Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

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

General Review Article

Current Drug Targets in Alzheimer’s Associated Memory Impairment: A Comprehensive Review

Author(s): Anna Mathew, Vignesh Balaji E, Sreedhara Ranganath K. Pai, Anoop Kishore, Vasudev Pai, Ramadevi Pemmireddy and Chandrashekar K.S*

Volume 22, Issue 2, 2023

Published on: 27 May, 2022

Page: [255 - 275] Pages: 21

DOI: 10.2174/1871527321666220401124719

Price: $65

Abstract

Alzheimer’s disease (AD) is the most prevalent form of dementia among geriatrics. It is a progressive, degenerative neurologic disorder that causes memory and cognition loss. The accumulation of amyloid fibrils and neurofibrillary tangles in the brain of AD patients is a distinguishing feature of the disease. Therefore, most of the current therapeutic goals are targeting inhibition of beta-amyloid synthesis and aggregation as well as tau phosphorylation and aggregation. There is also a loss of the cholinergic neurons in the basal forebrain, and first-generation therapeutic agents were primarily focused on compensating for this loss of neurons. However, cholinesterase inhibitors can only alleviate cognitive symptoms of AD and cannot reduce the progression of the disease. Understanding the molecular and cellular changes associated with AD pathology has advanced significantly in recent decades. The etiology of AD is complex, with a substantial portion of sporadic AD emerging from unknown reasons and a lesser proportion of early-onset familial AD (FAD) caused by a mutation in several genes, such as the amyloid precursor protein (APP), presenilin 1 (PS1), and presenilin 2 (PS2) genes. Hence, efforts are being made to discover novel strategies for these targets for AD therapy. A new generation of AChE and BChE inhibitors is currently being explored and evaluated in human clinical trials for AD symptomatic treatment. Other approaches for slowing the progression of AD include serotonergic modulation, H3 receptor antagonism, phosphodiesterase, COX-2, and MAO-B inhibition. The present review provides an insight into the possible therapeutic strategies and their molecular mechanisms, enlightening the perception of classical and future treatment approaches.

Keywords: Dementia, Alzheimer’s, neurodegeneration, long-term potentiation, cholinesterase inhibitors, beta-amyloid.

Graphical Abstract
[1]
Schachter AS, Davis KL. Alzheimer’s disease. Dialogues Clin Neurosci 2000; 2(2): 91-100.
[http://dx.doi.org/10.31887/DCNS.2000.2.2/asschachter] [PMID: 22034442]
[2]
Zhang XX, Tian Y, Wang ZT, Ma YH, Tan L, Yu JT. The epidemiology of Alzheimer’s disease modifiable risk factors and prevention. J Prev Alzheimers Dis 2021; 8(3): 313-21.
[PMID: 34101789]
[3]
Murphy MP, LeVine H III. Alzheimer’s disease and the amyloid-β peptide. J Alzheimers Dis 2010; 19(1): 311-23.
[http://dx.doi.org/10.3233/JAD-2010-1221] [PMID: 20061647]
[4]
Rabinovici GD. Late-onset Alzheimer disease. Continuum (Minneap Minn) 2019; 25(1): 14-33.
[http://dx.doi.org/10.1212/CON.0000000000000700] [PMID: 30707185]
[5]
Yiannopoulou KG, Papageorgiou SG. Current and future treatments for Alzheimer’s disease. Ther Adv Neurol Disord 2013; 6(1): 19-33.
[http://dx.doi.org/10.1177/1756285612461679] [PMID: 23277790]
[6]
Morsy A, Trippier PC. Current and emerging pharmacological targets for the treatment of Alzheimer’s disease. J Alzheimers Dis 2019; 72(s1): S145-76.
[http://dx.doi.org/10.3233/JAD-190744] [PMID: 31594236]
[7]
Loera-Valencia R, Cedazo-Minguez A, Kenigsberg PA, et al. Current and emerging avenues for Alzheimer’s disease drug targets. J Intern Med 2019; 286(4): 398-437.
[http://dx.doi.org/10.1111/joim.12959] [PMID: 31286586]
[8]
Contestabile A. The history of the cholinergic hypothesis. Behav Brain Res 2011; 221(2): 334-40.
[http://dx.doi.org/10.1016/j.bbr.2009.12.044] [PMID: 20060018]
[9]
Atack JR, Perry EK, Bonham JR, Perry RH. Molecular forms of acetylcholinesterase and butyrylcholinesterase in human plasma and cerebrospinal fluid. J Neurochem 1987; 48(6): 1845-50.
[http://dx.doi.org/10.1111/j.1471-4159.1987.tb05746.x] [PMID: 3572402]
[10]
Mesulam MM, Guillozet A, Shaw P, Levey A, Duysen EG, Lockridge O. Acetylcholinesterase knockouts establish central cholinergic pathways and can use butyrylcholinesterase to hydrolyze acetylcholine. Neuroscience 2002; 110(4): 627-39.
[http://dx.doi.org/10.1016/S0306-4522(01)00613-3] [PMID: 11934471]
[11]
Perrier AL, Massoulié J, Krejci E. PRiMA: The membrane anchor of acetylcholinesterase in the brain. Neuron 2002; 33(2): 275-85.
[http://dx.doi.org/10.1016/S0896-6273(01)00584-0] [PMID: 11804574]
[12]
Mori F, Cuadra G, Giacobini E. Metrifonate effects on acetylcholine and biogenic amines in rat cortex. Neurochem Res 1995; 20(9): 1081-8.
[http://dx.doi.org/10.1007/BF00995563] [PMID: 8570013]
[13]
Farlow M. A clinical overview of cholinesterase inhibitors in Alzheimer’s disease. Int Psychogeriatr 2002; 14(S1) (Suppl. 1): 93-126.
[http://dx.doi.org/10.1017/S1041610203008688] [PMID: 12636182]
[14]
Moss DE, Perez RG, Kobayashi H. Cholinesterase inhibitor therapy in Alzheimer’s disease: The limits and tolerability of irreversible CNS-selective acetylcholinesterase inhibition in primates. J Alzheimers Dis 2017; 55(3): 1285-94.
[http://dx.doi.org/10.3233/JAD-160733] [PMID: 27858711]
[15]
Giacobini E. Cholinesterases: New roles in brain function and in Alzheimer’s disease. Neurochem Res 2003; 28(3-4): 515-22.
[http://dx.doi.org/10.1023/A:1022869222652] [PMID: 12675140]
[16]
Greig NH, Utsuki T, Yu Q, et al. A new therapeutic target in Alzheimer’s disease treatment: Attention to butyrylcholinesterase. Curr Med Res Opin 2001; 17(3): 159-65.
[http://dx.doi.org/10.1185/03007990152673800] [PMID: 11900310]
[17]
Darreh-Shori T, Almkvist O, Guan ZZ, et al. Sustained cholinesterase inhibition in AD patients receiving rivastigmine for 12 months. Neurology 2002; 59(4): 563-72.
[http://dx.doi.org/10.1212/WNL.59.4.563] [PMID: 12196650]
[18]
Arendt T, Brückner MK, Lange M, Bigl V. Changes in acetylcholinesterase and butyrylcholinesterase in Alzheimer’s disease resemble embryonic development--a study of molecular forms. Neurochem Int 1992; 21(3): 381-96.
[http://dx.doi.org/10.1016/0197-0186(92)90189-X] [PMID: 1303164]
[19]
Lane RM, Potkin SG, Enz A. Targeting acetylcholinesterase and butyrylcholinesterase in dementia. Int J Neuropsychopharmacol 2006; 9(1): 101-24.
[http://dx.doi.org/10.1017/S1461145705005833] [PMID: 16083515]
[20]
Winblad B, Möbius HJ, Stöffler A. Glutamate receptors as a target for Alzheimer’s disease--are clinical results supporting the hope? J Neural Transm Suppl 2002; 62: 217-25.
