Open Access Open Access  Restricted Access Subscription Access
Open Access Open Access Open Access  Restricted Access Restricted Access Subscription Access

Repurposing Drugs for Management of Alzheimer Disease


Affiliations
1 Department of Pharmaceutics, JSS College of Pharmacy, Ooty, Tamilnadu-643001, India
2 Affiliated by JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
     

   Subscribe/Renew Journal


Alzheimer's disease (AD), or Alzheimer’s, It is neurodegenerative disease starts very slowly and with time it got worse one of the biggest challenge of the CNS drug delivery scientist. Mental and physic exercise, Cholinesterase Inhibitors, and avoiding adipositas may reduce the risk of the AD. Currently, existing/approved drugs for the treatment of the AD are based on neurotransmitter or enzyme replacement/modulation. Existing therapies gives just an optimum advantage and consequently, in present, there is an evident desire for advanced medications or therapies. Drug repurposing is one of the latest trend and the alternative approach in drug advancement, with a history of successful reuse of currently available drugs. A signify category of the drugs reuse in the management of the AD among them the prominent category is the anticancer, antiepileptic, antibiotics, ant diabetic etc. The review focuses on mechanism and application for the repurposing of the existing drugs in the light of the clinical and pre-clinical evidences.

Keywords

Alzheimer’s Disease, Repurposing Drugs, Repurposed Drugs Pathways, Preclinical Data, Clinical Evidences.
Subscription Login to verify subscription
User
Notifications
Font Size


