Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5273
ISSN (Online): 1996-3181

Review Article

Nanoparticles Based Intranasal Delivery of Drug to Treat Alzheimer’s Disease: A Recent Update

Author(s): Manisha Pandey*, Hira Choudhury*, Rohit Kumar Verma, Viney Chawla, Subrat Kumar Bhattamisra, Bapi Gorain, Maria Abdul Ghafoor Raja and Muhammad Wahab Amjad

Volume 19, Issue 9, 2020

Page: [648 - 662] Pages: 15

DOI: 10.2174/1871527319999200819095620

Price: $65

Abstract

Alzheimer Association Report (2019) stated that the 6th primary cause of death in the USA is Alzheimer’s Disease (AD), which leads to behaviour and cognitive impairment. Nearly 5.8 million peoples of all ages in the USA have suffered from this disease, including 5.6 million elderly populations. The statistics of the progression of this disease is similar to the global scenario. Still, the treatment of AD is limited to a few conventional oral drugs, which often fail to deliver an adequate amount of the drug in the brain. The reduction in the therapeutic efficacy of an anti-AD drug is due to poor solubility, existence to the blood-brain barrier and low permeability. In this context, nasal drug delivery emerges as a promising route for the delivery of large and small molecular drugs for the treatment of AD. This promising pathway delivers the drug directly into the brain via an olfactory route, which leads to the low systemic side effect, enhanced bioavailability, and higher therapeutic efficacy. However, few setbacks, such as mucociliary clearance and poor drug mucosal permeation, limit its translation from the laboratory to the clinic. The above stated limitation could be overcome by the adaption of nanoparticle as a drug delivery carrier, which may lead to prolong delivery of drugs with better permeability and high efficacy. This review highlights the latest work on the development of promising Nanoparticles (NPs) via the intranasal route for the treatment of AD. Additionally, the current update in this article will draw the attention of the researcher working on these fields and facing challenges in practical applicability.

Keywords: Alzheimer’s disease, nanoparticles, intranasal, permeability, blood-brain barrier, cognitive impairment.

Graphical Abstract
[1]
Gorain B, Choudhury H, Pandey M, et al. Mechanistic description of natural herbs in the treatment of dementia: a systematic review. Curr Psychopharmacol 2018; 7: 149-64.
[http://dx.doi.org/10.2174/2211556007666180420124544]
[2]
Livingston G, Sommerlad A, Orgeta V, et al. Dementia prevention intervention and care. Lancet 2017; 390(10113): 2673-734.
[http://dx.doi.org/10.1016/S0140-6736(17)31363-6 PMID: 28735855]
[3]
Naj AC, Schellenberg GD. Alzheimer’s Disease Genetics Consortium (ADGC). Am J Med Genet B Neuropsychiatr Genet 2017; 174(1): 5-26.
[4]
Apostolova LG, Risacher SL, Duran T, et al. Alzheimer’s disease neuroimaging initiative. JAMA Neurol 2018; 75: 328-41.
[http://dx.doi.org/10.1001/jamaneurol.2017.4198] [PMID: 29340569]
[5]
Safieh M, Korczyn AD, Michaelson DM. ApoE4: an emerging therapeutic target for Alzheimer’s disease. BMC Med 2019; 17(1): 64.
[http://dx.doi.org/10.1186/s12916-019-1299-4] [PMID: 30890171]
[6]
Irwin MR, Vitiello MV. Implications of sleep disturbance and inflammation for Alzheimer’s disease dementia. Lancet Neurol 2019; 18(3): 296-306.
[http://dx.doi.org/10.1016/S1474-4422(18)30450-2 PMID: 30661858]
[7]
Tezel G, Timur SS, Bozkurt İ, et al. A Snapshot on the current status of Alzheimer’s disease treatment perspectives in-vitro and in-vivo research studies and future opportunities. Chem Pharm Bull 2019; 67: 1030-41.
[8]
Ringman JM, Goate A, Masters CL, et al. Dominantly inherited Alzheimer network. Genetic heterogeneity in Alzheimer disease and implications for treatment strategies. Curr Neurol Neurosci Rep 2014; 14(11): 499.
[http://dx.doi.org/10.1007/s11910-014-0499-8] [PMID: 25217249]
[9]
Iqbal K, Liu F, Gong CX. Alzheimer disease therapeutics: focus on the disease and not just plaques and tangles. Biochem Pharmacol 2014; 88(4): 631-9.
[http://dx.doi.org/10.1016/j.bcp.2014.01.002] [PMID: 24418409]
[10]
Mizuno S, Iijima R, Ogishima S, et al. AlzPathway: a comprehensive map of signaling pathways of Alzheimer’s disease. BMC Syst Biol 2012; 6: 52.
[http://dx.doi.org/10.1186/1752-0509-6-52] [PMID: 22647208]
[11]
Cummings JL, Tong G, Ballard C. Treatment combinations for Alzheimer’s disease: current and future pharmacotherapy options. J Alzheimers Dis 2019; 67(3): 779-94.
[http://dx.doi.org/10.3233/JAD-180766] [PMID: 30689575]
[12]
Frölich L, Atri A, Ballard C, et al. Open-label multicenter phase III extension study of idalopirdine as adjunctive to donepezil for the treatment of mild-moderate Alzheimer’s disease. J Alzheimers Dis 2019; 67(1): 303-13.
[http://dx.doi.org/10.3233/JAD-180595] [PMID: 30636738]
[13]
Adlimoghaddam A, Neuendorff M, Roy B, Albensi BC. A review of clinical treatment considerations of donepezil in severe Alzheimer’s disease. CNS Neurosci Ther 2018; 24(10): 876-88.
[http://dx.doi.org/10.1111/cns.13035] [PMID: 30058285]
[14]
Chatterjee B, Gorain B, Mohananaidu K, Sengupta P, Mandal UK, Choudhury H. Targeted drug delivery to the brain via intranasal nanoemulsion: available proof of concept and existing challenges. Int J Pharm 2019; 565: 258-68.