[http://dx.doi.org/10.1007/978-3-7091-6139-5_20] [PMID: 12456065]
[21]
Molinuevo JL, Lladó A, Rami L. Memantine: Targeting glutamate excitotoxicity in Alzheimer’s disease and other dementias. Am J Alzheimers Dis Other Demen 2005; 20(2): 77-85.
[http://dx.doi.org/10.1177/153331750502000206] [PMID: 15844753]
[22]
Bloom GS. Amyloid-β and tau: The trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol 2014; 71(4): 505-8.
[http://dx.doi.org/10.1001/jamaneurol.2013.5847] [PMID: 24493463]
[23]
Avila J, Llorens-Martín M, Pallas-Bazarra N, et al. Cognitive decline in neuronal aging and Alzheimer’s disease: Role of NMDA receptors and associated proteins. Front Neurosci 2017; 11: 626.
[http://dx.doi.org/10.3389/fnins.2017.00626] [PMID: 29176942]
[24]
Mota SI, Ferreira IL, Rego AC. Neuropharmacology Dysfunctional synapse in Alzheimer’s disease - A focus on NMDA receptors. Neuropharmacology 2013; 1-11.
[25]
Traynelis SF, Wollmuth LP, McBain CJ, et al. Glutamate receptor ion channels: Structure, regulation, and function. Pharmacol Rev 2010; 62(3): 405-96.
[http://dx.doi.org/10.1124/pr.109.002451] [PMID: 20716669]
[26]
Wang R, Reddy PH. Role of glutamate and NMDA receptors in Alzheimer’s disease. J Alzheimers Dis 2017; 57(4): 1041-8.
[http://dx.doi.org/10.3233/JAD-160763] [PMID: 27662322]
[27]
Lüscher C, Malenka RC. NMDA receptor-dependent long-term potentiation and long-term depression (LTP/LTD). Cold Spring Harb Perspect Biol 2012; 4(6): 1-15.
[http://dx.doi.org/10.1101/cshperspect.a005710] [PMID: 22510460]
[28]
Kullmann DM, Lamsa KP. Long-term synaptic plasticity in hippocampal interneurons. Nat Rev Neurosci 2007; 8(9): 687-99.
[http://dx.doi.org/10.1038/nrn2207] [PMID: 17704811]
[29]
Olivares D, Deshpande VK, Shi Y, et al. N-Methyl D-Aspartate (NMDA) receptor antagonists and memantine treatment for Alzheimer’s disease, vascular dementia and Parkinson’s disease. Curr Alzheimer Res 2012; 9(6): 746-58.
[http://dx.doi.org/10.2174/156720512801322564] [PMID: 21875407]
[30]
Arundine M, Tymianski M. Molecular mechanisms of glutamate-dependent neurodegeneration in ischemia and traumatic brain injury. Cell Mol Life Sci 2004; 61(6): 657-68.
[http://dx.doi.org/10.1007/s00018-003-3319-x] [PMID: 15052409]
[31]
Lu XC, Williams AJ, Wagstaff JD, Tortella FC, Hartings JA. Effects of delayed intrathecal infusion of an NMDA receptor antagonist on ischemic injury and peri-infarct depolarizations. Brain Res 2005; 1056(2): 200-8.
[http://dx.doi.org/10.1016/j.brainres.2005.07.041] [PMID: 16112094]
[32]
Lipton SA. Failures and successes of NMDA receptor antagonists: Molecular basis for the use of open-channel blockers like memantine in the treatment of acute and chronic neurologic insults. NeuroRx 2004; 1(1): 101-10.
[http://dx.doi.org/10.1602/neurorx.1.1.101] [PMID: 15717010]
[33]
Garay RP, Grossberg GT. AVP-786 for the treatment of agitation in dementia of the Alzheimer’s type. Expert Opin Investig Drugs 2017; 26(1): 121-32.
[http://dx.doi.org/10.1080/13543784.2017.1267726] [PMID: 27936965]
[34]
The efficacy of galantamine treatment on attention in patients with Alzheimer’s disease Available from: https://www.clinicaltrials. gov/ct2/show/NCT01054976?term=galantamine&draw=3&rank=12(Accessed on 19 November 2020).
[35]
A study to evaluate the safety and tolerability of SAGE-718 in participants with mild cognitive impairment or mild dementia due to Alzheimer’s Disease (AD) Available from: https://www. clinicaltrials.gov/ct2/show/NCT04602624?term=sage718&draw=2&rank=2(Accessed on 19 November 2020).
[36]
Octohydroaminoacridine succinate tablet for mild-to-moderate Alzheimer’s disease. Available from: https://www.clinicaltrials. gov/ct2/show/NCT03283059?term=octohydroaminoacridine+succinate&draw=2&rank=2(Accessed on 19 November 2020).
[37]
Rivastigmine Bioequivalence Trial With Multiple Application of Transdermal Patches. Available from: https://www.clinicaltrials. gov/ct2/show/NCT03659435?term=RID-TDS&draw=2&rank=1(Accessed on 19 November 2020).
[38]
SUVN-502 with donepezil and memantine for the treatment of moderate alzheimer’s disease- phase 2a study Available from: https://www.clinicaltrials.gov/ct2/show/NCT02580305?term=Donepezil%2Fmemantine+extended+release&draw=2&rank=1(Accessed on 19 November 2020).
[39]
Magnetic Resonance Spectroscopy Study of Memantine in Alzheimer’s Disease - Full Text View - ClinicalTrials. Available from: https://www.clinicaltrials.gov/ct2/show/NCT00551161?term=memantine&draw=5&rank=37(Accessed on 19 November 2020).
[40]
NextStep. Study to evaluate safety,efficacy & tolerability of rivastigmine patch in mild to moderate Alzheimer’s patients Available from: https://www.clinicaltrials.gov/ct2/show/NCT02703636?term=rivastigmine&draw=2&rank=1(Accessed on 19 November 2020).
[41]
Efficacy, safety and tolerability of rivastigmine patch in patients with mild to moderate alzheimer’s disease switched from cholinesterase inhibitors. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01529619?term=galantamine+transdermal+patch&draw=2&rank=1(Accessed on 19 November 2020).
[42]
Jia J, Wei C, Chen W, et al. Safety and efficacy of donepezil 10 mg/day in patients with mild to moderate Alzheimer’s disease. J Alzheimers Dis 2020; 74(1): 199-211.
[http://dx.doi.org/10.3233/JAD-190940] [PMID: 31985467]
[43]
Bisnorcymserine in healthy adult volunteers.. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01747213?term=bisnorcymserine&draw=2&rank=1(Accessed on 19 November 2020).
[44]
A multinational, multi-center, randomized, double-blind, active comparator, phase III clinical trial to evaluate the efficacy and safety of donepezil transdermal patch in patients with alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03197740?term=donepezil+transdermal+patch&draw=2&rank=2(Accessed on 19 November 2020).
[45]
OLE for patients enrolled in phase 2b/3 study ANAVEX2-73-AD004. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04314934?term=anavex&draw=2&rank=3(Accessed on 19 November 2020).
[46]
Evaluate the safety and efficacy of AD-35 tablet in subjects with mild to moderate Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03790982?term=ad35&draw=2&rank=1(Accessed on 19 November 2020).
[47]
A dose titration study of CPC-201 in patients with dementia of Alzheimer’s type. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02549196?term=donepezil%2Fsolifenacin&draw=2&rank=1(Accessed on 19 November 2020).
[48]
Huperzine-A to Help With Mental Problems and the Inability to Care for Onself in Patients With Schizophrenia - Full Text View - ClinicalTrials. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01012830?term=huperzine+A&draw=4&rank=6(Accessed on 19 November 2020).
[49]
Reitz C. Alzheimer’s disease and the amyloid cascade hypothesis: A critical review. Int J Alzheimers Dis 2012; 2012: 369808.
