Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

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

Review Article

Transdermal Nutraceuticals Delivery System for CNS Disease

Author(s): Mohammed Asadullah Jahangir, Pooja Jain, Rishabh Verma, Mohamad Taleuzzaman*, Mohamed Jawed Ahsan, Ananda Kumar Chettupalli, Abdul Muheem and Mohd. Aamir Mirza*

Volume 21, Issue 10, 2022

Published on: 12 January, 2022

Page: [977 - 993] Pages: 17

DOI: 10.2174/1871527321666220112154051

Price: $65

Abstract

Herbal medicines are being used by humans since the oldest civilizations and have been an integral part of traditional and alternative medicines. In recent times, pharmaceutical and biomedical scientists are taking interest in developing nutraceutical-based medicines to overcome the side effects and adverse drug reactions caused by allopathic medicines. Nutraceuticals have started occupying the global market. Nutraceuticals have gained widespread acceptance due to their efficacy in treating difficult to treat diseases, low toxicity, low cost, easy accessibility, etc. Safety and efficacy are other important factors in the commercialization process of nutraceuticals. Different novel advanced drug delivery systems have been constantly studied to improve the efficacy and bioavailability of medicines obtained from herbal sources. The transdermal drug delivery system provides a potent alternative to the conventional method of using nutraceuticals. The development of transdermal system-based nutraceuticals could provide the advantage of enhanced bioavailability, improved solubility, bypass of the first-pass metabolism, and targeted delivery of drugs in brain-related disorders. It additionally provides the advantage of being non-invasive.

This article reviews the potential effects of various nutraceuticals in brain-related disorders as well as trends in transdermal nano-systems to deliver such nutraceuticals. We have also focused on advantages, applications as well as recent United States-based patents which emphasize emerging interest towards transdermal nutraceuticals in brain disorders.

Keywords: Nutraceuticals, transdermal, drug delivery, herbal drugs, brain, neuronal disease.