  • Holtzman DM, Morris JC, Goate AM. Alzheimer’s disease: the challenge of the second century. Science translational medicine.2011, 3(77), sr1-sr1.
  • Reisberg B, Ferris S, De Leon M. Senile dementia of the Alzheimer type: Diagnostic and differential diagnostic features with special reference to functional assessment staging. Senile dementia of the Alzheimer type: Springer; 1985, 18-37.
  • Hardy JA, Higgins GA. Alzheimer's disease: the amyloid cascade hypothesis. Science. 1992, 256(5054),184.
  • Duyc kaerts C, Potier M-C, Delatour B. Alzheimer disease models and human neuropathology: similarities and differences. Acta neuropathologica. 2008, 115(1), , 5-38.
  • Larson EB, Shadlen M-F, Wang L, McCormick WC, Bowen JD, Teri L, et al. Survival after initial diagnosis of Alzheimer disease. Annals of internal medicine. 2004, 140(7), 501-9.
  • Association As. 2017 Alzheimer's disease facts and figures. Alzheimer's & Dementia. 2017, 13(4), 325-73.
  • Fox PJ. Alzheimer's Disease: An Historical Overview. American Journal of Alzheimer's Care and Related Disorders. 1986, 1(4), 18-24.
  • Cipriani G, Danti S, Carlesi C. Three men in a (same) boat: Alzheimer, Pick, Lewy. Historical notes. European Geriatric Medicine. 2016, 7(6), 526-30.
  • Bertram L, Tanzi RE. Thirty years of Alzheimer's disease genetics: the implications of systematic meta-analyses. Nature Reviews Neuroscience. 2008, 9(10), 768.
  • Whitehouse PJ, George D. The myth of Alzheimer's: What you aren't being told about today's most dreaded diagnosis: Macmillan; 2008.
  • Janus C. Vaccines for Alzheimer’s disease. CNS drugs. 2003, 17(7), 457-74.
  • Finger EA. Alzheimer Demenz. 2015.
  • Helmuth L. New Alzheimer's treatments that may ease the mind. American Association for the Advancement of Science; 2002.
  • Hutton M, Pérez-Tur J, Hardy J. Genetics of Alzheimer’s. Essays in biochemistry. 1998, 33.
  • Selkoe DJ. Amyloid β-protein and the genetics of Alzheimer's disease. Journal of Biological Chemistry. 1996, 271(31), 18295-8.
  • St George-Hyslop PH. Molecular genetics of Alzheimer’s disease. Biological psychiatry. 2000, 47(3), 183-99.
  • Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, et al. β-Secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE. science. 1999, 286(5440), 735-41.
  • Scheuner D, Eckman C, Jensen M, Song X, Citron M, Suzuki N, et al. Secreted amyloid β–protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer's disease. Nature medicine. 1996, 2(8), 864.
  • Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J. Free radicals and antioxidants in normal physiological functions and human disease. The international journal of biochemistry & cell biology. 2007, 39(1),44-84.
  • Sayre LM, Perry G, Smith MA. Oxidative stress and neurotoxicity. Chemical research in toxicology. 2007, 21(1), 172-88.
  • Ittner LM, Götz J. Amyloid-β and tau—a toxic pas de deux in Alzheimer's disease. Nature Reviews Neuroscience. 2011, 12(2), 67.
  • Mandelkow E-M, Mandelkow E. Tau in Alzheimer's disease. Trends in cell biology. 1998, 8(11), 425-7.
  • Francis PT, Palmer AM, Snape M, Wilcock GK. The cholinergic hypothesis of Alzheimer’s disease: a review of progress. Journal of Neurology, Neurosurgery & Psychiatry. 1999, 66(2), 137-47.
  • Bartus RT, Dean Rr, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982, 217(4558), 408-14.
  • Cummings JL, Back C. The cholinergic hypothesis of neuropsychiatric symptoms in Alzheimer's disease. The American Journal of Geriatric Psychiatry. 1998, 6(2), S64-S78.
  • Kaduszkiewicz H, Zimmermann T, Beck-Bornholdt H-P, van den Bussche H. Cholinesterase inhibitors for patients with Alzheimer's disease: systematic review of randomised clinical trials. Bmj. 2005, 331(7512),321-7.
  • Hansen RA, Gartlehner G, Webb AP, Morgan LC, Moore CG, Jonas DE. Efficacy and safety of donepezil, galantamine, and rivastigmine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. Clinical interventions in aging. 2008, 3(2),211.
  • Jacob C, Koutsilieri E, Bartl J, Neuen-Jacob E, Arzberger T, Zander N, et al. Alterations in expression of glutamatergic transporters and receptors in sporadic Alzheimer's disease. Journal of Alzheimer's Disease. 2007, 11(1), 97-116.
  • Uddin MS, Al Mamun A, Kabir MT, Jakaria M, Mathew B, Barreto GE, et al. Nootropic and Anti-Alzheimer’s Actions of Medicinal Plants: Molecular Insight into Therapeutic Potential to Alleviate Alzheimer’s Neuropathology. Molecular neurobiology. 2018,1-20.