[http://dx.doi.org/10.1155/2012/369808]
[50]
Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease. Amyloid Int J Exp Clin Investig 2002; 297: 353-7.
[51]
Bhadbhade A, Cheng DW. Amyloid precursor protein processing in Alzheimer’s disease. Iran J Child Neurol 2012; 6: 1-4.
[52]
Findeis MA. The role of amyloid β peptide 42 in Alzheimer’s disease. Pharmacol Ther 2007; 116(2): 266-86.
[http://dx.doi.org/10.1016/j.pharmthera.2007.06.006] [PMID: 17716740]
[53]
Meyer-Luehmann M, Spires-Jones TL, Prada C, et al. Rapid appearance and local toxicity of amyloid-β plaques in a mouse model of Alzheimer’s disease. Nature 2008; 451(7179): 720-4.
[http://dx.doi.org/10.1038/nature06616] [PMID: 18256671]
[54]
Brion JP. Neurofibrillary tangles and Alzheimer’s disease. Eur Neurol 1998; 40(3): 130-40.
[http://dx.doi.org/10.1159/000007969] [PMID: 9748670]
[55]
Furukawa K, Sopher BL, Rydel RE, et al. Increased activity-regulating and neuroprotective efficacy of α-secretase-derived secreted amyloid precursor protein conferred by a C-terminal heparin-binding domain. J Neurochem 1996; 67(5): 1882-96.
[http://dx.doi.org/10.1046/j.1471-4159.1996.67051882.x] [PMID: 8863493]
[56]
Kuhn PH, Wang H, Dislich B, et al. ADAM10 is the physiologically relevant, constitutive α-secretase of the amyloid precursor protein in primary neurons. EMBO J 2010; 29(17): 3020-32.
[http://dx.doi.org/10.1038/emboj.2010.167] [PMID: 20676056]
[57]
De Strooper B, Vassar R, Golde T. The secretases: Enzymes with therapeutic potential in Alzheimer disease. Nat Rev Neurol 2010; 6(2): 99-107.
[http://dx.doi.org/10.1038/nrneurol.2009.218] [PMID: 20139999]
[58]
Filip V, Kolibás E. Selegiline in the treatment of Alzheimer’s disease: A long-term randomized placebo-controlled trial. J Psychiatry Neurosci 1999; 24(3): 234-43.
[PMID: 10354658]
[59]
Parvathy S, Ehrlich M, Pedrini S, et al. Atorvastatin-induced activation of Alzheimer’s α secretase is resistant to standard inhibitors of protein phosphorylation-regulated ectodomain shedding. J Neurochem 2004; 90(4): 1005-10.
[http://dx.doi.org/10.1111/j.1471-4159.2004.02521.x] [PMID: 15287907]
[60]
Yang L-B, Lindholm K, Yan R, et al. Elevated β-secretase expression and enzymatic activity detected in sporadic Alzheimer disease. Nat Med 2003; 9(1): 3-4.
[http://dx.doi.org/10.1038/nm0103-3] [PMID: 12514700]
[61]
Cai XD, Golde TE, Younkin SG. Release of excess amyloid β protein from a mutant amyloid β protein precursor. Science 1993; 259: 514-6.
[62]
Jonsson T, Atwal JK, Steinberg S, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature 2012; 488(7409): 96-9.
[http://dx.doi.org/10.1038/nature11283] [PMID: 22801501]
[63]
Yan R. Stepping closer to treating Alzheimer’s disease patients with BACE1 inhibitor drugs. Transl Neurodegener 2016; 5(1): 13.
[http://dx.doi.org/10.1186/s40035-016-0061-5] [PMID: 27418961]
[64]
A 24-Month study to evaluate the efficacy and safety of elenbecestat (E2609) in subjects with early Alzheimer’s disease (MissionAD1). 2020. Available from: https://clinicaltrials.gov/ct2/show/NCT02956486
[65]
De Strooper B, Annaert W. Proteolytic processing and cell biological functions of the amyloid precursor protein. J Cell Sci 2000; 113(Pt 11): 1857-70.
[http://dx.doi.org/10.1242/jcs.113.11.1857] [PMID: 10806097]
[66]
Esler WP, Kimberly WT, Ostaszewski BL, et al. Activity-dependent isolation of the presenilin- γ -secretase complex reveals nicastrin and a γ substrate. Proc Natl Acad Sci USA 2002; 99(5): 2720-5.
[http://dx.doi.org/10.1073/pnas.052436599] [PMID: 11867728]
[67]
MacLeod R, Hillert EK, Cameron RT, Baillie GS. The role and therapeutic targeting of α-, β- and γ-secretase in Alzheimer’s disease. Future Sci OA 2015; 1(3): FSO11.
[http://dx.doi.org/10.4155/fso.15.9] [PMID: 28031886]
[68]
Lobello K, Ryan JM, Liu E, Rippon G, Black R. Targeting Beta amyloid: A clinical review of immunotherapeutic approaches in Alzheimer’s disease. Int J Alzheimers Dis 2012; 2012: 628070.
[http://dx.doi.org/10.1155/2012/628070] [PMID: 22292124]
[69]
Mehta D, Jackson R, Paul G, Shi J, Sabbagh M. Why do trials for Alzheimer’s disease drugs keep failing? A discontinued drug perspective for 2010-2015. Expert Opin Investig Drugs 2017; 26(6): 735-9.
[http://dx.doi.org/10.1080/13543784.2017.1323868] [PMID: 28460541]
[70]
A study to evaluate safety and tolerability of aducanumab in participants with alzheimer’s disease who had previously participated in the aducanumab studies 221AD103, 221AD301, 221AD302 and 221AD205. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04241068?term=aducanumab&draw=2&rank=1(Accessed on 15 November 2020).
[71]
Safety, tolerability, PK and PD of Posiphen® in subjects with early Alzheimer’s disease. Available from: https://www.clinicaltrials. gov/ct2/show/NCT02925650?term=posiphen&draw=2&rank=3(Accessed on 15 November 2020).
[72]
Open-Label Extension of the Phase III Study With Tramiprosate (3APS) in Patients With Mild to Moderate Alzheimer’s Disease.. Available from: https://www.clinicaltrials.gov/ct2/show/NCT00314912?term=tramiprosate&draw=2&rank=2(Accessed on 15 November 2020).
[73]
A Study to Evaluate the Safety and Tolerability of Long-term Administration of Gantenerumab in Participants With Alzheimer’s Disease (AD). Available from: https://www.clinicaltrials.gov/ct2/show/NCT04339413?term=gantenerumab&draw=2&rank=3(Accessed on 16 November 2020).
[74]
Safety and efficacy study of ALZT-OP1 in subjects with evidence of early Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02547818?term=NCT02547818&draw=2&rank=1(Accessed on 16 November 2020).
[75]
MAD Phase I study to investigate contraloid acetate.. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03955380?term=contraloid&draw=2&rank=1(Accessed on 16 November 2020).
[76]
Lacosta A-M, Pascual-Lucas M, Pesini P, et al. Safety, tolerability and immunogenicity of an active anti-Aβ40 vaccine (ABvac40) in patients with Alzheimer’s disease: A randomised, double-blind, placebo-controlled, phase I trial. Alzheimers Res Ther 2018; 10(1): 12.
[http://dx.doi.org/10.1186/s13195-018-0340-8] [PMID: 29378651]
[77]
A repeated dose study of khk6640 in japanese patients with Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03093519?term=khk6640&draw=2&rank=3(Accessed on 16 November 2020).
[78]
Dominantly inherited Alzheimer network trial: An opportunity to prevent dementia. a study of potential disease modifying treatments in individuals at risk for or with a type of early onset Alzheimer’s disease caused by a genetic mutation. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04623242?term=Solanezumab&draw=2&rank=10(Accessed on 16 November 2020).