« Previous
Graphical Abstract
[1]
Das L, Bhaumik E, Raychaudhuri U, Chakraborty R. Role of nutraceuticals in human health. J Food Sci Technol 2012; 49(2): 173-83.
[http://dx.doi.org/10.1007/s13197-011-0269-4] [PMID: 23572839]
[2]
Dickinson A. History and overview of DSHEA. Fitoterapia 2011; 82(1): 5-10.
[http://dx.doi.org/10.1016/j.fitote.2010.09.001] [PMID: 20837113]
[3]
Ruchi S. Role of nutraceuticals in health care: A review. Int J Green Pharm 2017; 11(3): S385-94.
[http://dx.doi.org/10.22377/ijgp.v11i03.1146]
[4]
Rai VK, Gupta GD, Pottoo FH, Barkat MA. Potential of Nano-Structured Drug Delivery System for Phytomedicine Delivery. Nanophytomedicine. Singapore: Springer 2020; pp. 89-111.
[http://dx.doi.org/10.1007/978-981-15-4909-0_6]
[5]
Helal NA, Eassa HA, Amer AM, Eltokhy MA, Edafiogho I, Nounou MI. Nutraceuticals’ novel formulations: The good, the bad, the unknown and patents involved. Recent Pat Drug Deliv Formul 2019; 13(2): 105-56.
[http://dx.doi.org/10.2174/1872211313666190503112040] [PMID: 31577201]
[6]
Rai VK, Narang RK, Pottoo FH, Barkat MA. Pharmacokinetics, Interaction, and Toxicological Profile of Nanophytomedicine. Nanophytomedicine. Singapore: Springer 2020; pp. 133-49.
[http://dx.doi.org/10.1007/978-981-15-4909-0_8]
[7]
Nicoli MC, Anese M, Parpinel M. Influence of processing on the antioxidant properties of fruits and vegetables. Trends Food Sci Technol 1999; 10: 94-100.
[http://dx.doi.org/10.1016/S0924-2244(99)00023-0]
[8]
Kaur C, Kapoor HC. Antioxidants in fruits and vegetables-the millenium’s health. Int J Food Sci Technol 2001; 36: 703-25.
[http://dx.doi.org/10.1046/j.1365-2621.2001.00513.x]
[9]
Jahangir MA, Muheem A, Imam SS. Nigella Sativa encapsulated nano scaffold and their bioactivity significance. Biomarkers as Targeted Herbal Drug Discovery: A Pharmacological Approach to Nanomedicines. 1st Ed. Boca Raton: Apple Academic Press 2020.
[10]
Malik A. The potentials of Nutraceuticals. Pharmainfonet 6 Metchinkoff E (1907) The prolongation of life. New York: Putmans Sons 2008; pp. 151-83.
[11]
Dureja H, Kaushik D, Kumar V. Developments in nutraceuticals. Indian J Pharmacol 2003; 35: 363-72.
[12]
Sharma A, Yadav KS, Pottoo FH, Rai VK, Barkat MA. Nanomedicine Based Phytoformulation in Disease Diagnosis and Treatment. Nanophytomedicine. Singapore: Springer 2020; pp. 193-218.
[13]
Stellavato A, Pirozzi AVA, de Novellis F, et al. In vitro assessment of nutraceutical compounds and novel nutraceutical formulations in a liver-steatosis-based model. Lipids Health Dis 2018; 17(1): 24.
[http://dx.doi.org/10.1186/s12944-018-0663-2] [PMID: 29402273]
[14]
Bourbon AI, Pinheiro AC, Cerqueira MA, Vicente AA. In vitro digestion of lactoferrin-glycomacropeptide nanohydrogels incorporating bioactive compounds: Effect of a chitosan coating. Food Hydrocoll 2018; 84: 267-75.
[http://dx.doi.org/10.1016/j.foodhyd.2018.06.015]
[15]
Zhang X, Liu J, Qian C, Kan J, Jin CH. Effect of grafting method on the physical property and antioxidant potential of chitosan film functionalized with gallic acid. Food Hydrocoll 2019; 89: 1-10.
[http://dx.doi.org/10.1016/j.foodhyd.2018.10.023]
[16]
Jahangir MA, Anand C, Muheem A, et al. Nano phytomedicine based delivery system for CNS disease. Curr Drug Metab 2020; 21(9): 661-73.
[http://dx.doi.org/10.2174/1389200221666200523161003] [PMID: 32445453]
[17]
Chao J, Leung Y, Wang M, Chang RC. Nutraceuticals and their preventive or potential therapeutic value in Parkinson’s disease. Nutr Rev 2012; 70(7): 373-86.
[http://dx.doi.org/10.1111/j.1753-4887.2012.00484.x] [PMID: 22747840]
[18]
Shin GH, Li J, Cho JH, Kim JT, Park HJ. Enhancement of curcumin solubility by phase change from crystalline to amorphous in Cur-TPGS nanosuspension. J Food Sci 2016; 81(2): N494-501.
[http://dx.doi.org/10.1111/1750-3841.13208] [PMID: 26766628]
[19]
Liu F, Zhu Z, Ma C, et al. Fabrication of concentrated fish oil emulsions using dual-channelmicrofluidization: Impact of droplet concentration on physical properties and lipid oxidation. J Agric Food Chem 2016; 64(50): 9532-41.
[http://dx.doi.org/10.1021/acs.jafc.6b04413] [PMID: 27936671]
[20]
Uluata S, McClements DJ, Decker EA. Physical stability, autoxidation, and photosensitized oxidation of omega-3 oils in nanoemulsions prepared with natural and synthetic surfactants. J Agric Food Chem 2015; 63(42): 9333-40.
[http://dx.doi.org/10.1021/acs.jafc.5b03572] [PMID: 26452408]
[21]
Tan S, Ebrahimi A, Langrish T. Controlled release of caffeine from tablets of spray-dried casein gels. Food Hydrocoll 2019; 88: 13-20.
[http://dx.doi.org/10.1016/j.foodhyd.2018.09.038]
[22]
Kay DG, Maclellan A. Composition and method for improving cognitive function and brain bioavailability of ginseng and ginsenosides and treating neurodegenerative disease and neurological disorders. WO2018148821A1, 2018.
[23]
Pottoo FH, Sharma S, Javed MN, et al. Lipid-based nanoformulations in the treatment of neurological disorders. Drug Metab Rev 2020; 52(1): 185-204.
[http://dx.doi.org/10.1080/03602532.2020.1726942] [PMID: 32116044]
[24]
Raman S, Mahmood S, Hilles AR, Javed MN, Azmana M, Al-Japairai KAS. Polymeric nanoparticles for brain drug delivery - A review. Curr Drug Metab 2020; 21(9): 649-60.
[http://dx.doi.org/10.2174/1389200221666200508074348] [PMID: 32384025]
[25]
Wani TU, Mohi-Ud-Din R, Majeed A, Kawoosa S, Pottoo FH. Skin permeation of nanoparticles: Mechanisms involved and critical factors governing topical drug delivery. Curr Pharm Des 2020; 26(36): 4601-14.
[http://dx.doi.org/10.2174/1381612826666200701204010] [PMID: 32611291]
[26]
Aslam M, Javed MN, Deeb HH, et al. Lipid carriers mediated targeted delivery of nutraceuticals: Challenges, role of blood brain barrier and promises of nanotechnology based approaches in neuronal disorders. Curr Drug Metab 2020 (Withdrawn).