  • Giacobini E. Invited Review Cholinesterase inhibitors for Alzheimer’s disease therapy: from tacrine to future applications. Neurochemistry international. 1998, 32(5-6), 413-9.
  • Grutzendler J, Morris JC. Cholinesterase inhibitors for Alzheimer’s disease. Drugs. 2001, 61(1), 1-52.
  • Jones R. A review comparing the safety and tolerability of memantine with the acetylcholinesterase inhibitors. International Journal of Geriatric Psychiatry: A journal of the psychiatry of late life and allied sciences. 2010, 25(6), 547-53.
  • Kumar A, Singh A. A review on Alzheimer's disease pathophysiology and its management: an update. Pharmacological Reports. 2015, 67(2), 195-203.
  • Witt A, Macdonald N, Kirkpatrick P. Memantine hydrochloride. Nature Publishing Group; 2004.
  • De Felice FG, Velasco PT, Lambert MP, Viola K, Fernandez SJ, Ferreira ST, et al. Aβ oligomers induce neuronal oxidative stress through an N-methyl-D-aspartate receptor-dependent mechanism that is blocked by the Alzheimer drug memantine. Journal of Biological Chemistry. 2007, 282(15), 11590-601.
  • Kaltschmidt B, Uherek M, Volk B, Baeuerle PA, Kaltschmidt C. Transcription factor NF-κB is activated in primary neurons by amyloid β peptides and in neurons surrounding early plaques from patients with Alzheimer disease. Proceedings of the National Academy of Sciences. 1997, 94(6), 2642-7.
  • Park-Wyllie LY, Mamdani MM, Li P, Gill SS, Laupacis A, Juurlink DN. Cholinesterase inhibitors and hospitalization for bradycardia: a population-based study. PLoS medicine. 2009, 6(9), e1000157.
  • Pardridge WM. Blood–brain barrier delivery. Drug discovery today. 2007, 12(1-2), 54-61.
  • Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nature reviews Drug discovery. 2004, 3(8), 673.
  • Boolell M, Gepi‐Attee S, Gingell J, Allen M. Sildenafil, a novel effective oral therapy for male erectile dysfunction. British journal of urology. 1996, 78(2),257-61.
  • Appleby BS, Nacopoulos D, Milano N, Zhong K, Cummings JL. A review: treatment of Alzheimer’s disease discovered in repurposed agents. Dementia and geriatric cognitive disorders. 2013, 35(1-2), 1-22.
  • Vlassenko AG, Vaishnavi SN, Couture L, Sacco D, Shannon BJ, Mach RH, et al. Spatial correlation between brain aerobic glycolysis and amyloid-β (Aβ) deposition. Proceedings of the National Academy of Sciences. 2010, 107(41), 17763-7.
  • Monacelli F, Cea M, Borghi R, Odetti P, Nencioni A. Do cancer drugs counteract neurodegeneration? Repurposing for alzheimer’s disease. Journal of Alzheimer's Disease. 2017, 55(4),1295-306.
  • Roe CM, Behrens M, Xiong C, Miller J, Morris J. Alzheimer disease and cancer. Neurology. 2005, 64(5), 895-8.
  • Li G, Faibushevich A, Turunen BJ, Yoon SO, Georg G, Michaelis ML, et al. Stabilization of the cyclin‐dependent kinase 5 activator, p35, by paclitaxel decreases β-amyloid toxicity in cortical neurons. Journal of neurochemistry.2003, 84(2), 347-62.
  • Michaelis M, Ranciat N, Chen Y, Bechtel M, Ragan R, Hepperle M, et al. Protection against β‐amyloid toxicity in primary neurons by paclitaxel (taxol). Journal of neurochemistry. 1998, 70(4), 1623-7.
  • O'hare E, Jeggo R, Kim E-M, Barbour B, Walczak J-S, Palmer P, et al. Lack of support for bexarotene as a treatment for Alzheimer's disease. Neuropharmacology. 2016, 100, 124-30.
  • Bomben V, Holth J, Reed J, Cramer P, Landreth G, Noebels J. Bexarotene reduces network excitability in models of Alzheimer's disease and epilepsy. Neurobiology of aging. 2014, 35(9), 2091-5.
  • Cramer PE, Cirrito JR, Wesson DW, Lee CD, Karlo JC, Zinn AE, et al. ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficits in AD mouse models. science. 2012, 335(6075), 1503-6.
  • Bachmeier C, Beaulieu-Abdelahad D, Crawford F, Mullan M, Paris D. Stimulation of the retinoid X receptor facilitates beta-amyloid clearance across the blood–brain barrier. Journal of Molecular Neuroscience. 2013, 49(2), 270-6.
  • Frischer H, Ahmad T. Severe generalized glutathione reductase deficiency after antitumor chemotherapy with BCNU [1, 3-bis (chloroethyl)-1-nitrosourea]. The Journal of laboratory and clinical medicine. 1977, 89(5), 1080-91.
  • Hayes CD, Dey D, Palavicini JP, Wang H, Patkar KA, Minond D, et al. Striking reduction of amyloid plaque burden in an Alzheimer's mouse model after chronic administration of carmustine. BMC medicine. 2013, 11(1), 81.
  • Araki W. Potential repurposing of oncology drugs for the treatment of Alzheimer's disease. BMC medicine. 2013, 11(1), 82.
  • Jin N. Opposite effects of lithium and valproic acid on trophic deprivation-induced GSK-3 (Beta) activation, c-Jun expression and neuronal cell death. 2004.
