).Refrain from transvascular transportation of macromolecules.[42]

Further studies have evidenced that administering trehalose at nanoscale levels contributed to micromolar concentration efficacy and successful brain targeting [48]. Conclusively, it could be applied to preventative and therapeutic methodologies for a number of neurodegenerative disorders.

CONCLUDING REMARKS

The blood-brain barrier has long been attested as a significant hindrance to successful drug delivery to the central nervous system. The research and development of engineered and multifunctional NPs as pharmaceutical drug carriers has spurred exponential growth in applications to medicine throughout the last decade, proving them to be highly relevant, and indeed advantageous, for the treatment of neurological diseases. This is evident for an abundance of reasons. Firstly, their substances are able to permeate the blood-brain barrier, a common deterrent for central nervous system-targeted therapies. NPs also possess the ability to be engineered to interact with defined cellular subgroups and/or molecules, thereby affording specificity of treatment. Additionally, the materials are multifaceted in that numerous features may be applied to the NPs so as to ensure concurrent targeting, bioactivity, and successful delivery.

Many existing drug treatments have proven to be ineffective at healing, or even mitigating the progression of much neurological pathology. Again, this is accredited in large part to the role of the blood-brain barrier. Moreover, many central nervous system disorders’ disease mechanisms remain unmarked. Looking forward, however, NTs has every indication of advancing positively, particularly with the development of successful strategies to permeate the blood-brain barrier. As we continue to broaden our knowledge and comprehension on neuropathology and etiology, we will be able to continue applying it to NPs engineering and the vital role it holds in deriving original therapeutic treatments and approaches. Overall, NTs hold a great promise to deliver treatment for neurological diseases due to their beneficial features and imparts substantial opportunities for targeting by NPs.

CONSENT FOR PUBLICATION

Not applicable.

CONFLICT OF INTEREST

The author declares no conflict of interest, financial or otherwise.

ACKNOWLEDGEMENTS

Declared none.