[http://dx.doi.org/10.2174/1389200221999200728143511]
[27]
Iqubal MK, Saleem S, Iqubal A, et al. Natural, synthetic and their combinatorial nanocarriers based drug delivery system in treatment paradigm for wound healing via dermal targeting. Curr Pharm Des 2020; 26(36): 4551-68.
[http://dx.doi.org/10.2174/1381612826666200612164511] [PMID: 32532188]
[28]
Pottoo FH, Javed N, Rahman J, Abu-Izneid T, Khan FA. Targeted delivery of miRNA based therapeuticals in the clinical management of glioblastoma multiforme. Seminars in Cancer Biology. Cambridge: Academic Press 2020.
[29]
Pottoo FH, Barkat MA, Ansari MA, Javed MN, Jamal QM, Kamal MA. Nanotechnological based miRNA intervention in the therapeutic management of neuroblastoma. Seminars in Cancer Biology. Cambridge: Academic Press 2019.
[http://dx.doi.org/10.1016/j.semcancer.2019.09.017]
[30]
Isaac M, Holvey C. Transdermal patches: the emerging mode of drug delivery system in psychiatry. Ther Adv Psychopharmacol 2012; 2(6): 255-63.
[http://dx.doi.org/10.1177/2045125312458311] [PMID: 23983984]
[31]
Magner LN. A History of Medicine. 2nd ed. Boca Raton, FL: Taylor & Francis Group 2005.
[http://dx.doi.org/10.1201/b14213]
[32]
Geller MJ. Ancient Babylonian Medicine. 1st ed. Malden, MA: Wiley-Blackwell 2010.
[http://dx.doi.org/10.1002/9781444319996]
[33]
Mumtaz SM, Bhardwaj G, Goswami S, Tonk RK, Goyal RK, Pottoo FH. Management of glioblastoma multiforme by phytochemicals: Applications of nanoparticle based targeted drug delivery system. Curr Drug Targets 2021; 22(4): 429-42.
[http://dx.doi.org/10.2174/1389450121666200727115454] [PMID: 32718288]
[34]
Samala S, Veeresham C. Boswellic acids pretreatment enhances the bioavailability and hypoglycemic action of metformin in rats: Involvement of CYP3A inhibition. Int J Pharm Pharmacol 2018; 2: 132.
[35]
Mir RH, Shah AJ, Mohi-Ud-Din R, et al. Natural anti-inflammatory compounds as drug candidates in Alzheimer’s disease. Curr Med Chem 2021; 28(23): 4799-825.
[http://dx.doi.org/10.2174/0929867327666200730213215] [PMID: 32744957]
[36]
Alamgir ANM. Classification of Drugs, Nutraceuticals, Functional Food, and Cosmeceuticals; Proteins, Peptides, and Enzymes as Drugs. Therapeutic Use of Medicinal Plants and Their Extracts: Volume 1 Progress in Drug Research. Cham: Springer 2017; 73.
[37]
Chauhan B, Kumar G, Kalam N, Ansari SH. Current concepts and prospects of herbal nutraceutical: A review. J Adv Pharm Technol Res 2013; 4(1): 4-8.
[http://dx.doi.org/10.4103/2231-4040.107494] [PMID: 23662276]
[38]
Guilbot A, Bangratz M, Ait Abdellah S, Lucas C. A combination of coenzyme Q10, feverfew and magnesium for migraine prophylaxis: A prospective observational study. BMC Complement Altern Med 2017; 17(1): 433.
[http://dx.doi.org/10.1186/s12906-017-1933-7] [PMID: 28854909]
[39]
Rajapakse T, Pringsheim T. Nutraceuticals in migraine: A summary of existing guidelines for use. Headache 2016; 56(4): 808-16.
[http://dx.doi.org/10.1111/head.12789] [PMID: 26954394]
[40]
Campagnolo N, Johnston S, Collatz A, Staines D, Marshall- Gradisnik S. Dietary and nutrition interventions for the therapeutic treatment of chronic fatigue syndrome/myalgic encephalomyelitis: A systematic review. J Hum Nutr Diet 2017; 30(3): 247-59.
[http://dx.doi.org/10.1111/jhn.12435] [PMID: 28111818]
[41]
Castro-Marrero J, Sáez-Francàs N, Segundo MJ, et al. Effect of coenzyme Q10 plus nicotinamide adenine dinucleotide supplementation on maximum heart rate after exercise testing in chronic fatigue syndrome - A randomized, controlled, double-blind trial. Clin Nutr 2016; 35(4): 826-34.
[http://dx.doi.org/10.1016/j.clnu.2015.07.010] [PMID: 26212172]
[42]
Mischley LK, Lau RC, Bennett RD. Role of diet and nutritional supplements in Parkinson’s disease progression. Oxid Med Cell Longev 2017; 2017: 6405278.
[http://dx.doi.org/10.1155/2017/6405278] [PMID: 29081890]
[43]
Gaul C, Diener HC, Danesch U. Improvement of migraine symptoms with a proprietary supplement containing riboflavin, magnesium and Q10: A randomized, placebo-controlled, double-blind, multicenter trial. J Headache Pain 2015; 16: 516.
[http://dx.doi.org/10.1186/s10194-015-0516-6] [PMID: 25916335]
[44]
Bjørklund G, Dadar M, Pen JJ, Chirumbolo S, Aaseth J. Chronic fatigue syndrome (CFS): Suggestions for a nutritional treatment in the therapeutic approach. Biomed Pharmacother 2019; 109: 1000-7.
[http://dx.doi.org/10.1016/j.biopha.2018.10.076] [PMID: 30551349]
[45]
Curtis L, Epstein P. Nutritional treatment for acute and chronic traumatic brain injury patients. J Neurosurg Sci 2014; 58(3): 151-60.
[PMID: 24844176]
[46]
Perry E, Howes MJ. Medicinal plants and dementia therapy: herbal hopes for brain aging? CNS Neurosci Ther 2011; 17(6): 683-98.
[http://dx.doi.org/10.1111/j.1755-5949.2010.00202.x] [PMID: 22070157]
[47]
Wightman EL. Potential benefits of phytochemicals against Alzheimer’s disease. Proc Nutr Soc 2017; 76(2): 106-12.
[http://dx.doi.org/10.1017/S0029665116002962] [PMID: 28143625]
[48]
Kennedy DO, Wightman EL. Herbal extracts and phytochemicals: plant secondary metabolites and the enhancement of human brain function. Adv Nutr 2011; 2(1): 32-50.
[http://dx.doi.org/10.3945/an.110.000117] [PMID: 22211188]
[49]
Damar U, Gersner R, Johnstone JT, Schachter S, Rotenberg A. Huperzine A as a neuroprotective and antiepileptic drug: a review of preclinical research. Expert Rev Neurother 2016; 16(6): 671-80.
[http://dx.doi.org/10.1080/14737175.2016.1175303] [PMID: 27086593]
[50]
Li C, Shi S. Neuroprotective effect of huperzine A on d-galactose-induced hearing dysfunction. Ear Nose Throat J 2019; 100(Suppl_3): 269S-76S.
[http://dx.doi.org/10.1177/0145561319864570] [PMID: 31554431]
[51]
Tabira T, Kawamura N. A study of a supplement containing huperzine A and curcumin in dementia patients and individuals with mild cognitive impairment. J Alzheimers Dis 2018; 63(1): 75-8.