[http://dx.doi.org/10.1016/j.ijpharm.2019.05.032] [PMID: 31095983]
[15]
Gadhave D, Choudhury H, Kokare C. Neutropenia and leukopenia protective intranasal olanzapine-loaded lipid-based nanocarriers engineered for brain delivery. Appl Nanosci 2018; 9: 152-68.
[16]
Alam MI, Beg S, Samad A, et al. Strategy for effective brain drug delivery. Eur J Pharm Sci 2010; 40(5): 385-403.
[http://dx.doi.org/10.1016/j.ejps.2010.05.003] [PMID: 20497904]
[17]
Pardeshi CV, Belgamwar VS. Direct nose to brain drug delivery via integrated nerve pathways bypassing the blood-brain barrier: an excellent platform for brain targeting. Expert Opin Drug Deliv 2013; 10(7): 957-72.
[http://dx.doi.org/10.1517/17425247.2013.790887] [PMID: 23586809]
[18]
Gadhave D, Gorain B, Tagalpallewar A, et al. Intranasal teriflunomide microemulsion: an improved chemotherapeutic approach in glioblastoma. Drug Deliv Sci Technol 2019; 51: 276-89.
[http://dx.doi.org/10.1016/j.jddst.2019.02.013]
[19]
Choudhury H, Gorain B, Karmakar S, et al. Improvement of cellular uptake in vitro antitumor activity and sustained release profile with increased bioavailability from a nanoemulsion platform. Int J Pharm 2014; 460(1-2): 131-43.
[http://dx.doi.org/10.1016/j.ijpharm.2013.10.055] [PMID: 24239580]
[20]
Choudhury H, Pandey M, Chin PX, et al. Transferrin receptors-targeting nanocarriers for efficient targeted delivery and transcytosis of drugs into the brain tumors: a review of recent advancements and emerging trends. Drug Deliv Transl Res 2018; 8(5): 1545-63.
[http://dx.doi.org/10.1007/s13346-018-0552-2] [PMID: 29916012]
[21]
Choudhury H, Gorain B, Pandey M, et al. Recent advances in TPGS-based nanoparticles of docetaxel for improved chemotherapy. Int J Pharm 2017; 529(1-2): 506-22.
[http://dx.doi.org/10.1016/j.ijpharm.2017.07.018 PMID: 28711640]
[22]
Salatin S, Maleki Dizaj S, Yari Khosroushahi A. Effect of the surface modification size and shape on cellular uptake of nanoparticles. Cell Biol Int 2015; 39(8): 881-90.
[http://dx.doi.org/10.1002/cbin.10459] [PMID: 25790433]
[23]
Gorain B, Choudhury H, Pandey M, Kesharwani P. Paclitaxel loaded vitamin E-TPGS nanoparticles for cancer therapy. Mater Sci Eng C 2018; 91: 868-80.
[http://dx.doi.org/10.1016/j.msec.2018.05.054] [PMID: 30033322]
[24]
Kumar Thakur A, Kamboj P, Goswami K, et al. Pathophysiology and management of Alzheimer’s disease: an overview. Anal Pharm Res 2018; 7: 226-35.
[http://dx.doi.org/10.15406/japlr.2018.07.00230]
[25]
Dos Santos Picanco LC, Ozela PF, de Fatima de Brito Brito M, et al. Alzheimer’s disease: a review from the pathophysiology to diagnosis new perspectives for pharmacological treatment. Curr Med Chem 2018; 25(26): 3141-59.
[http://dx.doi.org/10.2174/0929867323666161213101126 PMID: 30191777]
[26]
Brandt R, Bakota L. Microtubule dynamics and the neurodegenerative triad of Alzheimer’s disease: the hidden connection. J Neurochem 2017; 143(4): 409-17.
[http://dx.doi.org/10.1111/jnc.14011] [PMID: 28267200]
[27]
Barbier P, Zejneli O, Martinho M, et al. Role of tau as a microtubule-associated protein: structural and functional aspects. Front Aging Neurosci 2019; 11: 204.
[http://dx.doi.org/10.3389/fnagi.2019.00204] [PMID: 31447664]
[28]
Esiri MM, Pearson RC, Steele JE, Bowen DM, Powell TP. A quantitative study of the neurofibrillary tangles and the choline acetyltransferase activity in the cerebral cortex and the amygdala in Alzheimer’s disease. J Neurol Neurosurg Psychiatry 1990; 53(2): 161-5.
[http://dx.doi.org/10.1136/jnnp.53.2.161] [PMID: 2313304]
[29]
Chow VW, Mattson MP, Wong PC, Gleichmann M. An overview of APP processing enzymes and products. Neuromolecular Med 2010; 12(1): 1-12.
[http://dx.doi.org/10.1007/s12017-009-8104-z] [PMID: 20232515]
[30]
Aisenbrey C, Borowik T, Byström R, et al. How is protein aggregation in amyloidogenic diseases modulated by biological membranes? Eur Biophys J 2008; 37(3): 247-55.
[http://dx.doi.org/10.1007/s00249-007-0237-0] [PMID: 18030461]
[31]
Bartus R, Dean R, Beer B, et al. The cholinergic hypothesis of geriatric memory dysfunction Science (80- ) 1982; 217: 408-14.
[http://dx.doi.org/10.1126/science.7046051]
[32]
Reinikainen KJ, Riekkinen PJ, Paljärvi L, et al. Cholinergic deficit in Alzheimer’s disease: a study based on CSF and autopsy data. Neurochem Res 1988; 13(2): 135-46.
[http://dx.doi.org/10.1007/BF00973325] [PMID: 3362291]
[33]
Reinikainen KJ, Soininen H, Riekkinen PJ. Neurotransmitter changes in Alzheimer’s disease: implications to diagnostics and therapy. J Neurosci Res 1990; 27(4): 576-86.