  • Qing H, He G, Ly PT, Fox CJ, Staufenbiel M, Cai F, et al. Valproic acid inhibits Aβ production, neuritic plaque formation, and behavioral deficits in Alzheimer's disease mouse models. Journal of Experimental Medicine. 2008, 205(12), 2781-9.
  • Ly PT, Wu Y, Zou H, Wang R, Zhou W, Kinoshita A, et al. Inhibition of GSK3β-mediated BACE1 expression reduces Alzheimer-associated phenotypes. The Journal of clinical investigation.2012, 123(1).
  • Hooper C, Killick R, Lovestone S. The GSK3 hypothesis of Alzheimer’s disease. Journal of neurochemistry. 2008, 104(6), 1433-9.
  • Leng Y, Chuang D-M. Endogenous α-synuclein is induced by valproic acid through histone deacetylase inhibition and participates in neuroprotection against glutamate-induced excitotoxicity. Journal of Neuroscience. 2006, 26(28), 7502-12.
  • Yao Z-G, Jing H-Y, Wang D-M, Lv B-B, Li J-M, Liu F-F, et al. Valproic acid ameliorates olfactory dysfunction in APP/PS1 transgenic mice of Alzheimer's disease: ameliorations from the olfactory epithelium to the olfactory bulb. Pharmacology Biochemistry and Behavior. 2016, 144, 53-9.
  • Kim H-S, Suh Y-H. Minocycline and neurodegenerative diseases. Behavioural brain research. 2009, 196(2), 168-79.
  • Hashimoto K. Microglial activation in schizophrenia and minocycline treatment. Progress in Neuropsychopharmacology & Biological Psychiatry. 2008, 7(32),1758-9.
  • Mao J. Glutamate transporter: an unexpected target for some antibiotics. Molecular pain. 2005, 1(1), 5.
  • Rothstein JD, Patel S, Regan MR, Haenggeli C, Huang YH, Bergles DE, et al. β-Lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature. 2005, 433(7021),73.
  • Tomiyama T, Shoji A, Kataoka K-i, Suwa Y, Asano S, Kaneko H, et al. Inhibition of amyloid protein aggregation and neurotoxicity by rifampicin its possible function as a hydroxyl radical scavenger. Journal of Biological Chemistry. 1996, 271(12), 6839-44.
  • Loeb MB, Molloy DW, Smieja M, Standish T, Goldsmith CH, Mahony J, et al. A randomized, controlled trial of doxycycline and rifampin for patients with Alzheimer's disease. Journal of the American Geriatrics Society. 2004, 52(3), 52(3), 381-7.
  • He G, Luo W, Li P, Remmers C, Netzer WJ, Hendrick J, et al. Gamma-secretase activating protein is a therapeutic target for Alzheimer’s disease. Nature. 2010, 467(7311), 95.
  • Jankowsky JL, Slunt HH, Ratovitski T, Jenkins NA, Copeland NG, Borchelt DR. Co-expression of multiple transgenes in mouse CNS: a comparison of strategies. Biomolecular engineering. 2001, 17(6), 157-65.
  • Balducci C, Mehdawy B, Mare L, Giuliani A, Lorenzini L, Sivilia S, et al. The γ-secretase modulator CHF5074 restores memory and hippocampal synaptic plasticity in plaque-free Tg2576 mice. Journal of Alzheimer's Disease. 2011, 24(4), 799-816.
  • Qosa H, Abuznait AH, Hill RA, Kaddoumi A. Enhanced brain amyloid-β clearance by rifampicin and caffeine as a possible protective mechanism against Alzheimer's disease. Journal of Alzheimer's Disease. 2012, 31(1), 151-65.
  • Yulug B, Hanoglu L, Kilic E, Schabitz WR. RIFAMPICIN: an antibiotic with brain protective function. Brain research bulletin. 2014, 107, 37-42.
  • Umeda T, Ono K, Sakai A, Yamashita M, Mizuguchi M, Klein WL, et al. Rifampicin is a candidate preventive medicine against amyloid-β and tau oligomers. Brain. 2016, 139(5),1568-86.
  • Abuznait AH, Cain C, Ingram D, Burk D, Kaddoumi A. Up‐regulation of P‐glycoprotein reduces intracellular accumulation of beta amyloid: investigation of P‐glycoprotein as a novel therapeutic target for Alzheimer's disease. Journal of Pharmacy and Pharmacology. 2011, 63(8), 1111-8.
  • Jenagaratnam L, McShane R. Clioquinol for the treatment of Alzheimer's Disease. Cochrane Database of Systematic Reviews. 2006.
  • Froestl W, Pfeifer A, Muhs A. Cognitive enhancers (Nootropics). Part 3: drugs interacting with targets other than receptors or enzymes. Disease-modifying drugs. Update 2014. Journal of Alzheimer's Disease. 2014, 42(4), 1079-149.
  • Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F. New pharmacological strategies for treatment of Alzheimer's disease: focus on disease modifying drugs. British journal of clinical pharmacology. 2012, 73(4), 504-17.
  • Csiszar A, Tucsek Z, Toth P, Sosnowska D, Gautam T, Koller A, et al. Synergistic effects of hypertension and aging on cognitive function and hippocampal expression of genes involved in β-amyloid generation and Alzheimer's disease. American Journal of Physiology-Heart and Circulatory Physiology. 2013, 305(8), H1120-H30.