[http://dx.doi.org/10.3233/JAD-171154] [PMID: 29614668]
[52]
Huang P, Li B, Guo YH, Feng S, Hu J, Liu QQ. Efficacy and safety of huperzine A in treating patients with mild cognitive impairment: A systematic review and Meta-analysis. Zhongguo Zhongyao Zazhi 2019; 44(3): 582-8.
[PMID: 30989926]
[53]
Li J, Yue M, Zhou D, Wang M, Zhang H. Abcb1a but not Abcg2 played a predominant role in limiting the brain distribution of huperzine A in mice. Food Chem Toxicol 2017; 107(Pt A): 68-73.
[http://dx.doi.org/10.1016/j.fct.2017.06.005]
[54]
Qian ZM, Ke Y, Huperzine A. Huperzine A: Is it an effective disease-modifying drug for Alzheimer’s disease? Front Aging Neurosci 2014; 6: 216.
[http://dx.doi.org/10.3389/fnagi.2014.00216] [PMID: 25191267]
[55]
Mao XY, Zhou HH, Li X, Liu ZQ, Huperzine A. Huperzine A Alleviates oxidative glutamate toxicity in hippocampal HT22 Cells via activating BDNF/TrkB-dependent PI3K/Akt/mTOR signaling pathway. Cell Mol Neurobiol 2016; 36(6): 915-25.
[http://dx.doi.org/10.1007/s10571-015-0276-5] [PMID: 26440805]
[56]
Poulose SM, Miller MG, Scott T, Shukitt-Hale B. Nutritional factors affecting adult neurogenesis and cognitive function. Adv Nutr 2017; 8(6): 804-11.
[http://dx.doi.org/10.3945/an.117.016261] [PMID: 29141966]
[57]
Kelly E, Vyas P, Weber JT. Biochemical properties and neuroprotective effects of compounds in various species of berries. Molecules 2017; 23(1): 26.
[http://dx.doi.org/10.3390/molecules23010026] [PMID: 29271934]
[58]
Dal-Pan A, Dudonné S, Bourassa P, et al. Cognitive-enhancing effects of a polyphenols-rich extract from fruits without changes in neuropathology in an animal model of Alzheimer’s disease. J Alzheimers Dis 2017; 55(1): 115-35.
[http://dx.doi.org/10.3233/JAD-160281] [PMID: 27662290]
[59]
Businaro R, Corsi M, Asprino R, et al. Modulation of inflammation as a way of delaying Alzheimer’s disease progression: The diet’s role. Curr Alzheimer Res 2018; 15(4): 363-80.
[http://dx.doi.org/10.2174/1567205014666170829100100] [PMID: 28847284]
[60]
Strathearn KE, Yousef GG, Grace MH, et al. Neuroprotective effects of anthocyanin- and proanthocyanidin-rich extracts in cellular models of Parkinson׳s disease. Brain Res 2014; 1555: 60-77.
[http://dx.doi.org/10.1016/j.brainres.2014.01.047] [PMID: 24502982]
[61]
Liu H, Zhao L, Yue L, et al. Pterostilbene attenuates early brain injury following subarachnoid hemorrhage via inhibition of the NLRP3 inflammasome and Nox2-related oxidative stress. Mol Neurobiol 2017; 54(8): 5928-40.
[http://dx.doi.org/10.1007/s12035-016-0108-8] [PMID: 27665283]
[62]
Bosch-Morell F, Villagrasa V, Ortega T, et al. Medicinal plants and natural products as neuroprotective agents in age-related macular degeneration. Neural Regen Res 2020; 15(12): 2207-16.
[http://dx.doi.org/10.4103/1673-5374.284978] [PMID: 32594032]
[63]
Siddiqui MJ, Saleh MSM, Basharuddin SNBB, et al. Saffron (Crocus sativus L.): As an antidepressant. J Pharm Bioallied Sci 2018; 10(4): 173-80.
[http://dx.doi.org/10.4103/JPBS.JPBS_83_18] [PMID: 30568374]
[64]
Finley JW, Gao S. A perspective on Crocus sativus L. (Saffron) constituent crocin: A potent water-soluble antioxidant and potential therapy for Alzheimer’s disease. J Agric Food Chem 2017; 65(5): 1005-20.
[http://dx.doi.org/10.1021/acs.jafc.6b04398] [PMID: 28098452]
[65]
Muheem A, Shakeel F, Jahangir MA, et al. A review on the strategies for oral delivery of proteins and peptides and their clinical perspectives. Saudi Pharm J 2016; 24(4): 413-28.
[http://dx.doi.org/10.1016/j.jsps.2014.06.004]
[66]
Muheem A, Jahangir MA, Jaiswal CP, et al. Recent patents, regulatory issues and toxicity of nanoparticles in neuronal disorders. Curr Drug Metab 2021; 22(4): 263-79.
[PMID: 33305703]
[67]
Hausenblas HA, Saha D, Dubyak PJ, Anton SD. Saffron (Crocus sativus L.) and major depressive disorder: A meta-analysis of randomized clinical trials. J Integr Med 2013; 11(6): 377-83.
[http://dx.doi.org/10.3736/jintegrmed2013056] [PMID: 24299602]
[68]
Khazdair MR, Boskabady MH, Hosseini M, Rezaee R, M Tsatsakis A. The effects of Crocus sativus (saffron) and its constituents on nervous system: A review. Avicenna J Phytomed 2015; 5(5): 376-91.
[PMID: 26468457]
[69]
Pitsikas N. Constituents of saffron (Crocus sativus L.) as potential candidates for the treatment of anxiety disorders and schizophrenia. Molecules 2016; 21(3): 303.
[http://dx.doi.org/10.3390/molecules21030303] [PMID: 26950102]
[70]
Batarseh YS, Bharate SS, Kumar V, et al. Crocus sativus extract tightens the blood-brain barrier, reduces amyloid β load and related toxicity in 5XFAD mice. ACS Chem Neurosci 2017; 8(8): 1756-66.
[http://dx.doi.org/10.1021/acschemneuro.7b00101] [PMID: 28471166]
[71]
Yun HM, Ban JO, Park KR, et al. Potential therapeutic effects of functionally active compounds isolated from garlic. Pharmacol Ther 2014; 142(2): 183-95.
[http://dx.doi.org/10.1016/j.pharmthera.2013.12.005] [PMID: 24333688]
[72]
Ray B, Chauhan NB, Lahiri DK. Oxidative insults to neurons and synapse are prevented by aged garlic extract and S-allyl-L-cysteine treatment in the neuronal culture and APP-Tg mouse model. J Neurochem 2011; 117(3): 388-402.
[http://dx.doi.org/10.1111/j.1471-4159.2010.07145.x] [PMID: 21166677]
[73]
Lin X, Yu S, Chen Y, Wu J, Zhao J, Zhao Y. Neuroprotective effects of diallyl sulfide against transient focal cerebral ischemia via anti-apoptosis in rats. Neurol Res 2012; 34(1): 32-7.
[http://dx.doi.org/10.1179/1743132811Y.0000000054] [PMID: 22196859]
[74]
Gilani SJ, Imam SS, Jafer M, Alshehri S, Taleuzzaman M, Jahangir MA. Curcumin Nanomedicines and Their Application in the Management of Disease. Biomarkers as Targeted Herbal Drug Discovery: A Pharmacological Approach to Nanomedicines. Boca Raton: Apple Academic Press 2020.