[79]
A Single ascending dose study to investigate the safety, tolerability, immunogenicity and pharmacokinetics of intravenously administered RO7126209 in healthy participants. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04023994?term=RO7126209&draw=2&rank=1(Accessed on 16 November 2020).
[80]
A study to evaluate efficacy and safety of treatment with BAN2401 in participants with preclinical alzheimer’s disease and elevated amyloid and also in participants with early preclinical alzheimer’s disease and intermediate amyloid. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04468659?term=ban2401&draw=2&rank=1(Accessed on 16 November 2020).
[81]
A study of donanemab (LY3002813) in participants with early alzheimer’s disease (TRAILBLAZER-ALZ 2) Available from: https://www.clinicaltrials.gov/ct2/show/NCT04437511?term=donanemab&draw=2&rank=1(Accessed on 16 November 2020).
[82]
An open-label crenezumab study in participants with alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03491150?term=crenezumab&draw=2&rank=1(Accessed on 18 November 2020).
[83]
A 24-month study to evaluate the efficacy and safety of elenbecestat (E2609) in subjects with early Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03036280?term=elenbecestat&draw=2&rank=2(Accessed on 18 November 2020).
[84]
Safety, Tolerability and abeta-specific antibody response of repeated i.m. injections of adjuvanted cad106 in mild alzheimer patients. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01097096?term=cad+106&draw=2&rank=7(Accessed on 18 November 2020).
[85]
A phase 3 study evaluating safety and effectiveness of Immune Globulin Intravenous (IGIV 10%) for the treatment of mild-tomoderate Alzheimer´s disease. Available from: https://www. clinicaltrials.gov/ct2/show/NCT00818662?term=flebogamma&draw=6&rank=43(Accessed on 18 November 2020).
[86]
A study of sodium oligomannate (GV-971) in participants with mild to moderate Alzheimer’s disease Available from: https://www.clinicaltrials.gov/ct2/show/NCT04520412?term=gv+971&draw=2&rank=1(Accessed on 18 November 2020).
[87]
An extension study of V203-AD study to evaluate the safety, tolerability, immunogenicity, and efficacy of UB-311 Available from: https://www.clinicaltrials.gov/ct2/show/NCT03531710?term=ub311&draw=2&rank=2(Accessed on 18 November 2020).
[88]
SAD/MAD study to assess safety, tolerability, PK & PD of MEDI1814 in subjects with mild-moderate Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02036645?term=medi1814&draw=2&rank=1(Accessed on 18 November 2020).
[89]
A study of LY3202626 on disease progression in participants with mild Alzheimer’s disease dementia. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02791191?term=ly3202626&draw=2&rank=3(Accessed on 18 November 2020).
[90]
Evaluate the efficacy and safety of ID1201 for dose-finding in mild Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03363269?term=id1201&draw=2&rank=1(Accessed on 18 November 2020).
[91]
Efficacy and safety of T-817MA in patients with mild cognitive impairment due to Alzheimer’s Disease (AD) or Mild AD Available from: https://www.clinicaltrials.gov/ct2/show/NCT04191486?term=t817ma&draw=2&rank=1(Accessed on 18 November 2020).
[92]
Study to evaluate dietary supplementation with a version of anatabloc(R) in subjects with alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01669876?term=anatabine&draw=2&rank=6(Accessed on 18 November 2020).
[93]
Evaluate the safety and efficacy of AD-35 tablet in subjects with mild to moderate alzheimer’s disease Available from: https://www.clinicaltrials.gov/ct2/show/NCT03790982?term=ad35&draw=2&rank=1(Accessed on 18 November 2020).
[94]
Efficacy and safety study of ELND005 as a treatment for agitation and aggression in Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01735630?term=scyllo+inositol&draw=2&rank=2(Accessed on 18 November 2020).
[95]
A single site, randomized, double-blind, placebo controlled trial of NIC5-15 in subjects with Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01928420?term=pinitol&draw=2&rank=3(Accessed on 18 November 2020).
[96]
Posiphen® dose-finding, biomarker study in early Alzheimer’s and Parkinson’s patients. Available from: https://www.clinicaltrials. gov/ct2/show/NCT04524351?term=posiphen&draw=2&rank=1(Accessed on 18 November 2020).
[97]
Rafii MS. Targeting tau protein in Alzheimer’s disease. Lancet 2016; 388(10062): 2842-4.
[http://dx.doi.org/10.1016/S0140-6736(16)32107-9] [PMID: 27863808]
[98]
Köpke E, Tung YC, Shaikh S, Alonso AC, Iqbal K, Grundke-Iqbal I. Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease. J Biol Chem 1993; 268(32): 24374-84.
[http://dx.doi.org/10.1016/S0021-9258(20)80536-5] [PMID: 8226987]
[99]
Alonso AC, Zaidi T, Grundke-Iqbal I, Iqbal K. Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease. Proc Natl Acad Sci USA 1994; 91(12): 5562-6.
[http://dx.doi.org/10.1073/pnas.91.12.5562] [PMID: 8202528]
[100]
Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, Binder LI. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci USA 1986; 83(13): 4913-7.
[http://dx.doi.org/10.1073/pnas.83.13.4913] [PMID: 3088567]
[101]
Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology 1992; 42(3 Pt 1): 631-9.
[http://dx.doi.org/10.1212/WNL.42.3.631] [PMID: 1549228]
[102]
Froelich S, Houlden H, Pickering-brown S, et al. Association] of missense and 5 J -splice-site mutations in. Nature 1998; 393: 702-5.
[http://dx.doi.org/10.1038/31508]
[103]
Roberson ED, Scearce-Levie K, Palop JJ, et al. Reducing endogenous tau ameliorates amyloid ß-induced deficits in an Alzheimer’s disease mouse model. Science (80- ) 2014; 316: 750-3.
[104]
Oddo S, Vasilevko V, Caccamo A, Kitazawa M, Cribbs DH, LaFerla FM. Reduction of soluble Abeta and tau, but not soluble Abeta alone, ameliorates cognitive decline in transgenic mice with plaques and tangles. J Biol Chem 2006; 281(51): 39413-23.
[http://dx.doi.org/10.1074/jbc.M608485200] [PMID: 17056594]
[105]
Ferrer I, Gomez-Isla T, Puig B, et al. Current advances on different kinases involved in tau phosphorylation, and implications in Alzheimer’s disease and tauopathies. Curr Alzheimer Res 2005; 2(1): 3-18.
[http://dx.doi.org/10.2174/1567205052772713] [PMID: 15977985]
[106]
Liu R, Tian Q. Protein phosphatase 2A, a key player in Alzheimer’s disease. Front Med China 2009; 3(1): 8-12.
[http://dx.doi.org/10.1007/s11684-009-0017-6]
[107]
Giese KP. GSK-3: A key player in neurodegeneration and memory. IUBMB Life 2009; 61(5): 516-21.
[http://dx.doi.org/10.1002/iub.187] [PMID: 19391164]
[108]
Wen Y, Planel E, Herman M, et al. Interplay between cyclin-dependent kinase 5 and glycogen synthase kinase 3 beta mediated by neuregulin signaling leads to differential effects on tau phosphorylation and amyloid precursor protein processing. J Neurosci 2008; 28(10): 2624-32.
[http://dx.doi.org/10.1523/JNEUROSCI.5245-07.2008] [PMID: 18322105]
[109]
Churcher I. Tau therapeutic strategies for the treatment of Alzheimer’s disease. Curr Top Med Chem 2006; 6(6): 579-95.
[http://dx.doi.org/10.2174/156802606776743057] [PMID: 16712493]
[110]
Meijer L, Borgne A, Mulner O, et al. Biochemical and cellular effects of roscovitine, a potent and selective inhibitor of the cyclin-dependent kinases cdc2, cdk2 and cdk5. Eur J Biochem 1997; 243(1-2): 527-36.