[http://dx.doi.org/10.1002/jnr.490270419] [PMID: 1981917]
[34]
Nagai T, McGeer PL, Peng JH, McGeer EG, Dolman CE. Choline acetyltransferase immunohistochemistry in brains of Alzheimer’s disease patients and controls. Neurosci Lett 1983; 36(2): 195-9.
[http://dx.doi.org/10.1016/0304-3940(83)90264-1] [PMID: 6866327]
[35]
Ballinger EC, Ananth M, Talmage DA, Role LW. Basal forebrain cholinergic circuits and signaling in cognition and cognitive decline. Neuron 2016; 91(6): 1199-218.
[http://dx.doi.org/10.1016/j.neuron.2016.09.006] [PMID: 27657448]
[36]
Ferris SH, Farlow M. Language impairment in Alzheimer’s disease and benefits of acetylcholinesterase inhibitors. Clin Interv Aging 2013; 8: 1007-14.
[http://dx.doi.org/10.2147/CIA.S39959] [PMID: 23946647]
[37]
Caccamo A, Fisher A, LaFerla FM. M1 agonists as a potential disease-modifying therapy for Alzheimer’s disease. Curr Alzheimer Res 2009; 6(2): 112-7.
[http://dx.doi.org/10.2174/156720509787602915] [PMID: 19355845]
[38]
Habtemariam S. Natural products in Alzheimer’s disease therapy: would old therapeutic approaches fix the broken promise of modern medicines? Molecules 2019; 24(8): 1519.
[http://dx.doi.org/10.3390/molecules24081519] [PMID: 30999702]
[39]
Lau A, Tymianski M. Glutamate receptors neurotoxicity and neurodegeneration. Pflugers Arch 2010; 460(2): 525-42.
[http://dx.doi.org/10.1007/s00424-010-0809-1] [PMID: 20229265]
[40]
Ferreira-Vieira TH, Guimaraes IM, Silva FR, Ribeiro FM. Alzheimer’s disease: targeting the cholinergic system. Curr Neuropharmacol 2016; 14(1): 101-15.
[http://dx.doi.org/10.2174/1570159X13666150716165726 PMID: 26813123]
[41]
Liu J, Chang L, Song Y, Li H, Wu Y. The role of NMDA receptors in Alzheimer’s disease. Front Neurosci 2019; 13: 43.
[http://dx.doi.org/10.3389/fnins.2019.00043] [PMID: 30800052]
[42]
Tönnies E, Trushina E. Oxidative stress synaptic dysfunctionand Alzheimer’s disease. J Alzheimers Dis 2017; 57(4): 1105-21.
[http://dx.doi.org/10.3233/JAD-161088] [PMID: 28059794]
[43]
Liu Z, Zhang A, Sun H, et al. Two decades of new drug discovery and development for Alzheimer’s disease. RSC Adv 2017; 7: 6046-58.
[http://dx.doi.org/10.1039/C6RA26737H]
[44]
Daulatzai MA. Fundamental role of pan-inflammation and oxidative-nitrosative pathways in neuropathogenesis of Alzheimer’s disease. Am J Neurodegener Dis 2016; 5(1): 1-28.
[PMID: 27073740]
[45]
Wen MM, El-Salamouni NS, El-Refaie WM, et al. Nanotechnology-based drug delivery systems for Alzheimer’s disease management: technical, industrial and clinical challenges. J Control Release 2017; 245: 95-107.
[http://dx.doi.org/10.1016/j.jconrel.2016.11.025] [PMID: 27889394]
[46]
Bartzokis G, Sultzer D, Cummings J, et al. In vivo evaluation of brain iron in Alzheimer disease using magnetic resonance imaging. Arch Gen Psychiatry 2000; 57(1): 47-53.
[http://dx.doi.org/10.1001/archpsyc.57.1.47] [PMID: 10632232]
[47]
Deraeve C, Pitie M, Meunier B. Influence of chelators and iron ions on the production and degradation of H2O2 by beta-amyloid-copper complexes. J Inorg Biochem 2006; 100(12): 2117-26.
[http://dx.doi.org/10.1016/j.jinorgbio.2006.08.005] [PMID: 17011628]
[48]
De Felice FG, Ferreira ST. Inflammation defective insulin signaling and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to Alzheimer disease. Diabetes 2014; 63(7): 2262-72.
[http://dx.doi.org/10.2337/db13-1954] [PMID: 24931033]
[49]
Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (N Y) 2018; 4: 575-90.
[http://dx.doi.org/10.1016/j.trci.2018.06.014] [PMID: 30406177]
[50]
Femminella GD, Thayanandan T, Calsolaro V, et al. Genetic variations underlying Alzheimer’s disease: evidence from genome-wide association studies and beyond. Int J Mol Sci 2018; 19: 3702.
[http://dx.doi.org/10.3390/ijms19123702]
[51]
Cuyvers E, Sleegers K. Update on Alzheimer’s disease therapy and prevention strategies. Lancet Neurol 2016; 15: 857-68.
[http://dx.doi.org/10.1016/S1474-4422(16)00127-7 PMID: 27302364]
[52]
Graham WV, Bonito-Oliva A, Sakmar TP. Update on Alzheimer’s disease therapy and prevention strategies. Annu Rev Med 2017; 68: 413-30.
[http://dx.doi.org/10.1146/annurev-med-042915-103753 PMID: 28099083]
[53]
Howard R, McShane R, Lindesay J, et al. Donepezil and memantine for moderate-to-severe Alzheimer’s disease. N Engl J Med 2012; 366(10): 893-903.
[http://dx.doi.org/10.1056/NEJMoa1106668] [PMID: 22397651]
[54]
Schneider LS. Alzheimer disease pharmacologic treatment and treatment research. Continuum (Minneap Minn) 2013; 19(2): 339-57.