[75]
Deng TS. Biological clocks, some clock-related diseases, and medicinal plants. PsyCh J 2018; 7(4): 197-205.
[http://dx.doi.org/10.1002/pchj.263] [PMID: 30561856]
[76]
Witkin JM, Li X. Curcumin, an active constiuent of the ancient medicinal herb Curcuma longa L.: some uses and the establishment and biological basis of medical efficacy. CNS Neurol Disord Drug Targets 2013; 12(4): 487-97.
[http://dx.doi.org/10.2174/1871527311312040007] [PMID: 23574161]
[77]
Karimian MS, Pirro M, Majeed M, Sahebkar A. Curcumin as a natural regulator of monocyte chemoattractant protein-1. Cytokine Growth Factor Rev 2017; 33: 55-63.
[http://dx.doi.org/10.1016/j.cytogfr.2016.10.001] [PMID: 27743775]
[78]
Lavanya DK, Shivanna MB, Ganeshan S. Total phenolic content and antioxidant activity of spilanthes species from Peninsular India. Int J Pharm Pharmacol 2017; 1: 115.
[79]
Ojha RP, Rastogi M, Devi BP, Agrawal A, Dubey GP. Neuroprotective effect of curcuminoids against inflammation-mediated dopaminergic neurodegeneration in the MPTP model of Parkinson’s disease. J Neuroimmune Pharmacol 2012; 7(3): 609-18.
[http://dx.doi.org/10.1007/s11481-012-9363-2] [PMID: 22527634]
[80]
Kalaycıoğlu Z, Gazioğlu I, Erim FB. Comparison of antioxidant, anticholinesterase, and antidiabetic activities of three curcuminoids isolated from Curcuma longa L. Nat Prod Res 2017; 31(24): 2914-7.
[http://dx.doi.org/10.1080/14786419.2017.1299727] [PMID: 28287280]
[81]
Kuboyama T, Tohda C, Komatsu K. Effects of Ashwagandha (roots of Withania somnifera) on neurodegenerative diseases. Biol Pharm Bull 2014; 37(6): 892-7.
[http://dx.doi.org/10.1248/bpb.b14-00022] [PMID: 24882401]
[82]
Gupta M, Kaur G. Withania somnifera as a potential anxiolytic and anti-inflammatory candidate against systemic lipopolysaccharide-induced neuroinflammation. Neuromolecular Med 2018; 20(3): 343-62.
[http://dx.doi.org/10.1007/s12017-018-8497-7] [PMID: 29846872]
[83]
Srivastav S, Fatima M, Mondal AC. Important medicinal herbs in Parkinson’s disease pharmacotherapy. Biomed Pharmacother 2017; 92: 856-63.
[http://dx.doi.org/10.1016/j.biopha.2017.05.137] [PMID: 28599249]
[84]
Pandey A, Bani S, Dutt P, Kumar Satti N, Avtar Suri K, Nabi Qazi G. Multifunctional neuroprotective effect of Withanone, a compound from Withania somnifera roots in alleviating cognitive dysfunction. Cytokine 2018; 102: 211-21.
[http://dx.doi.org/10.1016/j.cyto.2017.10.019] [PMID: 29108796]
[85]
Grover A, Shandilya A, Agrawal V, Bisaria VS, Sundar D. Computational evidence to inhibition of human acetyl cholinesterase by withanolide a for Alzheimer treatment. J Biomol Struct Dyn 2012; 29(4): 651-62.
[http://dx.doi.org/10.1080/07391102.2012.10507408] [PMID: 22208270]
[86]
Prakash J, Yadav SK, Chouhan S, Singh SP. Neuroprotective role of Withania somnifera root extract in maneb-paraquat induced mouse model of parkinsonism. Neurochem Res 2013; 38(5): 972-80.
[http://dx.doi.org/10.1007/s11064-013-1005-4] [PMID: 23430469]
[87]
Sehgal N, Gupta A, Valli RK, et al. Withania somnifera reverses Alzheimer’s disease pathology by enhancing low-density lipoprotein receptor-related protein in liver. Proc Natl Acad Sci USA 2012; 109(9): 3510-5.
[http://dx.doi.org/10.1073/pnas.1112209109] [PMID: 22308347]
[88]
Dubey T, Chinnathambi S. Brahmi (Bacopa monnieri): An ayurvedic herb against the Alzheimer’s disease. Arch Biochem Biophys 2019; 676: 108153.
[http://dx.doi.org/10.1016/j.abb.2019.108153] [PMID: 31622587]
[89]
Shinomol GK. Muralidhara, Bharath MM. Exploring the role of “Brahmi” (Bacopa monnieri and Centella asiatica) in brain function and therapy. Recent Pat Endocr Metab Immune Drug Discov 2011; 5(1): 33-49.
[http://dx.doi.org/10.2174/187221411794351833] [PMID: 22074576]
[90]
Apetz N, Munch G, Govindaraghavan S, Gyengesi E. Natural compounds and plant extracts as therapeutics against chronic inflammation in Alzheimer’s disease-a translational perspective. CNS Neurol Disord Drug Targets 2014; 13(7): 1175-91.
[http://dx.doi.org/10.2174/1871527313666140917110635] [PMID: 25230232]
[91]
Braithwaite MC, Tyagi C, Tomar LK, Kumar P, Choonara YE, Pillay V. Nutraceutical-based therapeutics and formulation strategies augmenting their efficiency to complement modern medicine: An overview. J Funct Foods 2014; 6: 82-99.
[http://dx.doi.org/10.1016/j.jff.2013.09.022]
[92]
Marwah H, Garg T, Goyal AK, Rath G. Permeation enhancer strategies in transdermal drug delivery. Drug Deliv 2016; 23(2): 564-78.
[http://dx.doi.org/10.3109/10717544.2014.935532] [PMID: 25006687]
[93]
Javed MN, Kohli K, Amin S. Risk assessment integrated QbD approach for development of optimized bicontinuous mucoadhesive limicubes for oral delivery of rosuvastatin. AAPS PharmSciTech 2018; 19(3): 1377-91.
[http://dx.doi.org/10.1208/s12249-018-0951-1] [PMID: 29388027]
[94]
Javed MN, Alam MS, Waziri A, et al. QbD applications for the development of nanopharmaceutical products. Pharmaceutical Quality by Design. Cambridge: Academic Press 2019; pp. 229-53.
[95]
Meinke MC, Darvin ME, Vollert H, Lademann J. Bioavailability of natural carotenoids in human skin compared to blood. Eur J Pharm Biopharm 2010; 76(2): 269-74.
[http://dx.doi.org/10.1016/j.ejpb.2010.06.004] [PMID: 20558286]
[96]
Alkilani AZ, McCrudden MT, Donnelly RF. Transdermal drug delivery: Innovative pharmaceutical developments based on disruption of the barrier properties of the stratum corneum. Pharmaceutics 2015; 7(4): 438-70.
[http://dx.doi.org/10.3390/pharmaceutics7040438] [PMID: 26506371]
[97]
Murphy M, Carmichael AJ. Transdermal drug delivery systems and skin sensitivity reactions. Incidence and management. Am J Clin Dermatol 2000; 1(6): 361-8.
[http://dx.doi.org/10.2165/00128071-200001060-00004] [PMID: 11702612]
[98]