[http://dx.doi.org/10.1111/j.1432-1033.1997.t01-2-00527.x] [PMID: 9030781]
[111]
Kesavapany S, Zheng YL, Amin N, Pant HC. Peptides derived from Cdk5 activator p35, specifically inhibit deregulated activity of Cdk5. Biotechnol J 2007; 2(8): 978-87.
[http://dx.doi.org/10.1002/biot.200700057] [PMID: 17526058]
[112]
Vijayan S, El-Akkad E, Grundke-Iqbal I, Iqbal K. A pool of beta-tubulin is hyperphosphorylated at serine residues in Alzheimer disease brain. FEBS Lett 2001; 509(3): 375-81.
[http://dx.doi.org/10.1016/S0014-5793(01)03201-X] [PMID: 11749959]
[113]
Tanimukai H, Grundke-Iqbal I, Iqbal K. Up-regulation of inhibitors of protein phosphatase-2A in Alzheimer’s disease. Am J Pathol 2005; 166(6): 1761-71.
[http://dx.doi.org/10.1016/S0002-9440(10)62486-8] [PMID: 15920161]
[114]
Irwin DJ. Tauopathies as clinicopathological entities David. Parkinsonism Relat Disord 2017; 22: 1-12.
[115]
Phase 2 study of biib092 in participants with early alzheimer’s disease Available from: https://www.clinicaltrials.gov/ct2/show/NCT03352557?term=BIIB&draw=5&rank=32(Accessed on 21 November 2020).
[116]
24 months safety and efficacy study of aadvac1 in patients with mild Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02579252?term=AADvac1&draw=2&rank=3(Accessed on 21 November 2020).
[117]
[118]
Tau PET imaging with 18F-AV-1451 in subjects with MAPT mutations Available from: https://www.clinicaltrials.gov/ct2/show/NCT03186989?term=MAPT&draw=3&rank=12(Accessed on 21 November 2020).
[119]
A study to evaluate the safety, tolerability and immunogenicity of tau targeted vaccines in participants with early Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04445831?term=aci35&draw=2&rank=1(Accessed on 21 November 2020).
[120]
A study of LY3303560 in participants with early symptomatic Alzheimer’s disease. Available from: https://www.clinicaltrials. gov/ct2/show/NCT03518073?term=ly3303560&draw=2&rank=3(Accessed on 21 November 2020).
[121]
An extension study of ABBV-8E12 in early Alzheimer’s Disease (AD) Available from: https://www.clinicaltrials.gov/ct2/show/NCT03712787?term=tilavonemab&draw=2&rank=1(Accessed on 21 November 2020).
[122]
A study of semorinemab in patients with moderate alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03828747?term=semorinemab&draw=2&rank=1(Accessed on 21 November 2020).
[123]
Safety and efficacy of TRx0237 in subjects with alzheimer’s disease followed by open-label treatment. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03446001?term=hydromethylthionine&draw=2&rank=2(Accessed on 21 November 2020).
[124]
Evaluation of [18F]RO6958948 as tracer for Positron Emission Tomography (PET) imaging of tau burden in Alzheimer’s disease participants Available from: https://www.clinicaltrials.gov/ct2/show/NCT02792179?term=RO6958948&draw=2&rank=1(Accessed on 21 November 2020).
[125]
Autopsy follow-up of subjects previously imaged with florbetapir F 18 (18F-AV-45) PET in trial 18F-AV-45-A07. Available from: https://www.clinicaltrials.gov/ct2/show/NCT01447719?term=florbetapir&draw=2&rank=9(Accessed on 21 November 2020).
[126]
Single-ascending-dose study of BIIB076 in healthy volunteers and participants with Alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03056729?term=BIIB076&draw=2&rank=1(Accessed on 21 November 2020).
[127]
A Study to Assess Safety, Tolerability, Pharmacokinetics (PK), Immunogenicity, and Pharmacodynamics (PD) of Intravenous Infusions of E2814 in Healthy Participants. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04231513?term=E2814&draw=2&rank=1(Accessed on 21 November 2020).
[128]
A study of JNJ-63733657 in participants with early alzheimer’s disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04619420?term=JNJ63733657&draw=2&rank=1(Accessed on 21 November 2020).
[129]
Study With Lu AF87908 in Healthy Subjects and Patients With Alzheimer’s Disease. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04149860?term=Lu-AF&draw=2&rank=10(Accessed on 21 November 2020).
[130]
The influence of vascular burden, amyloid plaque and tau protein in patients with vascular cognitive impairment and dementia with tauopathy. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04309253?term=PMPBB3&draw=2&rank=6(Accessed on 21 November 2020).
[131]
Cramer PE, Cirrito JR, Wesson DW, et al. ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficits in AD mouse models. Science (80- ) 2018; 335: 1503-6.
[132]
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]
[133]
Zhao J, Fu Y, Liu CC, et al. Retinoic acid isomers facilitate apolipoprotein E production and lipidation in astrocytes through the retinoid X receptor/retinoic acid receptor pathway. J Biol Chem 2014; 289(16): 11282-92.
[http://dx.doi.org/10.1074/jbc.M113.526095] [PMID: 24599963]
[134]
Skerrett R, Pellegrino MP, Casali BT, Taraboanta L, Landreth GE. Combined liver X receptor/peroxisome proliferator-activated receptor γ agonist treatment reduces amyloid β levels and improves behavior in amyloid precursor protein/presenilin 1 mice. J Biol Chem 2015; 290(35): 21591-602.
[http://dx.doi.org/10.1074/jbc.M115.652008] [PMID: 26163517]
[135]
Hong C, Tontonoz P. Liver X receptors in lipid metabolism: Opportunities for drug discovery. Nat Rev Drug Discov 2014; 13(6): 433-44.
[http://dx.doi.org/10.1038/nrd4280] [PMID: 24833295]
[136]
Ghosal K, Stathopoulos A, Thomas D, Phenis D, Vitek MP, Pimplikar SW. The apolipoprotein-E-mimetic COG112 protects amyloid precursor protein intracellular domain-overexpressing animals from Alzheimer’s disease-like pathological features. Neurodegener Dis 2013; 12(1): 51-8.
[http://dx.doi.org/10.1159/000341299] [PMID: 22965147]
[137]
Hudry E, Dashkoff J, Roe AD, et al. Gene transfer of human Apoe isoforms results in differential modulation of amyloid deposition and neurotoxicity in mouse brain. Sci Transl Med 2013; 5(212): 212ra161.
[http://dx.doi.org/10.1126/scitranslmed.3007000] [PMID: 24259049]
[138]
Suidan GL, Ramaswamy G. Targeting apolipoprotein E for Alzheimer’s disease: An industry perspective. Int J Mol Sci 2019; 20(9): E2161.
[http://dx.doi.org/10.3390/ijms20092161] [PMID: 31052389]
[139]
Lander ES. Brave new genome. N Engl J Med 2018; 169-74.
[PMID: 26039524]
[140]
Rabinowitz R, Kadair A, Ben-Zur T, Michaelson D, Offen D. ApoE4 allele specific knockout using a synthetic Cas9 variant as a potential gene therapy approach for Alzheimer’s disease. Cytotherapy 2019; 21(5): e7.
[http://dx.doi.org/10.1016/j.jcyt.2019.04.022]
[141]
Kuszczyk MA, Sanchez S, Pankiewicz J, et al. Blocking the interaction between apolipoprotein E and Aβ reduces intraneuronal accumulation of Aβ and inhibits synaptic degeneration. Am J Pathol 2013; 182(5): 1750-68.
[http://dx.doi.org/10.1016/j.ajpath.2013.01.034] [PMID: 23499462]
[142]
Shibuya Y, Chang CCY, Huang LH, Bryleva EY, Chang TY. Inhibiting ACAT1/SOAT1 in microglia stimulates autophagy-mediated lysosomal proteolysis and increases Aβ1-42 clearance. J Neurosci 2014; 34(43): 14484-501.