REFERENCES

[1]Neurology Collaborators GBD. Global, regional, and national burden of neurological disorders, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 2019; 18(5): 459-80.[http://dx.doi.org/10.1016/S1474-4422(18)30499-X] [PMID: 30879893][2]Micheli MR, Bova R, Magini A, Polidoro M, Emiliani C. Lipid-based nanocarriers for CNS-targeted drug delivery. Recent Patents CNS Drug Discov 2012; 7(1): 71-86.[http://dx.doi.org/10.2174/157488912798842241] [PMID: 22283231][3]Kang YJ, Cutler EG, Cho H. Therapeutic nanoplatforms and delivery strategies for neurological disorders. Nano Converg 2018; 5(1): 35.[http://dx.doi.org/10.1186/s40580-018-0168-8] [PMID: 30499047][4]Kim J, Ahn S. Nanotherapeutics Engineered to Cross the Blood-Brain Barrier for Advanced Drug Delivery to the Nervous System. J Ind Eng Chem 2019; 73: 8-18.[5]Edelblum K. L.; Turner, J., R. Epithelial Cells. 4th ed.2015.[6]Laksitorini M, Prasasty VD, Kiptoo PK, Siahaan TJ. Pathways and progress in improving drug delivery through the intestinal mucosa and blood-brain barriers. Ther Deliv 2014; 5(10): 1143-63.[http://dx.doi.org/10.4155/tde.14.67] [PMID: 25418271][7]Pardridge WM, Blood-Brain Barrier Drug Targeting M. Blood-brain barrier drug targeting: the future of brain drug development. Mol Interv 2003; 3(2): 90-105, 51.[http://dx.doi.org/10.1124/mi.3.2.90] [PMID: 14993430][8]Kreuter J. Nanoparticulate systems for brain delivery of drugs. Adv Drug Deliv Rev 2012; 64(1): 213-22.[http://dx.doi.org/10.1016/j.addr.2012.09.015] [PMID: 11251246][9]Kou L, Bhutia YD, Yao Q, He Z, Sun J, Ganapathy V. Transporter-guided delivery of nanoparticles to improve drug permeation across cellular barriers and drug exposure to selective cell types. Front Pharmacol 2018; 9: 27.[http://dx.doi.org/10.3389/fphar.2018.00027] [PMID: 29434548][10]Villaseñor R, Lampe J, Schwaninger M, Collin L. Intracellular transport and regulation of transcytosis across the blood-brain barrier. Cell Mol Life Sci 2019; 76(6): 1081-92.[http://dx.doi.org/10.1007/s00018-018-2982-x] [PMID: 30523362][11]De Jong WH, Borm PJA. Drug delivery and nanoparticles:applications and hazards. Int J Nanomedicine 2008; 3(2): 133-49.[http://dx.doi.org/10.2147/IJN.S596] [PMID: 18686775][12]Puri A, Loomis K, Smith B, et al. Lipid-based nanoparticles as pharmaceutical drug carriers: from concepts to clinic. Crit Rev Ther Drug Carrier Syst 2009; 26(6): 523-80.[http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.v26.i6.10] [PMID: 20402623][13]Mishra B, Patel B. Nanomedicine (Lond) 2010; 6(1): 9-24.[http://dx.doi.org/10.1016/j.nano.2009.04.008][14]Feng L, Mumper RJ. A critical review of lipid-based nanoparticles for taxane delivery. Cancer Lett 2013; 334(2): 157-75.[http://dx.doi.org/10.1016/j.canlet.2012.07.006] [PMID: 22796606][15]Jose S, Anju SS, Cinu TA, Aleykutty NA, Thomas S, Souto EB. In vivo pharmacokinetics and biodistribution of resveratrol-loaded solid lipid nanoparticles for brain delivery. Int J Pharm 2014; 474(1-2): 6-13.[http://dx.doi.org/10.1016/j.ijpharm.2014.08.003] [PMID: 25102112][16]Webb MS, Harasym TO, Masin D, Bally MB, Mayer LD. Sphingomyelin-cholesterol liposomes significantly enhance the pharmacokinetic and therapeutic properties of vincristine in murine and human tumour models. Br J Cancer 1995; 72(4): 896-904.[http://dx.doi.org/10.1038/bjc.1995.430] [PMID: 7547237][17]Hersh D. S.; Wadajkar, A., S.; Roberts, N.; Perez, J., G.; Connolly, N., P.; Frenkel, V.; Winkles, J., A.; Woodworth, G., F.; Kim, A., J. Evolving Drug Delivery Strategies to Overcome the Blood Brain Barrier 2016; 22(9): 1177-93.[18]Owens DE, III, Peppas NA. Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int J Pharm 2006; 307(1): 93-102.[http://dx.doi.org/10.1016/j.ijpharm.2005.10.010] [PMID: 16303268][19]Liu L, Guo K, Lu J, et al. Biologically active core/shell nanoparticles self-assembled from cholesterol-terminated PEG-TAT for drug delivery across the blood-brain barrier. Biomaterials 2008; 29(10): 1509-17.[http://dx.doi.org/10.1016/j.biomaterials.2007.11.014] [PMID: 18155137][20]Gao JQ, Lv Q, Li LM, et al. Glioma targeting and blood-brain barrier penetration by dual-targeting doxorubincin liposomes. Biomaterials 2013; 34(22): 5628-39.[http://dx.doi.org/10.1016/j.biomaterials.2013.03.097] [PMID: 23628475][21]Demeule M, Currie JC, Bertrand Y, et al. Involvement of the low-density lipoprotein receptor-related protein in the transcytosis of the brain delivery vector angiopep-2. J Neurochem 2008; 106(4): 1534-44.[http://dx.doi.org/10.1111/j.1471-4159.2008.05492.x] [PMID: 18489712][22]Albertazzi L, Gherardini L, Brondi M, et al.In vivo distribution and toxicity of PAMAM dendrimers in the central nervous system depend on their surface chemistry. Mol Pharm 2013; 10(1): 249-60.[http://dx.doi.org/10.1021/mp300391v] [PMID: 23163881][23]Jain KK. The Handbook of Nanomedicine. 2012.[http://dx.doi.org/10.1007/978-1-61779-983-9][24]Ke W, Shao K, Huang R, et al. Gene delivery targeted to the brain using an Angiopep-conjugated polyethyleneglycol-modified polyamidoamine dendrimer. Biomaterials 2009; 30(36): 6976-85.[http://dx.doi.org/10.1016/j.biomaterials.2009.08.049] [PMID: 19765819][25]Parveen S, Misra R, Sahoo SK, Nanoparticles K. Nanoparticles: a boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine 2012; 8(2): 147-66.