Mohanta BC, Javed MN, Hasnain MS, Nayak AK. Polyelectrolyte complexes of alginate for controlling drug release.Alginates in Drug Delivery. Cambridge: Academic Press 2020; pp. 297-321.
[99]
Imani M, Lahooti-Fard F, Taghizadeh SM, Takrousta M. Effect of adhesive layer thickness and drug loading on estradiol crystallization in a transdermal drug delivery system. AAPS PharmSciTech 2010; 11(3): 1268-75.
[http://dx.doi.org/10.1208/s12249-010-9494-9] [PMID: 20706877]
[100]
Dhiman S, Singh TG, Rehni AK. Transdermal patches: A recent approch to new drug delivery system. Int J Pharm Pharm Sci 2011.
[PMID: 14386374]
[101]
Rani S, Saroha K, Syan N, Mathur P. Transdermal patches a successful tool in transdermal drug delivery system: An overview. Pharm Sin 2011; 2(5): 17-29.
[102]
Mutalik S, Udupa N. Pharmacological evaluation of membrane- moderated transdermal system of glipizide. Clin Exp Pharmacol Physiol 2006; 33(1-2): 17-26.
[http://dx.doi.org/10.1111/j.1440-1681.2006.04318.x] [PMID: 16445694]
[103]
Stevenson CL, Santini JT Jr, Langer R. Reservoir-based drug delivery systems utilizing microtechnology. Adv Drug Deliv Rev 2012; 64(14): 1590-602.
[http://dx.doi.org/10.1016/j.addr.2012.02.005] [PMID: 22465783]
[104]
Hughes PJ, Freeman MK, Wensel TM. Appropriate use of transdermal drug delivery systems. J Nurs Educ Pract 2013; 3(10): 129-38.
[http://dx.doi.org/10.5430/jnep.v3n10p129]
[105]
Cherukuri S, Batchu UR, Mandava K, Cherukuri V, Ganapuram KR. Formulation and evaluation of transdermal drug delivery of topiramate. Int J Pharm Investig 2017; 7(1): 10-7.
[http://dx.doi.org/10.4103/jphi.JPHI_35_16] [PMID: 28405574]
[106]
Abd E, Yousef SA, Pastore MN, et al. Skin models for the testing of transdermal drugs. Clin Pharmacol Adv Appl 2016; 8: 163-176.
[http://dx.doi.org/10.2147/CPAA.S64788]
[107]
Trommer H, Neubert RH. Overcoming the stratum corneum: The modulation of skin penetration. A review. Skin Pharmacol Physiol 2006; 19(2): 106-21.
[http://dx.doi.org/10.1159/000091978] [PMID: 16685150]
[108]
Zimmer Ł, Kasperek R. Modern polymers in matrix tablets technology. Polim Med 2014; 44(3): 189-96.
[109]
Al Hanbali OA, Khan HMS, Sarfraz M, Arafat M, Ijaz S, Hameed A. Transdermal patches: Design and current approaches to painless drug delivery. Acta Pharm 2019; 69(2): 197-215.
[http://dx.doi.org/10.2478/acph-2019-0016] [PMID: 31259729]
[110]
Bidve SC, Kadam VB, Malpathak NP. Metabolite profiling and principle component analysis of a mangrove plant Aegiceras corniculatum L (Blanco). Int J Pharm Pharmacol 2018; 2: 125.
[http://dx.doi.org/10.31531/2581-3080.1000125]
[111]
Radhika B, Samreen N, Ramya N, et al. Pharmacognostic and phytochemical evaluation of Hibiscus hirtus Linn. Int J Biomed Investig 2019; 2: 117.
[112]
Padamwar MN, Pokharkar VB. Development of vitamin loaded topical liposomal formulation using factorial design approach: Drug deposition and stability. Int J Pharm 2006; 320(1-2): 37-44.
[http://dx.doi.org/10.1016/j.ijpharm.2006.04.001] [PMID: 16707237]
[113]
Montanari J, Vera M, Mensi E, Morilla M, Romero E. Nanoberries for topical delivery of antioxidants. J Cosmet Sci 2013; 64(6): 469-81.
[PMID: 24397884]
[114]
Lee WC, Tsai TH. Preparation and characterization of liposomal coenzyme Q10 for in vivo topical application. Int J Pharm 2010; 395(1-2): 78-83.
[http://dx.doi.org/10.1016/j.ijpharm.2010.05.006] [PMID: 20635514]
[115]
Parham S, Kharazi AZ, Bakhsheshi-Rad HR, et al. Electrospun nano-fibers for biomedical and tissue engineering applications: A comprehensive review. Materials (Basel) 2020; 13(9): 2153.
[http://dx.doi.org/10.3390/ma13092153] [PMID: 32384813]
[116]
Xue J, Wu T, Dai Y, Xia Y. Electrospinning and electrospun nanofibers: Methods, materials, and applications. Chem Rev 2019; 119(8): 5298-415.
[http://dx.doi.org/10.1021/acs.chemrev.8b00593] [PMID: 30916938]
[117]
Taepaiboon P, Rungsardthong U, Supaphol P. Vitamin-loaded electrospun cellulose acetate nanofiber mats as transdermal and dermal therapeutic agents of vitamin A acid and vitamin E. Eur J Pharm Biopharm 2007; 67(2): 387-97.
[http://dx.doi.org/10.1016/j.ejpb.2007.03.018] [PMID: 17498935]
[118]
Suwantong O, Opanasopit P, Ruktanonchai U, Supaphol P. Electrospun cellulose acetate fiber mats containing curcumin and release characteristic of the herbal substance. Polymer (Guildf) 2007; 48(26): 7546-57.
[http://dx.doi.org/10.1016/j.polymer.2007.11.019]
[119]
Suwantong O, Ruktanonchai U, Supaphol P. Electrospun cellulose acetate fiber mats containing asiaticoside or Centella asiatica crude extract and the release characteristics of asiaticoside. Polymer (Guildf) 2008; 49(19): 4239-47.
[http://dx.doi.org/10.1016/j.polymer.2008.07.020]
[120]
Mohanty D, Rani MJ, Haque MA, et al. Preparation and evaluation of transdermal naproxen niosomes: Formulation optimization to preclinical anti-inflammatory assessment on murine model. J Liposome Res 2020; 30(4): 377-387.
[PMID: 31412744]
[121]
Paneva D, Manolova N, Argirova M, Rashkov I. Antibacterial electrospun poly(ɛ-caprolactone)/ascorbyl palmitate nanofibrous materials. Int J Pharm 2011; 416(1): 346-55.
[http://dx.doi.org/10.1016/j.ijpharm.2011.06.032] [PMID: 21726615]
[122]
Zhang D, Lv P, Zhou C, Zhao Y, Liao X, Yang B. Cyclodextrin-based delivery systems for cancer treatment. Mater Sci Eng C 2019; 96: 872-86.
[http://dx.doi.org/10.1016/j.msec.2018.11.031] [PMID: 30606602]
[123]
Gonnet M, Lethuaut L, Boury F. New trends in encapsulation of liposoluble vitamins. J Control Release 2010; 146(3): 276-90.
[http://dx.doi.org/10.1016/j.jconrel.2010.01.037] [PMID: 20600399]
[124]
Yuan C, Jin Z, Xu X. Inclusion complex of astaxanthin with hydroxypropyl-β-cyclodextrin: UV, FTIR, 1H NMR and molecular modeling studies. Carbohydr Polym 2012; 89(2): 492-6.
[http://dx.doi.org/10.1016/j.carbpol.2012.03.033] [PMID: 24750749]
[125]