[http://dx.doi.org/10.1523/JNEUROSCI.2567-14.2014] [PMID: 25339759]
[143]
Castranio EL, Wolfe CM, Nam KN, et al. ABCA1 haplodeficiency affects the brain transcriptome following traumatic brain injury in mice expressing human APOE isoforms. Acta Neuropathol Commun 2018; 6(1): 69.
[http://dx.doi.org/10.1186/s40478-018-0569-2] [PMID: 30049279]
[144]
Boehm-Cagan A, Bar R, Harats D, et al. Differential effects of apoE4 and activation of ABCA1 on brain and plasma lipoproteins. PLoS One 2016; 11(11): e0166195.
[http://dx.doi.org/10.1371/journal.pone.0166195] [PMID: 27824936]
[145]
Huynh TV, Liao F, Francis CM, et al. Age-dependent effects of apoE reduction using antisense oligonucleotides in a model of β-amyloidosis. Neuron 2017; 96(5): 1013-1023.e4.
[http://dx.doi.org/10.1016/j.neuron.2017.11.014] [PMID: 29216448]
[146]
Buhot MC, Martin S, Segu L. Role of serotonin in memory impairment. Ann Med 2000; 32(3): 210-21.
[http://dx.doi.org/10.3109/07853890008998828] [PMID: 10821328]
[147]
Barnes NM, Sharp T. A review of central 5-HT receptors and their function. Neuropharmacology 1999; 38(8): 1083-152.
[http://dx.doi.org/10.1016/S0028-3908(99)00010-6] [PMID: 10462127]
[148]
Bender E, Pindon A, van Oers I, et al. Structure of the human serotonin 5-HT4 receptor gene and cloning of a novel 5-HT4 splice variant. J Neurochem 2000; 74(2): 478-89.
[http://dx.doi.org/10.1046/j.1471-4159.2000.740478.x] [PMID: 10646498]
[149]
Claeysen S, Bockaert J, Giannoni P. Serotonin: A new hope in Alzheimer’s disease? ACS Chem Neurosci 2015; 6(7): 940-3.
[http://dx.doi.org/10.1021/acschemneuro.5b00135] [PMID: 26011650]
[150]
Cirrito JR, Disabato BM, Restivo JL, et al. Serotonin signaling is associated with lower amyloid-β levels and plaques in transgenic mice and humans. Proc Natl Acad Sci USA 2011; 108(36): 14968-73.
[http://dx.doi.org/10.1073/pnas.1107411108] [PMID: 21873225]
[151]
Sheline YI, West T, Yarasheski K, et al. An antidepressant decreases CSF Aβ production in healthy individuals and in transgenic AD mice. Sci Transl Med 2014; 6(236): 1-20.
[http://dx.doi.org/10.1126/scitranslmed.3008169]
[152]
Claeysen S. Early administration of RS 67333, a specific 5-HT 4 receptor agonist, prevents amyloidogenesis and behavioral deficits in the 5XFAD mouse model of Alzheimer ’ s disease. Front Aging Neurosci 2013; 5: 1-12.
[153]
Wilkinson D, Windfeld K, Colding-Jørgensen E. Safety and efficacy of idalopirdine, a 5-HT6 receptor antagonist, in patients with moderate Alzheimer’s disease (LADDER): A randomised, double-blind, placebo-controlled phase 2 trial. Lancet Neurol 2014; 13(11): 1092-9.
[http://dx.doi.org/10.1016/S1474-4422(14)70198-X] [PMID: 25297016]
[154]
Lecoutey C, Hedou D, Freret T, et al. Design of donecopride, a dual serotonin subtype 4 receptor agonist/acetylcholinesterase inhibitor with potential interest for Alzheimer’s disease treatment. Proc Natl Acad Sci USA 2014; 111(36): E3825-30.
[http://dx.doi.org/10.1073/pnas.1410315111] [PMID: 25157130]
[155]
Leurs R, Vischer HF, Wijtmans M, de Esch IJ. En route to new blockbuster anti-histamines: Surveying the offspring of the expanding histamine receptor family. Trends Pharmacol Sci 2011; 32(4): 250-7.
[http://dx.doi.org/10.1016/j.tips.2011.02.004] [PMID: 21414671]
[156]
Haas HL, Sergeeva OA, Selbach O. Histamine in the nervous system. Physiol Rev 2008; 88(3): 1183-241.
[http://dx.doi.org/10.1152/physrev.00043.2007] [PMID: 18626069]
[157]
Bacciottini L, Passani MB, Mannaioni PF, Blandina P. Interactions between histaminergic and cholinergic systems in learning and memory. Behav Brain Res 2001; 124(2): 183-94.
[http://dx.doi.org/10.1016/S0166-4328(01)00230-3] [PMID: 11640972]
[158]
Passani MB, Blandina P. Histamine receptors in the CNS as targets for therapeutic intervention. Trends Pharmacol Sci 2011; 32(4): 242-9.
[http://dx.doi.org/10.1016/j.tips.2011.01.003] [PMID: 21324537]
[159]
Fox G, Esbenshade T, Pan J, et al. Pharmacological properties of ABT-239 [4-(2-{2-[(2R) -2-Methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitrile]: II. neurophysiological characterization and broad preclinical efficacy in cognition and schizophrenia of a potent and selective histamine H3 receptor antagonist. Pharmacology 2005; 313: 176-90.
[PMID: 15608077]
[160]
Fu Q, Dai H, He P, et al. The H3 receptor antagonist clobenpropit protects against Abeta42-induced neurotoxicity in differentiated rat PC12 cells. Pharmazie 2010; 65(4): 257-60.
[PMID: 20432621]
[161]
Bitner RS, Markosyan S, Nikkel AL, Brioni JD. In-vivo histamine H3 receptor antagonism activates cellular signaling suggestive of symptomatic and disease modifying efficacy in Alzheimer’s disease. Neuropharmacology 2011; 60(2-3): 460-6.
[http://dx.doi.org/10.1016/j.neuropharm.2010.10.026] [PMID: 21044639]
[162]
Bitner RS, Nikkel AL, Markosyan S, Otte S, Puttfarcken P, Gopalakrishnan M. Selective α7 nicotinic acetylcholine receptor activation regulates glycogen synthase kinase3β and decreases tau phosphorylation in vivo. Brain Res 2009; 1265: 65-74.
[http://dx.doi.org/10.1016/j.brainres.2009.01.069] [PMID: 19230830]
[163]
Giannoni P, Medhurst AD, Passani MB, et al. Regional differential effects of the novel histamine H 3 hydrochloride (GSK189254) on histamine release in the central nervous system of freely moving rats. J Pharmacol Exp Ther 2010; 332: 164-72.
[http://dx.doi.org/10.1124/jpet.109.158444] [PMID: 19815811]
[164]
Ligneau X, Perrin D, Landais L, et al. BF2.649 [1-3-[3-(4-Chlorophenyl)propoxy]propylpiperidine, hydrochloride], a nonimidazole inverse agonist/antagonist at the human histamine H3 receptor: Preclinical pharmacology. J Pharmacol Exp Ther 2007; 320(1): 365-75.
[http://dx.doi.org/10.1124/jpet.106.111039] [PMID: 17005916]
[165]
García-Osta A, Cuadrado-Tejedor M, García-Barroso C, Oyarzábal J, Franco R. Phosphodiesterases as therapeutic targets for Alzheimer’s disease. ACS Chem Neurosci 2012; 3(11): 832-44.
[http://dx.doi.org/10.1021/cn3000907] [PMID: 23173065]
[166]
Tully T. Regulation of gene expression and its role in long-term memory and synaptic plasticity. Proc Natl Acad Sci USA 1997; 94(9): 4239-41.
[http://dx.doi.org/10.1073/pnas.94.9.4239] [PMID: 9113972]
[167]
DeNoble VJ. Vinpocetine enhances retrieval of a step-through passive avoidance response in rats. Pharmacol Biochem Behav 1987; 26(1): 183-6.