[http://dx.doi.org/10.1212/01.CON.0000429180.60095.d0 PMID: 23558481]
[55]
Grossberg GT, Manes F, Allegri RF, et al. The safety tolerability and efficacy of once-daily memantine (28 mg): a multinational randomized double-blind placebo-controlled trial in patients with moderate-to-severe Alzheimer’s disease taking cholinesterase inhibitors. CNS Drugs 2013; 27(6): 469-78.
[http://dx.doi.org/10.1007/s40263-013-0077-7] [PMID: 23733403]
[56]
Ito K, Tatebe T, Suzuki K, et al. Memantine reduces the production of amyloid-β peptides through modulation of amyloid precursor protein trafficking. Eur J Pharmacol 2017; 798: 16-25.
[http://dx.doi.org/10.1016/j.ejphar.2017.02.001] [PMID: 28167259]
[57]
Massoud F, Gauthier S. Update on the pharmacological treatment of Alzheimer’s disease. Curr Neuropharmacol 2010; 8(1): 69-80.
[http://dx.doi.org/10.2174/157015910790909520] [PMID: 20808547]
[58]
Jauch-Chara K, Friedrich A, Rezmer M, et al. Intranasal insulin suppresses food intake via enhancement of brain energy levels in humans. Diabetes 2012; 61(9): 2261-8.
[http://dx.doi.org/10.2337/db12-0025] [PMID: 22586589]
[59]
Frey D, Methods G. Methods pharmaceutical compositions and articles of manufacture for administering therapeutic cells to the animal central nervous system. United States Patent US 8, 283, 160 2012.
[60]
Md S, Bhattmisra SK, Zeeshan F, et al. Nano-carrier enabled drug delivery systems for nose to brain targeting for the treatment of neurodegenerative disorders. Drug Deliv Sci Technol 2018; 43: 295-310.
[http://dx.doi.org/10.1016/j.jddst.2017.09.022]
[61]
Gupta PP, Pandey RD, Jha D, Shrivastav V, Kumar S. Role of traditional nonsteroidal anti-inflammatory drugs in Alzheimer’s disease: a meta-analysis of randomized clinical trials. Am J Alzheimers Dis Other Demen 2015; 30(2): 178-82.
[http://dx.doi.org/10.1177/1533317514542644] [PMID: 25024454]
[62]
Lee LK, Shahar S, Chin AV, Yusoff NA. Docosahexaenoic acid-concentrated fish oil supplementation in subjects with Mild Cognitive Impairment (MCI): a 12-month randomised, double-blind, placebo-controlled trial. Psychopharmacology (Berl) 2013; 225(3): 605-12.
[http://dx.doi.org/10.1007/s00213-012-2848-0] [PMID: 22932777]
[63]
Bo Y, Zhang X, Wang Y, et al. The n-3 polyunsaturated fatty acids supplementation improved the cognitive function in the Chinese elderly with mild cognitive impairment: a double-blind randomized controlled trial. Nutrients 2017; 9(1): 54.
[http://dx.doi.org/10.3390/nu9010054] [PMID: 28075381]
[64]
Littlejohns TJ, Henley WE, Lang IA, et al. Vitamin D and the risk of dementia and Alzheimer disease. Neurology 2014; 83(10): 920-8.
[http://dx.doi.org/10.1212/WNL.0000000000000755 PMID: 25098535]
[65]
Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline. Alzheimers Dement (N Y) 2019; 5: 272-93.
[http://dx.doi.org/10.1016/j.trci.2019.05.008] [PMID: 31334330]
[66]
Crowe TP, Greenlee MHW, Kanthasamy AG, Hsu WH. Mechanism of intranasal drug delivery directly to the brain. Life Sci 2018; 195: 44-52.
[http://dx.doi.org/10.1016/j.lfs.2017.12.025] [PMID: 29277310]
[67]
Hanson LR, Frey WH. II. Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci 2008; 9(3): S5.
[http://dx.doi.org/10.1186/1471-2202-9-S3-S5] [PMID: 19091002]
[68]
Garcia-Garcia E, Andrieux K, Gil S, Couvreur P. Colloidal carriers and Blood-Brain Barrier (BBB) translocation: a way to deliver drugs to the brain? Int J Pharm 2005; 298(2): 274-92.
[http://dx.doi.org/10.1016/j.ijpharm.2005.03.031] [PMID: 15896933]
[69]
Erdő F, Bors LA, Farkas D, Bajza Á, Gizurarson S. Evaluation of intranasal delivery route of drug administration for brain targeting. Brain Res Bull 2018; 143: 155-70.
[http://dx.doi.org/10.1016/j.brainresbull.2018.10.009 PMID: 30449731]
[70]
Dhuria SV, Hanson LR, Frey WH II. Intranasal delivery to the central nervous system: mechanisms and experimental considerations. J Pharm Sci 2010; 99(4): 1654-73.
[http://dx.doi.org/10.1002/jps.21924] [PMID: 19877171]
[71]
Miyake MM, Bleier BS. The blood-brain barrier and nasal drug delivery to the central nervous system. Am J Rhinol Allergy 2015; 29(2): 124-7.
[http://dx.doi.org/10.2500/ajra.2015.29.4149] [PMID: 25785753]
[72]
Thorne RG, Frey WH II. Delivery of neurotrophic factors to the central nervous system: pharmacokinetic considerations. Clin Pharmacokinet 2001; 40(12): 907-46.
[http://dx.doi.org/10.2165/00003088-200140120-00003 PMID: 11735609]
[73]
Illum L. Is nose-to-brain transport of drugs in man a reality? J Pharm Pharmacol 2004; 56(1): 3-17.
[http://dx.doi.org/10.1211/0022357022539] [PMID: 14979996]
[74]
Kandimalla KK, Donovan MD. Localization and differential activity of P-glycoprotein in the bovine olfactory and nasal respiratory mucosae. Pharm Res 2005; 22(7): 1121-8.