[http://dx.doi.org/10.1016/j.nano.2011.05.016] [PMID: 21703993][26]Shumoni O, Shi B, Adlard P, Bush AI. Delivery of fluorescent nanoparticles to the brain. J Mol Neurosci 2016; 60(3): 405-9.[http://dx.doi.org/10.1007/s12031-016-0833-5] [PMID: 27660216][27]Sun C, Lee JS, Zhang M. Magnetic nanoparticles in MR imaging and drug delivery. Adv Drug Deliv Rev 2008; 60(11): 1252-65.[http://dx.doi.org/10.1016/j.addr.2008.03.018] [PMID: 18558452][28]Yih TC, Al-Fandi M. Engineered nanoparticles as precise drug delivery systems. J Cell Biochem 2006; 97(6): 1184-90.[http://dx.doi.org/10.1002/jcb.20796] [PMID: 16440317][29]Ni D, Zhang J, Bu W, et al. Dual-targeting upconversion nanoprobes across the blood-brain barrier for magnetic resonance/fluorescence imaging of intracranial glioblastoma. ACS Nano 2014; 8(2): 1231-42.[http://dx.doi.org/10.1021/nn406197c] [PMID: 24397730][30]Xing H, Zheng X, Ren Q, et al. Computed tomography imaging-guided radiotherapy by targeting upconversion nanocubes with significant imaging and radiosensitization enhancements. 1751; Vol. 3[31]Thanvi B, Lo N, Robinson T. Levodopa-induced dyskinesia in Parkinson’s disease: clinical features, pathogenesis, prevention and treatment. Postgrad Med J 2007; 83(980): 384-8.[http://dx.doi.org/10.1136/pgmj.2006.054759] [PMID: 17551069][32]Ott A, Breteler MM, van Harskamp F, et al. Prevalence of Alzheimer’s disease and vascular dementia: association with education. The Rotterdam study. BMJ 1995; 310(6985): 970-3.[http://dx.doi.org/10.1136/bmj.310.6985.970] [PMID: 7728032][33]Akiyama H, Mori H, Saido T, Kondo H, Ikeda K, McGeer PL. Occurrence of the diffuse amyloid beta-protein (Abeta) deposits with numerous Abeta-containing glial cells in the cerebral cortex of patients with Alzheimer’s disease. Glia 1999; 25(4): 324-31.[http://dx.doi.org/10.1002/(SICI)1098-1136(19990215)25:4<324::AID-GLIA2>3.0.CO;2-5] [PMID: 10028915][34]Liu G, Men P, Kudo W, Perry G, Smith MA. Nanoparticle-chelator conjugates as inhibitors of amyloid-beta aggregation and neurotoxicity: a novel therapeutic approach for Alzheimer disease. Neurosci Lett 2009; 455(3): 187-90.[http://dx.doi.org/10.1016/j.neulet.2009.03.064] [PMID: 19429118][35]van Tellingen O, Yetkin-Arik B, de Gooijer MC, Wesseling P, Wurdinger T, de Vries HE. Overcoming the blood-brain tumor barrier for effective glioblastoma treatment. Drug Resist Updat 2015; 19: 1-12.[http://dx.doi.org/10.1016/j.drup.2015.02.002] [PMID: 25791797][36]Sarin H, Kanevsky AS, Wu H, et al. Effective transvascular delivery of nanoparticles across the blood-brain tumor barrier into malignant glioma cells. J Transl Med 2008; 6(80): 80.[http://dx.doi.org/10.1186/1479-5876-6-80] [PMID: 19094226][37]Madhankumar AB, Slagle-Webb B, Mintz A, Sheehan JM, Connor JR. Interleukin-13 receptor-targeted nanovesicles are a potential therapy for glioblastoma multiforme. Mol Cancer Ther 2006; 5(12): 3162-9.[http://dx.doi.org/10.1158/1535-7163.MCT-06-0480] [PMID: 17172420][38]Michael JS, Lee BS, Zhang M, Yu JS. Nanotechnology for Treatment of Glioblastoma Multiforme. J Transl Int Med 2018; 6(3): 128-33.[http://dx.doi.org/10.2478/jtim-2018-0025] [PMID: 30425948][39]Riegler J, Liew A, Hynes SO, et al. Superparamagnetic iron oxide nanoparticle targeting of MSCs in vascular injury. Biomaterials 2013; 34(8): 1987-94.[http://dx.doi.org/10.1016/j.biomaterials.2012.11.040] [PMID: 23237516][40]Paszkowiak JJ, Dardik A. Arterial wall shear stress: observations from the bench to the bedside. Vasc Endovascular Surg 2003; 37(1): 47-57.[http://dx.doi.org/10.1177/153857440303700107] [PMID: 12577139][41]Lu D, Kassab GS. Role of shear stress and stretch in vascular mechanobiology. J R Soc Interface 2011; 8(63): 1379-85.[http://dx.doi.org/10.1098/rsif.2011.0177] [PMID: 21733876][42]Dessai N. Challenges in development of nanoparticle-based therapeutics. AAPS J 2012; 14(2): 282-95.[http://dx.doi.org/10.1208/s12248-012-9339-4] [PMID: 22407288][43]Zhou J, Patel TR, Sirianni RW, et al. Highly penetrative, drug-loaded nanocarriers improve treatment of glioblastoma. Proceedings of the National Academy of Sciences of the United States of America Vol. 110: 11751-6.[http://dx.doi.org/10.1073/pnas.1304504110][44]Jang S, H, Wientjes M, G, Lu D, Au J, L, S. Drug delivery and transport to solid tumors Pharmnaceutical research 2003; 20(9): 1337-50.[45]Maeda H, Sawa T, Konno T. Mechanism of tumor-targeted delivery of macromolecular drugs, including the EPR effect in solid tumor and clinical overview of the prototype polymeric drug SMANCS Journal of controlled release: official journal of the Controlled Release Society 2001; 74(1-3): 47-61.[http://dx.doi.org/10.1016/S0168-3659(01)00309-1][46]Jain R, K. Transport of molecules across tumor vasculature. Cancer Metastasis Rev 1987; 6(4): 559-93.[47]Rey, L; May, J (3rd ed.), 3rd ed.2010.[48]Debnath K, Pradhan N, Singh BK, Jana NR, Jana NR. Poly(trehalose) nanoparticles prevent amyloid aggregation and suppress polyglutamine aggregation in a huntington’s disease model mouse. ACS Appl Mater Interfaces 2017; 9(28): 24126-39.[http://dx.doi.org/10.1021/acsami.7b06510] [PMID: 28632387]