[http://dx.doi.org/10.1016/0091-3057(87)90552-1] [PMID: 3562490]
[168]
Boess FG, Hendrix M, van der Staay FJ, et al. Inhibition of phosphodiesterase 2 increases neuronal cGMP, synaptic plasticity and memory performance. Neuropharmacology 2004; 47(7): 1081-92.
[http://dx.doi.org/10.1016/j.neuropharm.2004.07.040] [PMID: 15555642]
[169]
Tully T, Bourtchouladze R, Scott R, Tallman J. Targeting the CREB pathway for memory enhancers. Nat Rev Drug Discov 2003; 2(4): 267-77.
[http://dx.doi.org/10.1038/nrd1061] [PMID: 12669026]
[170]
Barad M, Bourtchouladze R, Winder DG, Golan H, Kandel E. Rolipram, a type IV-specific phosphodiesterase inhibitor, facilitates the establishment of long-lasting long-term potentiation and improves memory. Proc Natl Acad Sci USA 1998; 95(25): 15020-5.
[http://dx.doi.org/10.1073/pnas.95.25.15020] [PMID: 9844008]
[171]
Vardigan JD, Converso A, Hutson PH, Uslaner JM. The selective phosphodiesterase 9 (PDE9) inhibitor PF-04447943 attenuates a scopolamine-induced deficit in a novel rodent attention task. J Neurogenet 2011; 25(4): 120-6.
[http://dx.doi.org/10.3109/01677063.2011.630494] [PMID: 22070409]
[172]
Rodefer JS, Murphy ER, Baxter MG. PDE10A inhibition reverses subchronic PCP-induced deficits in attentional set-shifting in rats. Eur J Neurosci 2005; 21(4): 1070-6.
[http://dx.doi.org/10.1111/j.1460-9568.2005.03937.x] [PMID: 15787711]
[173]
Hoozemans JJM, O’Banion MK. The role of COX-1 and COX-2 in Alzheimer’s disease pathology and the therapeutic potentials of non-steroidal anti-inflammatory drugs. Curr Drug Targets CNS Neurol Disord 2005; 4(3): 307-15.
[http://dx.doi.org/10.2174/1568007054038201] [PMID: 15975032]
[174]
Kotilinek LA, Westerman MA, Wang Q, et al. Cyclooxygenase-2 inhibition improves amyloid-β-mediated suppression of memory and synaptic plasticity. Brain 2008; 131(Pt 3): 651-64.
[http://dx.doi.org/10.1093/brain/awn008] [PMID: 18292081]
[175]
Weggen S, Eriksen JL, Das P, et al. A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity. Nature 2001; 414(6860): 212-6.
[http://dx.doi.org/10.1038/35102591] [PMID: 11700559]
[176]
Teather LA, Packard MG, Bazan NG. Post-training cyclooxygenase-2 (COX-2) inhibition impairs memory consolidation. Learn Mem 2002; 9(1): 41-7.
[http://dx.doi.org/10.1101/lm.43602] [PMID: 11917005]
[177]
Aisen PS. The potential of anti-inflammatory drugs for the treatment of Alzheimer’s disease. Lancet Neurol 2002; 1(5): 279-84.
[http://dx.doi.org/10.1016/S1474-4422(02)00133-3] [PMID: 12849425]
[178]
Pasinetti GM, Aisen PS. Cyclooxygenase-2 expression is increased in frontal cortex of Alzheimer’s disease brain. Neuroscience 1998; 87(2): 319-24.
[http://dx.doi.org/10.1016/S0306-4522(98)00218-8] [PMID: 9740394]
[179]
Lim GP, Yang F, Chu T, et al. Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J Neurosci 2000; 20(15): 5709-14.
[http://dx.doi.org/10.1523/JNEUROSCI.20-15-05709.2000] [PMID: 10908610]
[180]
Cai Z. Monoamine oxidase inhibitors: Promising therapeutic agents for Alzheimer’s disease (Review). Mol Med Rep 2014; 9(5): 1533-41.
[http://dx.doi.org/10.3892/mmr.2014.2040] [PMID: 24626484]
[181]
Jo S, Yarishkin O, Hwang YJ, et al. GABA from reactive astrocytes impairs memory in mouse models of Alzheimer’s disease. Nat Med 2014; 20(8): 886-96.
[http://dx.doi.org/10.1038/nm.3639] [PMID: 24973918]
[182]
Schedin-Weiss S, Inoue M, Hromadkova L, et al. Monoamine oxidase B is elevated in Alzheimer disease neurons, is associated with γ-secretase and regulates neuronal amyloid β-peptide levels. Alzheimers Res Ther 2017; 9(1): 57.
[http://dx.doi.org/10.1186/s13195-017-0279-1] [PMID: 28764767]
[183]
Borroni E, Bohrmann B, Grueninger F, et al. Sembragiline  A novel, selective monoamine oxidase type B inhibitor for the treatment of Alzheimer’s disease. J Pharmacol Exp Ther 2017; 362(3): 413-23.
[http://dx.doi.org/10.1124/jpet.117.241653] [PMID: 28642233]
[184]
Bell KFS, de Kort GJL, Steggerda S, Shigemoto R, Ribeiro-da-Silva A, Cuello AC. Structural involvement of the glutamatergic presynaptic boutons in a transgenic mouse model expressing early onset amyloid pathology. Neurosci Lett 2003; 353(2): 143-7.
[http://dx.doi.org/10.1016/j.neulet.2003.09.027] [PMID: 14664921]
[185]
Kehrer C, Maziashvili N, Dugladze T, Gloveli T. Altered excitatory-inhibitory balance in the NMDA-hypofunction model of schizophrenia. Front Mol Neurosci 2008; 1: 6.
[http://dx.doi.org/10.3389/neuro.02.006.2008] [PMID: 18946539]
[186]
Mitew S, Kirkcaldie MTK, Dickson TC, Vickers JC. Altered synapses and gliotransmission in Alzheimer’s disease and AD model mice. Neurobiol Aging 2013; 34(10): 2341-51.
[http://dx.doi.org/10.1016/j.neurobiolaging.2013.04.010] [PMID: 23643146]
[187]
DeBoer P, Westerink BHC. GABAergic modulation of striatal cholinergic interneurons: An in vivo microdialysis study. J Neurochem 1994; 62(1): 70-5.
[http://dx.doi.org/10.1046/j.1471-4159.1994.62010070.x] [PMID: 8263546]
[188]
Calvo-Flores Guzmán B, Vinnakota C, Govindpani K, Waldvogel HJ, Faull RLM, Kwakowsky A. The GABAergic system as a therapeutic target for Alzheimer’s disease. J Neurochem 2018; 146(6): 649-69.
[http://dx.doi.org/10.1111/jnc.14345] [PMID: 29645219]
[189]
Pilipenko V, Narbute K, Beitnere U, et al. Very low doses of muscimol and baclofen ameliorate cognitive deficits and regulate protein expression in the brain of a rat model of streptozocin-induced Alzheimer’s disease. Eur J Pharmacol 2018; 818: 381-99.
[http://dx.doi.org/10.1016/j.ejphar.2017.11.012] [PMID: 29133125]
[190]
Wu L, Zhao H, Weng H, Ma D. Lasting effects of general anesthetics on the brain in the young and elderly: “Mixed picture” of neurotoxicity, neuroprotection and cognitive impairment. J Anesth 2019; 33(2): 321-35.
[http://dx.doi.org/10.1007/s00540-019-02623-7] [PMID: 30859366]
[191]
Li Y, Sun H, Chen Z, Xu H, Bu G, Zheng H. Implications of GABAergic neurotransmission in Alzheimer’s disease. Front Aging Neurosci 2016; 8: 31.