[http://dx.doi.org/10.1007/s11095-005-5420-3] [PMID: 16028013]
[75]
Graff CL, Pollack GM. Functional evidence for P-glycoprotein at the nose-brain barrier. Pharm Res 2005; 22(1): 86-93.
[http://dx.doi.org/10.1007/s11095-004-9013-3] [PMID: 15771234]
[76]
Wioland MA, Fleury-Feith J, Corlieu P, et al. CFTR MDR1 and MRP1 immunolocalization in normal human nasal respiratory mucosa. J Histochem Cytochem 2000; 48(9): 1215-22.
[http://dx.doi.org/10.1177/002215540004800905] [PMID: 10950878]
[77]
Kandimalla KK, Donovan MD. Carrier mediated transport of chlorpheniramine and chlorcyclizine across bovine olfactory mucosa: implications on nose-to-brain transport. J Pharm Sci 2005; 94(3): 613-24.
[http://dx.doi.org/10.1002/jps.20284] [PMID: 15666293]
[78]
Sarkar MA. Drug metabolism in the nasal mucosa. Pharm Res 1992; 9(1): 1-9.
[http://dx.doi.org/10.1023/A:1018911206646] [PMID: 1589391]
[79]
Ritza KN. Shifting the paradigm: from bariatric surgery to metabolic Surgery. J Surg Acad 2015; 5: 1-3.
[80]
Türker S, Onur E, Ozer Y. Nasal route and drug delivery systems. Pharm World Sci 2004; 26(3): 137-42.
[http://dx.doi.org/10.1023/B:PHAR.0000026823.82950.ff PMID: 15230360]
[81]
Vyas TK, Babbar AK, Sharma RK, Singh S, Misra A. Intranasal mucoadhesive microemulsions of clonazepam: preliminary studies on brain targeting. J Pharm Sci 2006; 95(3): 570-80.
[http://dx.doi.org/10.1002/jps.20480] [PMID: 16419051]
[82]
Vyas TK, Babbar AK, Sharma RK, Singh S, Misra A. Preliminary brain-targeting studies on intranasal mucoadhesive microemulsions of sumatriptan. AAPS PharmSciTech 2006; 7(1): 49-57.
[http://dx.doi.org/10.1208/pt070108] [PMID: 28290023]
[83]
Kumar M, Misra A, Babbar AK, Mishra AK, Mishra P, Pathak K. Intranasal nanoemulsion based brain targeting drug delivery system of risperidone. Int J Pharm 2008; 358(1-2): 285-91.
[http://dx.doi.org/10.1016/j.ijpharm.2008.03.029] [PMID: 18455333]
[84]
Vyas TK, Babbar AK, Sharma RK, Misra A. Intranasal mucoadhesive microemulsions of zolmitriptan: preliminary studies on brain-targeting. J Drug Target 2005; 13(5): 317-24.
[http://dx.doi.org/10.1080/10611860500246217] [PMID: 16199375]
[85]
Zhang Q-Z, Zha L-S, Zhang Y, et al. The brain targeting efficiency following nasally applied MPEG-PLA nanoparticles in rats. J Drug Target 2006; 14(5): 281-90.
[http://dx.doi.org/10.1080/10611860600721051] [PMID: 16882548]
[86]
Wang X, He H, Leng W, Tang X. Evaluation of brain-targeting for the nasal delivery of estradiol by the microdialysis method. Int J Pharm 2006; 317(1): 40-6.
[http://dx.doi.org/10.1016/j.ijpharm.2006.02.055] [PMID: 16631329]
[87]
Wang X, Chi N, Tang X. Preparation of estradiol chitosan nanoparticles for improving nasal absorption and brain targeting. Eur J Pharm Biopharm 2008; 70(3): 735-40.
[http://dx.doi.org/10.1016/j.ejpb.2008.07.005] [PMID: 18684400]
[88]
Davis SS, Illum L. Absorption enhancers for nasal drug delivery. Clin Pharmacokinet 2003; 42(13): 1107-28.
[http://dx.doi.org/10.2165/00003088-200342130-00003 PMID: 14531723]
[89]
Chen SC, Eiting K, Cui K, et al. Therapeutic utility of a novel tight junction modulating peptide for enhancing intranasal drug delivery. J Pharm Sci 2006; 95(6): 1364-71.
[http://dx.doi.org/10.1002/jps.20510] [PMID: 16625659]
[90]
Dufes C, Olivier JC, Gaillard F, Gaillard A, Couet W, Muller JM. Brain delivery of vasoactive intestinal peptide (VIP) following nasal administration to rats. Int J Pharm 2003; 255(1-2): 87-97.
[http://dx.doi.org/10.1016/S0378-5173(03)00039-5 PMID: 12672605]
[91]
Sakane T, Akizuki M, Taki Y, Yamashita S, Sezaki H, Nadai T. Direct drug transport from the rat nasal cavity to the cerebrospinal fluid: the relation to the molecular weight of drugs. J Pharm Pharmacol 1995; 47(5): 379-81.
[http://dx.doi.org/10.1111/j.2042-7158.1995.tb05814.x PMID: 7494186]
[92]
Loftus LT, Li HF, Gray AJ, et al. In vivo protein transduction to the CNS. Neurosci 2006; 139(3): 1061-7.
[http://dx.doi.org/10.1016/j.neuroscience.2006.01.041 PMID: 16529872]
[93]
Charlton ST, Whetstone J, Fayinka ST, Read KD, Illum L, Davis SS. Evaluation of direct transport pathways of glycine receptor antagonists and an angiotensin antagonist from the nasal cavity to the central nervous system in the rat model. Pharm Res 2008; 25(7): 1531-43.
[http://dx.doi.org/10.1007/s11095-008-9550-2] [PMID: 18293062]
[94]
Gao X, Tao W, Lu W, et al. Lectin-conjugated PEG-PLA nanoparticles: preparation and brain delivery after intranasal administration. Biomaterials 2006; 27(18): 3482-90.