Molecular Mechanism of Therapeutic Actions of Some Nanoparticles in Some Diseases

Bello Aminu Bello1,*,Ibrahim Khalil Adam1,Sani S. Usman2,Yahaya Saidu Gwarzo1,Luqman Shah3,Fatima Sulaiman Abdullahi4
1 Department of Biochemistry, Federal University Dutse, P.M.B.7156 Dutse, Jigawa State, Nigeria
2 Department of Biological Sciences, Federal University Kashere, P.M.B. 0182 Kashere, Gombe, Nigeria
3 Department of Biochemistry, Hazara University Mansehra, Mansehra, Khyber Pakhtunkhwa, Pakistan
4 Department of Food Science and Technology, Federal University Dutsin-MA, P.M.B. 5001, Katsina State, Nigeria

Abstract

This chapter covers a detailed description of various molecular mechanisms of therapeutic actions of silver nanoparticles (AgNPs), gold nanoparticles (AuNPs), Iron Oxide nanoparticles (FeO-NPs), Titanium Dioxide nanoparticles (TiO2-NPs), Cerium Oxide Nanoparticles (CNPs), Zinc Oxide nanoparticles (ZnO-NPs), Nitric oxide releasing nanoparticles (NO-NPs) among others in cancer, diabetes, bacterial, fungal, viral and inflammatory diseases. The mechanisms of anticancer activity of the nanoparticles (NPs) range from ultra-structure disruption, generation of reactive oxygen species (ROS), induction of DNA damage, inactivation of proteins that regulate signalling pathways, inhibition of migration and angiogenesis as well as induction of apoptosis. The mechanism of anti-diabetic activity of the NPs is through inhibition of α-amylase and protein tyrosine phosphatase 1B. The antibacterial and anti-fungal activities of the NPs are by disruption of membrane and induced DNA damage as a result of generation of ROS and dissolved metal ions. The diseases associated with viral infections are treated by restricting the entrance of the virus into the host and by binding to the ap120 site on the viral membrane, thereby regulating its function. The therapeutic mechanism of the NPs in inflammatory diseases is through blocking the production of pro-inflammatory cytokines, inhibiting mast cell proliferation, suppressing lipopolysaccharides (LPS) induced cyclooxygenase (COX-2) gene expression, reducing vascular endothelial growth factor level, decreasing Hypoxia-Inducible Factor (HIF) 1 α -gene expression, suppressing the inducible nitric oxide synthases (INO) gene expression as well as preventing mucin hypersecretion. Therefore, it is clear that the NPs possess various effective and efficient mechanisms of action against both infectious and degenerative diseases.