[http://dx.doi.org/10.3389/fnagi.2016.00031] [PMID: 26941642]
[192]
Etherington LA, Mihalik B, Pálvölgyi A, et al. Selective inhibition of extra-synaptic α5-GABAA receptors by S44819, a new therapeutic agent. Neuropharmacology 2017; 125: 353-64.
[http://dx.doi.org/10.1016/j.neuropharm.2017.08.012] [PMID: 28807671]
[193]
Thomas AW, Knust H, Achermann G, et al. The discovery and unique pharmacological profile of RO4938581 and RO4882224 as potent and selective GABAA alpha5 inverse agonists for the treatment of cognitive dysfunction. Bioorg Med Chem Lett 2015; 19: 5940-4.
[194]
Chaudhary A, Maurya PK, Yadav BS, Singh S, Mani A. Current therapeutic targets for Alzheimer’s disease. J Biomed (Syd) 2018; 3: 74-84.
[http://dx.doi.org/10.7150/jbm.26783]
[195]
Vinnakota C, Govindpani K, Tate WP, et al. An 5 GABAA Receptor Inverse Agonist, 5IA, attenuates amyloid beta-induced neuronal death in mouse hippocampal cultures. Int J Mol Sci 2020; 21(9): 1-20.
[http://dx.doi.org/10.3390/ijms21093284] [PMID: 32384683]
[196]
Horton W, Sood A, Peerannawar S, et al. Synthesis and application of β-carbolines as novel multi-functional anti-Alzheimer’s disease agents. Bioorg Med Chem Lett 2017; 27(2): 232-6.
[http://dx.doi.org/10.1016/j.bmcl.2016.11.067] [PMID: 27923619]
[197]
Ishikawa M, Yoshitomi T, Covey DF, Zorumski CF, Izumi Y. Neurosteroids and oxysterols as potential therapeutic agents for glaucoma and Alzheimer’s disease. Neuropsychiatry (London) 2018; 8(1): 344-59.
[http://dx.doi.org/10.4172/Neuropsychiatry.1000356] [PMID: 30774720]
[198]
Touchon J, Ousset PJ, Pasquier F, et al. Treatment with PXT-864 showed stabilisation of cognitive disability in mild Alzheimer’s disease after 36 weeks. Alzheimers Dement 2017; 13(7S_Part_12): 9-10.
[http://dx.doi.org/10.1016/j.jalz.2017.06.653]
[199]
Xu Y, Zhao M, Han Y, Zhang H. GABAergic inhibitory interneuron deficits in Alzheimer’s disease: Implications for treatment. Front Neurosci 2020; 14: 660.
[http://dx.doi.org/10.3389/fnins.2020.00660] [PMID: 32714136]
[200]
Kim GH, Kim JE, Rhie SJ, Yoon S. The role of oxidative stress in neurodegenerative diseases. Exp Neurobiol 2015; 24(4): 325-40.
[http://dx.doi.org/10.5607/en.2015.24.4.325] [PMID: 26713080]
[201]
Badía MC, Giraldo E, Dasí F, et al. Reductive stress in young healthy individuals at risk of Alzheimer disease. Free Radic Biol Med 2013; 63: 274-9.
[http://dx.doi.org/10.1016/j.freeradbiomed.2013.05.003] [PMID: 23665394]
[202]
Swerdlow RH. Brain aging, Alzheimer’s disease, and mitochondria. Biochim Biophys Acta 2011; 1812(12): 1630-9.
[http://dx.doi.org/10.1016/j.bbadis.2011.08.012] [PMID: 21920438]
[203]
Cervellati C, Romani A, Seripa D, et al. Oxidative balance, homocysteine, and uric acid levels in older patients with late onset Alzheimer’s disease or vascular dementia. J Neurol Sci 2014; 337(1-2): 156-61.
[PMID: 24321755]
[204]
Markesbery WR. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic Biol Med 1997; 23(1): 134-47.
[http://dx.doi.org/10.1016/S0891-5849(96)00629-6] [PMID: 9165306]
[205]
Hawking ZL. Review article Alzheimer’s disease  The role of mitochondrial dysfunction and potential new therapies. Biosci Horiz 2016; 9: 1-11.
[http://dx.doi.org/10.1093/biohorizons/hzw014]
[206]
Malm T, Mariani M, Donovan LJ, Neilson L, Landreth GE. Activation of the nuclear receptor PPARδ is neuroprotective in a transgenic mouse model of Alzheimer’s disease through inhibition of inflammation. J Neuroinflammation 2015; 12(1): 7.
[http://dx.doi.org/10.1186/s12974-014-0229-9] [PMID: 25592770]
[207]
Trushina ET, Oxidative Stress E. Synaptic Dysfunction, and Alzheimer’s Disease. J Alzheimers Dis 2016; 1-17.
[208]
Baierle M, Nascimento SN, Moro AM, et al. Relationship between inflammation and oxidative stress and cognitive decline in the institutionalized elderly. Oxid Med Cell Longev 2015; 2015: 804198.
[http://dx.doi.org/10.1155/2015/804198] [PMID: 25874023]
[209]
Paragh G, Balla P, Katona E, Seres I, Egerházi A, Degrell I. Serum paraoxonase activity changes in patients with Alzheimer’s disease and vascular dementia. Eur Arch Psychiatry Clin Neurosci 2002; 252(2): 63-7.
[http://dx.doi.org/10.1007/s004060200013] [PMID: 12111338]
[210]
Commenges D, Scotet V, Renaud S, Jacqmin-Gadda H, Barberger-Gateau P, Dartigues JF. Intake of flavonoids and risk of dementia. Eur J Epidemiol 2000; 16(4): 357-63.
[http://dx.doi.org/10.1023/A:1007614613771] [PMID: 10959944]
[211]
Zhang C, Browne A, Child D, Tanzi RE. Curcumin decreases amyloid-β peptide levels by attenuating the maturation of amyloid-β precursor protein. J Biol Chem 2010; 285(37): 28472-80.
[http://dx.doi.org/10.1074/jbc.M110.133520] [PMID: 20622013]
[212]
Clausen A, Xu X, Bi X, Baudry M. Effects of the superoxide dismutase/catalase mimetic EUK-207 in a mouse model of Alzheimer’s disease: Protection against and interruption of progression of amyloid and tau pathology and cognitive decline. J Alzheimers Dis 2012; 30(1): 183-208.
[http://dx.doi.org/10.3233/JAD-2012-111298] [PMID: 22406441]
[213]
Luo W, Sun W, Taldone T, Rodina A, Chiosis G. Heat shock protein 90 in neurodegenerative diseases. Mol Neurodegener 2010; 5(1): 24.
[http://dx.doi.org/10.1186/1750-1326-5-24] [PMID: 20525284]
[214]
Tan BL, Norhaizan ME, Liew WP. Nutrients and oxidative stress: Friend or foe? Oxid Med Cell Longev 2018; 2018: 9719584.
[http://dx.doi.org/10.1155/2018/9719584] [PMID: 29643982]
[215]
Aisen PS, Schafer KA, Grundman M, et al. Effects of rofecoxib or naproxen vs. placebo on Alzheimer disease progression: A randomized controlled trial. JAMA 2003; 289(21): 2819-26.
[http://dx.doi.org/10.1001/jama.289.21.2819] [PMID: 12783912]
[216]
Tripathi KD. Essentials of Medical Pharmacology. (7th ed.), New Delhi, India: Jaypee Brothers Medical Publisher 2013.
[217]
Esposito Z, Belli L, Toniolo S, Sancesario G, Bianconi C, Martorana A. Amyloid β glutamate, excitotoxicity in Alzheimer’s disease: Are we on the right track? CNS Neurosci Ther 2013; 19(8): 549-55.
[http://dx.doi.org/10.1111/cns.12095] [PMID: 23593992]
[218]
Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer’s disease: An appraisal for the development of therapeutics. Nat Rev Drug Discov 2011; 10(9): 698-712.
[http://dx.doi.org/10.1038/nrd3505] [PMID: 21852788]

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