[http://dx.doi.org/10.1016/j.biomaterials.2006.01.038 PMID: 16510178]
[95]
Gao X, Chen J, Tao W, et al. UEA I-bearing nanoparticles for brain delivery following intranasal administration. Int J Pharm 2007; 340(1-2): 207-15.
[http://dx.doi.org/10.1016/j.ijpharm.2007.03.039] [PMID: 17499948]
[96]
Gao X, Wu B, Zhang Q, et al. Brain delivery of vasoactive intestinal peptide enhanced with the nanoparticles conjugated with wheat germ agglutinin following intranasal administration. J Control Release 2007; 121(3): 156-67.
[http://dx.doi.org/10.1016/j.jconrel.2007.05.026] [PMID: 17628165]
[97]
Hanafy AS, Farid RM, Helmy MW, ElGamal SS. Pharmacological, toxicological and neuronal localization assessment of galantamine/chitosan complex nanoparticles in rats: future potential contribution in Alzheimer’s disease management. Drug Deliv 2016; 23(8): 3111-22.
[http://dx.doi.org/10.3109/10717544.2016.1153748 PMID: 26942549]
[98]
Tosi G, Musumeci T, Ruozi B, et al. The fate of polymeric and lipid nanoparticles for brain delivery and targeting: strategies and mechanism of blood–brain barrier crossing and trafficking into the central nervous system. J Drug Deliv Sci Technol 2016; 32: 66-76.
[http://dx.doi.org/10.1016/j.jddst.2015.07.007]
[99]
Antimisiaris SG, Mourtas S, Markoutsa E, et al. Nanoparticles for diagnosis and/or treatment of Alzheimer’s disease.In: Advanced Healthcare Materials. Hoboken: Wiley & Sons, Inc. 2014; pp. 87-179.
[http://dx.doi.org/10.1002/9781118774205.ch4]
[100]
Wong HL, Wu XY, Bendayan R. Nanotechnological advances for the delivery of CNS therapeutics. Adv Drug Deliv Rev 2012; 64(7): 686-700.
[http://dx.doi.org/10.1016/j.addr.2011.10.007] [PMID: 22100125]
[101]
ElBayoumi TA, Torchilin VP. Current trends in liposome research. Methods Mol Biol 2010; 605: 1-27.
[http://dx.doi.org/10.1007/978-1-60327-360-2_1]
[102]
Brasnjevic I, Steinbusch HWM, Schmitz C, Martinez-Martinez P. European nanobiopharmaceutics research initiative. Delivery of peptide and protein drugs over the blood-brain barrier. Prog Neurobiol 2009; 87(4): 212-51.
[http://dx.doi.org/10.1016/j.pneurobio.2008.12.002 PMID: 19395337]
[103]
Al Asmari AK, Ullah Z, Tariq M, Fatani A. Preparation, characterization and in vivo evaluation of intranasally administered liposomal formulation of donepezil. Drug Des Devel Ther 2016; 10: 205-15.
[PMID: 26834457]
[104]
Yang ZZ, Zhang YQ, Wang ZZ, Wu K, Lou JN, Qi XR. Enhanced brain distribution and pharmacodynamics of rivastigmine by liposomes following intranasal administration. Int J Pharm 2013; 452(1-2): 344-54.
[http://dx.doi.org/10.1016/j.ijpharm.2013.05.009] [PMID: 23680731]
[105]
Li W, Zhou Y, Zhao N, Hao B, Wang X, Kong P. Pharmacokinetic behavior and efficiency of acetylcholinesterase inhibition in rat brain after intranasal administration of galanthamine hydrobromide loaded flexible liposomes. Environ Toxicol Pharmacol 2012; 34(2): 272-9.
[http://dx.doi.org/10.1016/j.etap.2012.04.012] [PMID: 22613079]
[106]
Sood S, Jain K, Gowthamarajan K. Optimization of curcumin nanoemulsion for intranasal delivery using design of experiment and its toxicity assessment. Colloids Surf B Biointerfaces 2014; 113: 330-7.
[http://dx.doi.org/10.1016/j.colsurfb.2013.09.030] [PMID: 24121076]
[107]
Nasr M. Development of an optimized hyaluronic acid-based lipidic nanoemulsion co-encapsulating two polyphenols for nose to brain delivery. Drug Deliv 2016; 23(4): 1444-52.
[http://dx.doi.org/10.3109/10717544.2015.1092619 PMID: 26401600]
[108]
Jaiswal M, Kumar A, Sharma SJ. Nanoemulsions loaded Carbopol® 934 based gel for intranasal delivery of neuroprotective centella asiatica extract: in-vitro and ex-vivo permeation study. Pharm Investig 2016; 46: 79-89.
[http://dx.doi.org/10.1007/s40005-016-0228-1]
[109]
Shah BM, Misra M, Shishoo CJ, Padh H. Nose to brain microemulsion-based drug delivery system of rivastigmine: formulation and ex-vivo characterization. Drug Deliv 2015; 22(7): 918-30.
[http://dx.doi.org/10.3109/10717544.2013.878857] [PMID: 24467601]
[110]
Pehlivan SB. Nanotechnology-based drug delivery systems for targeting imaging and diagnosis of neurodegenerative diseases. Pharm Res 2013; 30(10): 2499-511.
[http://dx.doi.org/10.1007/s11095-013-1156-7] [PMID: 23959851]
[111]
Klementieva O, Benseny-Cases N, Gella A, Appelhans D, Voit B, Cladera J. Dense shell glycodendrimers as potential nontoxic anti-amyloidogenic agents in Alzheimer’s disease. Amyloid-dendrimer aggregates morphology and cell toxicity. Biomacromolecules 2011; 12(11): 3903-9.