Kim SH, Youn JY, Kim KM, Kang KC, Pyo HB, Lee SJ. Characterization of an inclusion complex of, 7-dehydrocholesterol and cyclodextrin. J Ind Eng Chem 2010; 16(1): 119-21.
[http://dx.doi.org/10.1016/j.jiec.2010.01.008]
[126]
Tomren MA, Másson M, Loftsson T, Tønnesen HH. Studies on curcumin and curcuminoids XXXI. Symmetric and asymmetric curcuminoids: Stability, activity and complexation with cyclodextrin. Int J Pharm 2007; 338(1-2): 27-34.
[http://dx.doi.org/10.1016/j.ijpharm.2007.01.013] [PMID: 17298869]
[127]
Patravale VB, Mandawgade SD. Novel cosmetic delivery systems: An application update. Int J Cosmet Sci 2008; 30(1): 19-33.
[http://dx.doi.org/10.1111/j.1468-2494.2008.00416.x] [PMID: 18377627]
[128]
Ansari KA, Vavia PR, Trotta F, Cavalli R. Cyclodextrin-based nanosponges for delivery of resveratrol: in vitro characterisation, stability, cytotoxicity and permeation study. AAPS PharmSciTech 2011; 12(1): 279-86.
[http://dx.doi.org/10.1208/s12249-011-9584-3] [PMID: 21240574]
[129]
Lobo S, Sachdeva S, Goswami T. Role of pressure-sensitive adhesives in transdermal drug delivery systems. Ther Deliv 2016; 7(1): 33-48.
[http://dx.doi.org/10.4155/tde.15.87] [PMID: 26652621]
[130]
Jung SY, Kang EY, Choi YJ, Chun IK, Lee BK, Gwak HS. Formulation and evaluation of ubidecarenone transdermal delivery systems. Drug Dev Ind Pharm 2009; 35(9): 1029-34.
[http://dx.doi.org/10.1080/03639040902755205] [PMID: 19365779]
[131]
Ita K. Transdermal delivery of drugs with microneedles-potential and challenges. Pharmaceutics 2015; 7(3): 90-105.
[http://dx.doi.org/10.3390/pharmaceutics7030090] [PMID: 26131647]
[132]
Yan Q, Wang W, Weng J, et al. Dissolving microneedles for transdermal delivery of huperzine A for the treatment of Alzheimer’s disease. Drug Deliv 2020; 27(1): 1147-55.
[http://dx.doi.org/10.1080/10717544.2020.1797240] [PMID: 32729341]
[133]
Jahangir MA, Taleuzzaman M, Beg S, Verma S, Gilani SJ, Alam P. A review of Eugenol-based nanomedicine: Recent advancements. Curr Bioact Compd 2020; 16: 1.
[http://dx.doi.org/10.2174/1573407216999200525145633]
[134]
Li Y, Zheng J, Xiao H, McClements DJ. Nanoemulsion-based delivery systems for poorly water-soluble bioactive compounds: Influence of formulation parameters on Polymethoxyflavone crystallization. Food Hydrocoll 2012; 27(2): 517-28.
[http://dx.doi.org/10.1016/j.foodhyd.2011.08.017] [PMID: 22685367]
[135]
Mitri K, Shegokar R, Gohla S, Anselmi C, Müller RH. Lutein nanocrystals as antioxidant formulation for oral and dermal delivery. Int J Pharm 2011; 420(1): 141-6.
[http://dx.doi.org/10.1016/j.ijpharm.2011.08.026] [PMID: 21884768]
[136]
Kong M, Chen XG, Kweon DK, Park HJ. Investigations on skin permeation of hyaluronic acid based nanoemulsion as transdermal carrier. Carbohydr Polym 2011; 89(2): 837-43.
[http://dx.doi.org/10.1016/j.carbpol.2011.05.027]
[137]
Teeranachaideekul V, Souto EB, Junyaprasert VB, Müller RH. Cetyl palmitate-based NLC for topical delivery of Coenzyme Q(10) - development, physicochemical characterization and in vitro release studies. Eur J Pharm Biopharm 2007; 67(1): 141-8.
[http://dx.doi.org/10.1016/j.ejpb.2007.01.015] [PMID: 17346953]
[138]
Hasnain MS, Javed MN, Alam MS, et al. Purple heart plant leaves extract-mediated silver nanoparticle synthesis: Optimization by Box-Behnken design. Mater Sci Eng C 2019; 99: 1105-14.
[http://dx.doi.org/10.1016/j.msec.2019.02.061] [PMID: 30889643]
[139]
Alam MS, Garg A, Pottoo FH, et al. Gum ghatti mediated, one pot green synthesis of optimized gold nanoparticles: Investigation of process-variables impact using Box-Behnken based statistical design. Int J Biol Macromol 2017; 104(Pt A): 758-67.
[http://dx.doi.org/10.1016/j.ijbiomac.2017.05.129] [PMID: 28601649]
[140]
Alam MS, Javed MN, Pottoo FH, et al. QbD approached comparison of reaction mechanism in microwave synthesized gold nanoparticles and their superior catalytic role against hazardous nirto-dye. Appl Organomet Chem 2019; 33(9): e5071.
[http://dx.doi.org/10.1002/aoc.5071]
[141]
Haghighi P, Ghaffari S, Bidgoli SA, Qomi M, Haghighat S. Preparation, characterization and evaluation of Ginkgo biloba solid lipid nanoparticles. Nanomed Res J 2018; 3: 71-8.
[http://dx.doi.org/10.22034/nmrj.2018.02.003]
[142]
Kandimalla KK, Babu RJ, Singh M. Biphasic flux profiles of melatonin: the Yin-Yang of transdermal permeation enhancement mediated by fatty alcohol enhancers. J Pharm Sci 2010; 99(1): 209-18.
[http://dx.doi.org/10.1002/jps.21812] [PMID: 19530074]
[143]
Teskac K, Kristl J. The evidence for solid lipid nanoparticles mediated cell uptake of resveratrol. Int J Pharm 2010; 390(1): 61-9.
[http://dx.doi.org/10.1016/j.ijpharm.2009.10.011] [PMID: 19833178]
[144]
Khayata N, Abdelwahed W, Chehna MF, Charcosset C, Fessi H. Stability study and lyophilization of vitamin E-loaded nanocapsules prepared by membrane contactor. Int J Pharm 2012; 439(1-2): 254-9.
[http://dx.doi.org/10.1016/j.ijpharm.2012.09.032] [PMID: 23010283]
[145]
Esposito E, Nastruzzi C, Sguizzato M, Cortesi R. Nanomedicines to treat skin pathologies with natural molecules. Curr Pharm Des 2019; 25(21): 2323-37.
[http://dx.doi.org/10.2174/1381612825666190709210703] [PMID: 31584367]
[146]
Chinembiri TN, Gerber M, du Plessis LH, du Preez JL, Hamman JH, du Plessis J. Topical delivery of Withania somnifera crude extracts in niosomes and solid lipid nanoparticles. Pharmacogn Mag 2017; 13(Suppl. 3): S663-71.
[http://dx.doi.org/10.4103/pm.pm_489_16] [PMID: 29142430]
[147]
Fukushima K, Ise A, Morita H, et al. Two-layered dissolving microneedles for percutaneous delivery of peptide/protein drugs in rats. Pharm Res 2011; 28(1): 7-21.
[http://dx.doi.org/10.1007/s11095-010-0097-7] [PMID: 20300802]
[148]
Pathan IB, Setty CM. Chemical penetration enhancers for transdermal drug delivery systems. Trop J Pharm Res 2009; 8: 2.
[http://dx.doi.org/10.4314/tjpr.v8i2.44527]
[149]
Wohlrab J, Kreft B, Tamke B. Skin tolerability of transdermal patches. Expert Opin Drug Deliv 2011; 8(7): 939-48.
[http://dx.doi.org/10.1517/17425247.2011.574689] [PMID: 21506903]
[150]
Gupta H, Babu RJ. Transdermal delivery: Product and patent update. DDF 2013; 7(3): 184-205.
[http://dx.doi.org/10.2174/187221130703131128121747] [PMID: 24025130]
[151]
Akhtar N, Singh V, Yusuf M, Khan RA. Non-invasive drug delivery technology: Development and current status of transdermal drug delivery devices, techniques and biomedical applications. Biomed Tech (Berl) 2020; 65(3): 243-72.
[http://dx.doi.org/10.1515/bmt-2019-0019] [PMID: 31926064]
[152]
Maher S, Brayden DJ, Casettari L, Illum L. Application of permeation enhancers in oral delivery of macromolecules: An update. Pharmaceutics 2019; 11(1): 41.
[http://dx.doi.org/10.3390/pharmaceutics11010041] [PMID: 30669434]
[153]
Pandey M, Saleem S, Nautiyal H, Pottoo FH, Javed MN. PINK1/Parkin in Neurodegenerative Disorders: Crosstalk Between Mitochondrial Stress and Neurodegeneration.Uddin MS, Ashraf GM. Quality Control of Cellular Protein in Neurodegenerative Disorders. Pennsylvania: IGI Global 2020; pp. 282-301.
[154]
Pottoo FH, Javed MN, Barkat MA, et al. Estrogen and serotonin: Complexity of interactions and implications for epileptic seizures and epileptogenesis. Curr Neuropharmacol 2019; 17(3): 214-31.
[http://dx.doi.org/10.2174/1570159X16666180628164432] [PMID: 29956631]
[155]
Pottoo FH, Tabassum N, Javed MN, et al. Raloxifene potentiates the effect of fluoxetine against maximal electroshock induced seizures in mice. Eur J Pharm Sci 2020; 146: 105261.
[http://dx.doi.org/10.1016/j.ejps.2020.105261] [PMID: 32061655]
[156]
Pottoo FH, Tabassum N, Javed MN, et al. The synergistic effect of raloxifene, fluoxetine, and bromocriptine protects against pilocarpine-induced status epilepticus and temporal lobe epilepsy. Mol Neurobiol 2019; 56(2): 1233-47.
[http://dx.doi.org/10.1007/s12035-018-1121-x] [PMID: 29881945]
[157]
Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol 2008; 26(11): 1261-8.
[http://dx.doi.org/10.1038/nbt.1504] [PMID: 18997767]
[158]
Nigar S, Pottoo FH, Tabassum N, Verma SK, Javed MN. Molecular insights into the role of inflammation and oxidative stress in epilepsy. JAMPS 2016; 10(1): 1-9.
[http://dx.doi.org/10.9734/JAMPS/2016/24441]
[159]
Nasri H, Baradaran A, Shirzad H, Rafieian-Kopaei M. New concepts in nutraceuticals as alternative for pharmaceuticals. Int J Prev Med 2014; 5(12): 1487-99.
[PMID: 25709784]
[160]
Yamagishi N, Namioka T, Okura N, et al. Application of a reservoir-type calcitriol transdermal patch in dairy cattle. J Vet Med Sci 2009; 71(6): 845-8.
[http://dx.doi.org/10.1292/jvms.71.845] [PMID: 19578302]
[161]
D’Angelo Costa GM, Sales de Oliveira Pinto CA, Rodrigues Leite-Silva V, Rolim Baby A, Robles Velasco MV. Is vitamin D3 transdermal formulation feasible? An ex vivo skin retention and permeation. AAPS PharmSciTech 2018; 19(5): 2418-25.
[http://dx.doi.org/10.1208/s12249-018-1065-5] [PMID: 29869312]
[162]
Alsaqr A, Rasoully M, Musteata FM. Investigating transdermal delivery of vitamin D3. AAPS PharmSciTech 2015; 16(4): 963-72.
[http://dx.doi.org/10.1208/s12249-015-0291-3] [PMID: 25609377]
[163]
Fetoni AR, Piacentini R, Fiorita A, Paludetti G, Troiani D. Water- soluble Coenzyme Q10 formulation (Q-ter) promotes outer hair cell survival in a guinea pig model of noise induced hearing loss (NIHL). Brain Res 2009; 1257: 108-16.
[http://dx.doi.org/10.1016/j.brainres.2008.12.027] [PMID: 19133240]
[164]
Cevc G, Vierl U. Nanotechnology and the transdermal route: A state of the art review and critical appraisal. J Control Release 2010; 141(3): 277-99.
[http://dx.doi.org/10.1016/j.jconrel.2009.10.016] [PMID: 19850095]
[165]
Thong HY, Zhai H, Maibach HI. Percutaneous penetration enhancers: An overview. Skin Pharmacol Physiol 2007; 20(6): 272-82.
[http://dx.doi.org/10.1159/000107575] [PMID: 17717423]
[166]
Park JH, Lee JW, Kim YC, Prausnitz MR. The effect of heat on skin permeability. Int J Pharm 2008; 359(1-2): 94-103.
[http://dx.doi.org/10.1016/j.ijpharm.2008.03.032] [PMID: 18455889]
[167]
DiPierro G, Giannos SA. Biosynchronous transdermal drug delivery for longevity, anti-aging, fatigue management, obesity, weight loss, weight management, delivery of nutraceuticals, and the treatment of hyperglycemia, Alzheimer's disease, sleep disorders, Parkinson's disease, aids, epilepsy, attention deficit disorder, nicotine addiction, cancer, headache and pain control, asthma, angina, hypertension, depression, cold, flu and the like. US10258778, 2019.
[168]
Fu L, Wang K, Qiang F, et al. Application of ginsenoside RG3 in preparing medicine for preventing and/or treating dementia, and medicine for treating dementia. US10238676, 2019.
[169]
Jonathan E. Transdermal delivery of large agents. US20190343773, 2019.
[170]
Gokaraju GR, Gokaraju RR, Gokaraju VKRR, et al. Synergistic anti-inflammatory compositions comprising Boswellia serrata extract. US10206964, 2019.
[171]
Kariman A. Compound and method for reducing neuropathic pain and depression. US20190255036, 2019.
[172]
Sang JK, Lee K, Seong HK. Peptide having effects of increasing telomerase activity and extending telomere, and composition containing same. US20190142894, 2019.
[173]
Garti N, Levi SG. Dilutable formulations of cannabinoids and processes for their preparation. US20190314326, 2019.
[174]
Callizot N. Combination composition comprising huperzine. US10596166, 2020.
[175]
Vales RL, Quijorna IF. Maresins for use in the treatment of CNS injuries. US20200163923, 2020.
[176]
Nagano K, Harada K, Higashisaka K, et al. Curcumin-containing medicinal preparation. US20200009211, 2020.

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