  • Paris D, Bachmeier C, Patel N, Quadros A, Volmar C-H, Laporte V, et al. Selective antihypertensive dihydropyridines lower Aβ accumulation by targeting both the production and the clearance of Aβ across the blood-brain barrier. Molecular Medicine.2011, 17(3-4), 149.
  • Cho H, Ueda M, Shima K, Mizuno A, Hayashimatsu M, Ohnaka Y, et al. Dihydropyrimidines: novel calcium antagonists with potent and long-lasting vasodilative and anti-hypertensive activity. Journal of medicinal chemistry. 1989, 32(10), 2399-406.
  • HOWLETT DR, GEORGE AR, MARKWELL RE. Common structural features determine the effectiveness of carvedilol, daunomycin and rolitetracycline as inhibitors of Alzheimer β-amyloid fibril formation. Biochemical Journal. 1999, 343(2), 419-23.
  • Wang J, Ono K, Dickstein DL, Arrieta-Cruz I, Zhao W, Qian X, et al. Carvedilol as a potential novel agent for the treatment of Alzheimer's disease. Neurobiology of aging. 2011, 32(12), 2321. e1-. e12.
  • Arrieta-Cruz I, Wang J, Pavlides C, Pasinetti GM. Carvedilol reestablishes long-term potentiation in a mouse model of Alzheimer's disease. Journal of Alzheimer's Disease. 2010, 21(2), 649-54.
  • Daulatzai MA. Quintessential risk factors: their role in promoting cognitive dysfunction and Alzheimer’s disease. Neurochemical research. 2012, 37(12), 2627-58.
  • Wolk DA, Dickerson BC, Initiative AsDN. Apolipoprotein E (APOE) genotype has dissociable effects on memory and attentional–executive network function in Alzheimer's disease. Proceedings of the National Academy of Sciences. 2010:201001412.
  • Dhikav V, Anand K. Potential predictors of hippocampal atrophy in Alzheimer’s disease. Drugs & aging. 2011, 28(1), 1-11.
  • Watson GS, Cholerton BA, Reger MA, Baker LD, Plymate SR, Asthana S, et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. The American journal of geriatric psychiatry. 2005, 13(11), 950-8.
  • Pedersen WA, McMillan PJ, Kulstad JJ, Leverenz JB, Craft S, Haynatzki GR. Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Experimental neurology. 2006, 199(2), 265-73.
  • Mandrekar-Colucci S, Karlo JC, Landreth GE. Mechanisms underlying the rapid peroxisome proliferator-activated receptor-γ-mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimer's disease. Journal of Neuroscience. 2012, 32(30), 10117-28.
  • Escribano L, Simón A-M, Gimeno E, Cuadrado-Tejedor M, De Maturana RL, García-Osta A, et al. Rosiglitazone rescues memory impairment in Alzheimer's transgenic mice: mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology. 2010, 35(7), 1593.
  • Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A, Dewachter I, Kuiperi C, et al. Acute treatment with the PPARγ agonist pioglitazone and ibuprofen reduces glial inflammation and Aβ1–42 levels in APPV717I transgenic mice. Brain. 2005, 128(6), 1442-53.
  • Searcy JL, Phelps JT, Pancani T, Kadish I, Popovic J, Anderson KL, et al. Long-term pioglitazone treatment improves learning and attenuates pathological markers in a mouse model of Alzheimer's disease. Journal of Alzheimer's Disease. 2012, 30(4), 943-61.
  • Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP, Heneka MT. PPARγ/RXRα-induced and CD36-mediated microglial amyloid-β phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. Journal of Neuroscience. 2012, 32(48), 17321-31.
  • Craft S, Peskind E, Schwartz MW, Schellenberg GD, Raskind M, Porte D. Cerebrospinal fluid and plasma insulin levels in Alzheimer's disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology. 1998, 50(1), 164-8.
  • Gasparini L, Xu H. Potential roles of insulin and IGF-1 in Alzheimer's disease. Trends in neurosciences. 2003, 26(8), 404-6.
  • Steen E, Terry BM, J Rivera E, Cannon JL, Neely TR, Tavares R, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease–is this type 3 diabetes? Journal of Alzheimer's disease. 2005, 7(1), 63-80.
  • De Felice FG, Vieira MN, Bomfim TR, Decker H, Velasco PT, Lambert MP, et al. Protection of synapses against Alzheimer's-linked toxins: insulin signaling prevents the pathogenic binding of Aβ oligomers. Proceedings of the National Academy of Sciences. 2009:pnas. 0809158106.
  • Qu T, Manev R, Manev H. 5-Lipoxygenase (5-LOX) promoter polymorphism in patients with early-onset and late-onset Alzheimer's disease. The Journal of neuropsychiatry and clinical neurosciences. 2001, 13(2), 304-5.
  • Drazen JM, Yandava CN, Dubé L, Szczerback N, Hippensteel R, Pillari A, et al. Pharmacogenetic association between ALOX5 promoter genotype and the response to anti-asthma treatment. Nature genetics. 1999, 22(2), 168.