[http://dx.doi.org/10.1021/bm2008636] [PMID: 21936579]
[112]
Luppi B, Bigucci F, Corace G, et al. Albumin nanoparticles carrying cyclodextrins for nasal delivery of the anti-Alzheimer drug tacrine. Eur J Pharm Sci 2011; 44(4): 559-65.
[http://dx.doi.org/10.1016/j.ejps.2011.10.002] [PMID: 22009109]
[113]
Wong LR, Ho PC. Role of serum albumin as a nanoparticulate carrier for nose-to-brain delivery of R-flurbiprofen: implications for the treatment of Alzheimer’s disease. J Pharm Pharmacol 2018; 70(1): 59-69.
[http://dx.doi.org/10.1111/jphp.12836] [PMID: 29034965]
[114]
Silva AC, González-Mira E, Lobo JM, Amaral MH. Current progresses on nanodelivery systems for the treatment of neuropsychiatric diseases: Alzheimer’s and schizophrenia. Curr Pharm Des 2013; 19(41): 7185-95.
[http://dx.doi.org/10.2174/138161281941131219123329 PMID: 23489198]
[115]
Naahidi S, Jafari M, Edalat F, Raymond K, Khademhosseini A, Chen P. Biocompatibility of engineered nanoparticles for drug delivery. J Control Release 2013; 166(2): 182-94.
[http://dx.doi.org/10.1016/j.jconrel.2012.12.013] [PMID: 23262199]
[116]
Patel A, Patel M, Yang X, Mitra AK. Recent advances in protein and Peptide drug delivery: a special emphasis on polymeric nanoparticles. Protein Pept Lett 2014; 21(11): 1102-20.
[http://dx.doi.org/10.2174/0929866521666140807114240 PMID: 25106908]
[117]
Fazil M, Md S, Haque S, et al. Development and evaluation of rivastigmine loaded chitosan nanoparticles for brain targeting. Eur J Pharm Sci 2012; 47(1): 6-15.
[http://dx.doi.org/10.1016/j.ejps.2012.04.013] [PMID: 22561106]
[118]
Hanafy AS, Farid RM, ElGamal SS. Complexation as an approach to entrap cationic drugs into cationic nanoparticles administered intranasally for Alzheimer’s disease management: preparation and detection in rat brain. Drug Dev Ind Pharm 2015; 41(12): 2055-68.
[http://dx.doi.org/10.3109/03639045.2015.1062897 PMID: 26133084]
[119]
Bhavna Md S, Ali M, et al. Donepezil nanosuspension intended for nose to brain targeting: in vitro and in vivo safety evaluation. Int J Biol Macromol 2014; 67: 418-25.
[http://dx.doi.org/10.1016/j.ijbiomac.2014.03.022] [PMID: 24705169]
[120]
Muntimadugu E, Dhommati R, Jain A, Challa VG, Shaheen M, Khan W. Intranasal delivery of nanoparticle encapsulated tarenflurbil: a potential brain targeting strategy for Alzheimer’s disease. Eur J Pharm Sci 2016; 92: 224-34.
[http://dx.doi.org/10.1016/j.ejps.2016.05.012] [PMID: 27185298]
[121]
Zhang C, Chen J, Feng C, et al. Intranasal nanoparticles of basic fibroblast growth factor for brain delivery to treat Alzheimer’s disease. Int J Pharm 2014; 461(1-2): 192-202.
[http://dx.doi.org/10.1016/j.ijpharm.2013.11.049] [PMID: 24300213]
[122]
Meng Q, Wang A, Hua H, et al. Intranasal delivery of Huperzine A to the brain using lactoferrin-conjugated N-trimethylated chitosan surface-modified PLGA nanoparticles for treatment of Alzheimer’s disease. Int J Nanomedicine 2018; 13: 705-18.
[http://dx.doi.org/10.2147/IJN.S151474] [PMID: 29440896]
[123]
Sunena, Singh SK, Mishra DN. Nose to brain delivery of galantamine loaded nanoparticles: in-vivo pharmacodynamic and biochemical study in mice. Curr Drug Deliv 2019; 16(1): 51-8.
[http://dx.doi.org/10.2174/1567201815666181004094707 PMID: 30289074]
[124]
Elnaggar YSR, Etman SM, Abdelmonsif DA, et al. Intranasal piperine-loaded chitosan nanoparticles as brain-targeted therapy in Alzheimer’s disease: optimization biological efficacy and potential toxicity. J Pharm Sci 2015; 104: 3544-56.
[http://dx.doi.org/10.1002/jps.24557]
[125]
Shah B, Khunt D, Bhatt H, Misra M, Padh H. Application of quality by design approach for intranasal delivery of rivastigmine loaded solid lipid nanoparticles: effect on formulation and characterization parameters. Eur J Pharm Sci 2015; 78: 54-66.
[http://dx.doi.org/10.1016/j.ejps.2015.07.002] [PMID: 26143262]
[126]
Zhao Y, Wang Y, Ran F, et al. A comparison between sphere and rod nanoparticles regarding their in vivo biological behavior and pharmacokinetics. Sci Rep 2017; 7(1): 4131.
[http://dx.doi.org/10.1038/s41598-017-03834-2] [PMID: 28646143]
[127]
Bednarski M, Dudek M, Knutelska J, et al. The influence of the route of administration of gold nanoparticles on their tissue distribution and basic biochemical parameters: in vivo studies. Pharmacol Rep 2015; 67(3): 405-9.
[http://dx.doi.org/10.1016/j.pharep.2014.10.019] [PMID: 25933945]
[128]
Bahadar H, Maqbool F, Niaz K, Abdollahi M. Toxicity of nanoparticles and an overview of current experimental models. Iran Biomed J 2016; 20(1): 1-11.
[PMID: 26286636]
[129]
Zhang B, Sai Lung P, Zhao S, Chu Z, Chrzanowski W, Li Q. Shape dependent cytotoxicity of PLGA-PEG nanoparticles on human cells. Sci Rep 2017; 7(1): 7315.