Keywords: Cancer, Diabetes, Diseases, Inflammation, Molecular Mechanism, Nanoparticles, Therapeutic Action.
*Corresponding author Bello Aminu Bello: Department of Biochemistry, Federal University Dutse, P.M.B. 7156 Dutse, Jigawa State, Nigeria; Email: [email protected]

1. INTRODUCTION

The field nano-biotechnology is considered one of the most dynamic and rapidly growing research fields with diverse applications. It deals with synthesis, strategy and manipulation of new materials at a scale between 1nm and 100 nm [1]. Therefore, the major product of nano-biotechnology called nanoparticles which can be synthesized through different methods that range from physical, chemical, electrochemical, photochemical, biological as well as the use of irradiative techniques [2]. In physical and chemical methods, the involvement of high radiation and high concentrations of both the reductants and stabilizing agents are extremely harmful to the environment, humans and other living organisms, thereby limiting their vast applications [3]. Even though these methods were recorded for being successful in generating pure and well defined NPs, they are highly expensive and release dangerous products to the environment [4]. The biological method, on the other hand, involves a single step bio-reduction process which is cost-effective, can be operated easily at an industrial scale, requires less energy and eco-friendly products (NPs) that are safe for humans with therapeutic importance are generated [5, 6]. Many bio-based substances such as plant extracts, fungi, bacteria, algae and enzymes act as reducing and protecting agents in the green synthesis of NPs [7, 8]. The plant based NPs synthesis is one of the emerging fields of nanotechnology in the recent era [8] that has been continuously drawing the attention of more researchers due to their numerous applications in different fields as a result of their inherent properties and pose no harmful effects to the environment. It is usually carried out at a neutral pH and ambient temperatures [4, 9]. These plant materials or extracts served as both reducing and capping agents for metallic ions have more advantages than other biological material [10]. Because of their safer applications, plant-based metallic NPs are the most demanding and most effective in the field nano-biotechnology. The different metallic NPs are synthesized using metallic ions such as silver, gold, zinc, copper, titanium, magnetite and nickel (Fig. 1) and different parts of the plants or extracts such as stem, roots, fruits, leaves and flowers are largely investigated with various biological potentials [4].

Due to their completely new and enhanced properties such as their high surface-to-volume ratio and small size, distribution and morphology which allow for the ability to surpass barriers and gain access to biological molecules and, particularly in microorganisms [11], NPs are continuously gaining applications in many fields such as health care, implants, prosthetics, in-vitro diagnostics, cosmetics, biomedical, food and feeds, drug-gene delivery, environmental study, mechanics, optics, optoelectronic devices, bio-imaging devices, sensors and many others [12]. The metallic NPs have been widely used in these fields because of their high stability, solubility, multi-functionality, bio compatibility, adhesive as well as therapeutic properties. The current trend is the development of nanoparticles which have better therapeutic properties as well as being environment-friendly [13-15]. Among these metallic NPs, the NPs from noble metals such as gold (Au), silver [1] and platinum (Pt) are well studied and reported to have significant applications in electronics, magnetic, optoelectronics and information storage. Recently, it was reported that, NPs could serve as drug carriers either by active or passive mechanism [1]. The most studied among the metallic NPs are the AgNPs and were reported to be used in producing products that have direct contact with human systems such as shampoos, soaps, detergents, cosmetics, toothpaste, medical, pharmaceuticals, among others [16]. The nanoparticles (NPs) are reported to exhibit a wider range of superior physical, chemical, mechanical, thermal as well as biological properties when compared with the bulk or starting materials. All of these properties are important to the biological and research fields because they influence their antibacterial [17], antifungal [4], anti-inflammatory [18, 19], antiviral [20], anti-diabetic [21], antioxidant [22] and anti-angiogenic potential with improved catalytic activities [15, 23, 24]. AgNPs are reported to have potential applications in cancer diagnosis and treatment [25-27]. They are reported to be used in the treatment of ulcer [28-30] and cardiovascular disorders [31].

Fig. (1)) The metallic-nanoparticles synthesized from different plant extracts.