  • Paris D, Town T, Mori T, Parker TA, Humphrey J, Mullan M. Soluble β-amyloid peptides mediate vasoactivity via activation of a pro-inflammatory pathway. Neurobiology of aging. 2000, 21(2), 183-97.
  • Manev H, Uz T, Sugaya K, Qu T. Putative role of neuronal 5-lipoxygenase in an aging brain. The FASEB Journal. 2000, 14(10), 1464-9.
  • Chu J, Li J-G, Pratico D. Zileuton improves memory deficits, amyloid and tau pathology in a mouse model of Alzheimer’s disease with plaques and tangles. PLoS One. 2013, 8(8), e70991.
  • Costa Silva B, Silva de Miranda A, Guimaraes Rodrigues F, Leticia Malheiros Silveira A, Henrique de Souza Resende G, Flavio Dutra Moraes M, et al. The 5-lipoxygenase (5-LOX) inhibitor zileuton reduces inflammation and infarct size with improvement in neurological outcome following cerebral ischemia. Current neurovascular research. 2015, 12(4), 398-403.
  • Setter SM, Iltz JL, Fincham JE, Campbell RK, Baker DE. Phosphodiesterase 5 inhibitors for erectile dysfunction. Annals of Pharmacotherapy. 2005, 39(7-8),1286-95.
  • Ahn GJ, Yu JY, Choi SM, Kang KK, Ahn BO, Kwon JW, et al. Chronic administration of phosphodiesterase 5 inhibitor improves erectile and endothelial function in a rat model of diabetes. International journal of andrology. 2005, 28(5), 260-6.
  • Cuadrado‐Tejedor M, Hervias I, Ricobaraza A, Puerta E, Pérez‐Roldán J, García‐Barroso C, et al. Sildenafil restores cognitive function without affecting β‐amyloid burden in a mouse model of Alzheimer's disease. British journal of pharmacology. 2011, 164(8), 2029-41.
  • Garcia-Osta A, Aguirre N, Franco R, Garcia-Barroso C, Perez-Roldan J, Puerta E, et al. Sildenafil restores cognitive function without affecting β-amyloid burden in a mouse model of Alzheimer's disease. 2011.
  • Ding Y, Qiao A, Wang Z, Goodwin JS, Lee E-S, Block ML, et al. Retinoic acid attenuates β-amyloid deposition and rescues memory deficits in an Alzheimer's disease transgenic mouse model. Journal of Neuroscience. 2008, 28(45), 11622-34.
  • Jarvis C, Goncalves M, Clarke E, Dogruel M, Kalindjian S, Thomas S, et al. Retinoic acid receptor‐α signalling antagonizes both intracellular and extracellular amyloid‐β production and prevents neuronal cell death caused by amyloid‐β. European Journal of Neuroscience. 2010, 32(8),1246-55.
  • Shudo K, Fukasawa H, Nakagomi M, Yamagata N. Towards retinoid therapy for Alzheimer's disease. Current Alzheimer Research. 2009, 6(3), 302-11.
  • Tippmann F, Hundt J, Schneider A, Endres K, Fahrenholz F. Up-regulation of the α-secretase ADAM10 by retinoic acid receptors and acitretin. The FASEB Journal. 2009, 23(6), 1643-54.
  • Radha Mahendran, Suganya Jeyabasker, Astral Francis, Sharanya Manoharan. Homology Modeling and in silico docking analysis of BDNF in the treatment of Alzheimer’s disease. Research J. Pharm. and Tech. 2017; 10(9): 2899-2906.
  • Vivek Kumar Sharma. Current Therapeutic Strategies for Alzheimer’s disease: A Lost Direction or A Hope Remains?. Research J. Pharmacology and Pharmacodynamics. 2010; 2(3): 215-220.
  • D Rajesh Kumar,M Siva Shankar,P Prathap Reddy, B Ram Sarath Kumar, N Sumalatha A Review on Alzheimer’s Disease. Research Journal of Pharmacology and Pharmacodynamics. 2014; 6(1): 59-63.
  • M. Vijey Aanandhi, Niventhi. A, Rujaswini. T, Hemalatha C.N, Praveen. D. A Comprehensive Review on the Role of Tau Proteins in Alzheimer’s Pathology. Research J. Pharm. and Tech 2018; 11(2):788-790.
  • Chirag K Patel, B Panigrahi, R Badmanaban, CN Patel. Biochemical Origins of Alzheimer’s Disease with Treatment Techniques. Research J. Pharmacology and Pharmacodynamics. 2010; 2(1):33-38.
  • Saudagar RB, Buchake VV, Bachhav RS. Role of Retinoids in treatment of Alzheimer’s disease. Research J. Pharmacology and Pharmacodynamics. 2012; 4(3): 144-149 .
  • Neveda Baskeran. Iron Deficiency and Brain Disorders. Research J. Pharm. and Tech. 7(3): Mar., 2014; Page 352-353.
  • Sree Lekshmi. R. S, P. Shanmugasundaram. Neuroprotective Properties of Statins. Research J. Pharm. and Tech 2018; 11(8): 3581-3584.
  • M. Vijey Aanandhi, Yeshwanth Prasanna Kumar. B, Ranadheer Chowdary. P, Praveen. D. A Review on the Role of Presenilin in Alzheimer’s Disease . Research J. Pharm. and Tech 2018; 11(5):2149-2151.
  • Vasudev Pai, Chandrashekar K. S, C. S. Shreedhara, Aravinda Pai. In-Silico and In-Vitro correlation studies of natural β-secretase inhibitor: An approach towards Alzheimer’s Disease. Research J. Pharm. and Tech 2017; 10(10):3506-3510.

Abstract Views: 192

PDF Views: 0




  • Repurposing Drugs for Management of Alzheimer Disease

Abstract Views: 192  |  PDF Views: 0

Authors

Senthil Venkatachalam
Department of Pharmaceutics, JSS College of Pharmacy, Ooty, Tamilnadu-643001, India
Ayush Jaiswal
Department of Pharmaceutics, JSS College of Pharmacy, Ooty, Tamilnadu-643001, India
Anindita De
Affiliated by JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
Rohith Krishnan Vijayakumar
Affiliated by JSS Academy of Higher Education and Research, Mysuru, Karnataka, India

Abstract


Alzheimer's disease (AD), or Alzheimer’s, It is neurodegenerative disease starts very slowly and with time it got worse one of the biggest challenge of the CNS drug delivery scientist. Mental and physic exercise, Cholinesterase Inhibitors, and avoiding adipositas may reduce the risk of the AD. Currently, existing/approved drugs for the treatment of the AD are based on neurotransmitter or enzyme replacement/modulation. Existing therapies gives just an optimum advantage and consequently, in present, there is an evident desire for advanced medications or therapies. Drug repurposing is one of the latest trend and the alternative approach in drug advancement, with a history of successful reuse of currently available drugs. A signify category of the drugs reuse in the management of the AD among them the prominent category is the anticancer, antiepileptic, antibiotics, ant diabetic etc. The review focuses on mechanism and application for the repurposing of the existing drugs in the light of the clinical and pre-clinical evidences.

Keywords


Alzheimer’s Disease, Repurposing Drugs, Repurposed Drugs Pathways, Preclinical Data, Clinical Evidences.

References