[http://dx.doi.org/10.1038/s41598-017-07588-9] [PMID: 28779154]
[130]
Kermanizadeh A, Jacobsen NR, Roursgaard M, Loft S, Møller P. Hepatic toxicity assessment of cationic liposome exposure in healthy and chronic alcohol fed mice. Heliyon 2017; 3(11)e00458
[http://dx.doi.org/10.1016/j.heliyon.2017.e00458] [PMID: 29234737]
[131]
Gorain B, Choudhury H, Tekade RK, Karan S, Jaisankar P, Pal TK. Comparative biodistribution and safety profiling of olmesartan medoxomil oil-in-water oral nanoemulsion. Regul Toxicol Pharmacol 2016; 82: 20-31.
[http://dx.doi.org/10.1016/j.yrtph.2016.10.020] [PMID: 27815174]
[132]
Choudhury H, Gorain B, Tekade RK, Pandey M, Karmakar S, Pal TK. Safety against nephrotoxicity in paclitaxel treatment: oral nanocarrier as an effective tool in preclinical evaluation with marked in vivo antitumor activity. Regul Toxicol Pharmacol 2017; 91: 179-89.
[http://dx.doi.org/10.1016/j.yrtph.2017.10.023] [PMID: 29080846]
[133]
Hossen S, Hossain MK, Basher MK, Mia MNH, Rahman MT, Uddin MJ. Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: a review. J Adv Res 2018; 15: 1-18.
[http://dx.doi.org/10.1016/j.jare.2018.06.005] [PMID: 30581608]
[134]
Gabal YM, Kamel AO, Sammour OA, Elshafeey AH. Effect of surface charge on the brain delivery of nanostructured lipid carriers in situ gels via the nasal route. Int J Pharm 2014; 473(1-2): 442-57.
[http://dx.doi.org/10.1016/j.ijpharm.2014.07.025] [PMID: 25062866]
[135]
Jia YP, Ma BY, Wei XW, et al. The in vitro and in vivo toxicity of gold nanoparticles. Chin Chem Lett 2017; 28: 691-702.
[http://dx.doi.org/10.1016/j.cclet.2017.01.021]
[136]
Casettari L, Illum L. Chitosan in nasal delivery systems for therapeutic drugs. J Control Release 2014; 190: 189-200.
[http://dx.doi.org/10.1016/j.jconrel.2014.05.003] [PMID: 24818769]
[137]
Mistry A, Stolnik S, Illum L. Nose-to-brain delivery: investigation of the transport of nanoparticles with different surface characteristics and sizes in excised porcine olfactory epithelium. Mol Pharm 2015; 12(8): 2755-66.
[http://dx.doi.org/10.1021/acs.molpharmaceut.5b00088 PMID: 25997083]
[138]
Sonvico F, Clementino A, Buttini F, et al. Surface-modified nanocarriers for nose-to-brain delivery: from bioadhesion to targeting. Pharmaceutics 2018; 10(1): 34.
[http://dx.doi.org/10.3390/pharmaceutics10010034 PMID: 29543755]
[139]
Patel D, Naik S, Misra A. Improved transnasal transport and brain uptake of tizanidine HCl-loaded thiolated chitosan nanoparticles for alleviation of pain. J Pharm Sci 2012; 101(2): 690-706.
[http://dx.doi.org/10.1002/jps.22780] [PMID: 22006260]
[140]
Patel D, Naik S, Chuttani K, Mathur R, Mishra AK, Misra A. Intranasal delivery of cyclobenzaprine hydrochloride-loaded thiolated chitosan nanoparticles for pain relief. J Drug Target 2013; 21(8): 759-69.
[http://dx.doi.org/10.3109/1061186X.2013.818676 PMID: 23879335]
[141]
Jain R, Nabar S, Dandekar P, Patravale V. Micellar nanocarriers: potential nose-to-brain delivery of zolmitriptan as novel migraine therapy. Pharm Res 2010; 27(4): 655-64.
[http://dx.doi.org/10.1007/s11095-009-0041-x] [PMID: 20151180]
[142]
Jain R, Nabar S, Dandekar P, et al. Formulation and evaluation of novel micellar nanocarrier for nasal delivery of sumatriptan. Nanomedicine (Lond) 2010; 5(4): 575-87.
[http://dx.doi.org/10.2217/nnm.10.28] [PMID: 20528453]
[143]
Wen Z, Yan Z, He R, et al. Brain targeting and toxicity study of odorranalectin-conjugated nanoparticles following intranasal administration. Drug Deliv 2011; 18(8): 555-61.
[http://dx.doi.org/10.3109/10717544.2011.596583] [PMID: 21812752]
[144]
Liu Q, Shao X, Chen J, et al. In vivo toxicity and immunogenicity of wheat germ agglutinin conjugated poly(ethylene glycol)-poly(lactic acid) nanoparticles for intranasal delivery to the brain. Toxicol Appl Pharmacol 2011; 251(1): 79-84.
[http://dx.doi.org/10.1016/j.taap.2010.12.003] [PMID: 21163285]
[145]
Aderibigbe BA, Naki T. Chitosan-based nanocarriers for nose to brain delivery. Appl Sci (Basel) 2019; 9: 2219.
[http://dx.doi.org/10.3390/app9112219]
[146]
Gorain B, Rajeswary DC, Pandey M, Kesharwani P, Kumbhar SA, Choudhury H. Nose to brain delivery of nanocarriers towards attenuation of demented condition. Curr Pharm Des 2020; 26(19): 2233-46.
[http://dx.doi.org/10.2174/1381612826666200313125613 PMID: 32167424]
[147]
Djupesland PG. Nasal drug delivery devices: characteristics and performance in a clinical perspective-a review. Drug Deliv Transl Res 2013; 3(1): 42-62.
[http://dx.doi.org/10.1007/s13346-012-0108-9] [PMID: 23316447]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy