Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-18T05:26:52.879Z Has data issue: false hasContentIssue false

5 - Repurposed Agents in Alzheimer’s Disease Drug Development

from Section 1 - Advancing Alzheimer’s Disease Therapies in a Collaborative Science Ecosystem

Published online by Cambridge University Press:  03 March 2022

Jeffrey Cummings
Affiliation:
University of Nevada, Las Vegas
Jefferson Kinney
Affiliation:
University of Nevada, Las Vegas
Howard Fillit
Affiliation:
Alzheimer’s Drug Discovery Foundation
Get access

Summary

Drug repositioning refers to the development of a drug for an indication other that in the marketing authorisation and drug repurposing is the use of known drugs for new indications. Both repositioning and repurposing are opportunities to complement traditional drug development and may shorten the time for a drug to reach the patient. This chapter provides a detailed overview of the pharmacological, preclinical, clinical and epidemiological evidence for four drugs or drug classes currently considered as the highest priority candidates for repurposing in Alzheimer’s disease: fasudil, phenserine, antiviral drugs and glucagon-like peptide 1 (GLP-1) analogs. In addition, key considerations on the future of repurposing are provided, including the role of transcriptional approaches and targeting risk genes and growth factors.

Type
Chapter
Information
Alzheimer's Disease Drug Development
Research and Development Ecosystem
, pp. 54 - 61
Publisher: Cambridge University Press
Print publication year: 2022

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Ballard, C, Aarsland, D, Cummings, J, et al. Drug repositioning and repurposing for Alzheimer disease. Nat Rev Neurol 2020; 16: 661–73.CrossRefGoogle ScholarPubMed
Corbett, A, Pickett, J, Burns, A, et al. Drug repositioning for Alzheimer’s disease. Nat Rev Drug Discov 2012; 11: 833–46.CrossRefGoogle ScholarPubMed
Hou, Y, Zhou, L, Yang, QD, et al. Changes in hippocampal synapses and learning-memory abilities in a streptozotocin treated rat model and intervention by using fasudil hydrochloride. Neuroscience 2012; 200: 120–9.Google Scholar
Rush, T, Martinez-Hernandez, J, Dollmeyer, M, et al. Synaptotoxicity in Alzheimer’s disease involved a dysregulation of actin cytoskeleton dynamics through cofilin 1 phosphorylation. J Neurosci 2018; 38: 10349–61.Google Scholar
Yu, JZ, Li, YH, Liu, CY, et al. Multitarget therapeutic effect of fasudil in APP/PS1transgenic mice. CNS Neurol Disord Drug Targets 2017; 16: 199209.Google Scholar
Hamano, T, Shirafuji, N, Yen, SH, et al. Rho-kinase ROCK inhibitors reduce oligomeric tau protein. Neurobiol Aging 2020; 89: 4154.CrossRefGoogle ScholarPubMed
Zhang, X, Ye, P, Wang, D, et al. Involvement of RhoA/ROCK signaling in Aβ-induced chemotaxis, cytotoxicity and inflammatory response of microglial BV2 cells. Cell Mol Neurobiol 2019; 39: 637–50.Google Scholar
Chen, J, Sun, Z, Jin, M, et al. Inhibition of AGEs/RAGE/Rho/ROCK pathway suppresses non-specific neuroinflammation by regulating BV2 microglial M1/M2 polarization through the NF-κB pathway. J Neuroimmunol 2017; 305: 108–14.CrossRefGoogle ScholarPubMed
Elliott, C, Rojo, AI, Ribe, E, et al. A role for APP in Wnt signalling links synapse loss with β-amyloid production. Transl Psychiatry 2018; 8: 179.Google Scholar
Guo, MF, Zhang, HY, Li, YH, et al. Fasudil inhibits the activation of microglia and astrocytes of transgenic Alzheimer’s disease mice via the downregulation of TLR4/Myd88/NF-κB pathway. J Neuroimmunol 2020; 346: 577284.Google Scholar
Vicari, RM, Chaitman, B, Keefe, D, et al.; Fasudil study group. Efficacy and safety of fasudil in patients with stable angina: a double-blind, placebo-controlled, phase 2 trial. J Am Coll Cardiol 2005; 46: 1803–11.CrossRefGoogle Scholar
Kamei, S, Oishi, M, Takasu, T. Evaluation of fasudil hydrochloride treatment for wandering symptoms in cerebrovascular dementia with 31P-magnetic resonance spectroscopy and Xe-computed tomography. Clin Neuropharmacol 1996; 19: 428–38.Google Scholar
Fukumoto, Y, Yamada, N, Matsubara, H, et al. Double-blind, placebo-controlled clinical trial with a rho-kinase inhibitor in pulmonary arterial hypertension. Circ J 2013; 77: 2619–25.CrossRefGoogle ScholarPubMed
Yan, B, Sun, F, Duan, L, et al. Curative effect of fasudil injection combined with nimodipine on Alzheimer disease of elderly patients. J Clin Med Pract 2011; 14: 36.Google Scholar
Winblad, B. Giacobini, E. Frölich, L. et al. Phenserine efficacy in Alzheimer’s disease. J Alzheimers Dis 2010; 22: 1201–8.Google ScholarPubMed
Lahiri, DK, Alley, GM, Tweedie, D, et al. Differential effects of two hexahydropyrroloindole carbamate-based anticholinesterase drugs on the amyloid beta protein pathway involved in Alzheimer’s disease. Neuromol Med 2007; 9: 157–68.CrossRefGoogle ScholarPubMed
Tabrez, S, Damanhouri, GA. Computational and kinetic studies of acetylcholine esterase inhibition by phenserine. Curr Pharm Des 2019; 25: 2108–12.Google Scholar
Lilja, AM, Röjdner, J, Mustafiz, T, et al. Age-dependent neuroplasticity mechanisms in Alzheimer Tg2576 mice following modulation of brain amyloid-β levels. PLoS One 2013; 8: e58752.CrossRefGoogle ScholarPubMed
Lilja, AM, Luo, Y, Yu, QS, et al. Neurotrophic and neuroprotective actions of (−)- and (+)-phenserine, candidate drugs for Alzheimer’s disease. PLoS One 2013; 8: e54887.Google Scholar
Sugaya, K, Kwak, YD, Ohmitsu, O, et al. Practical issues in stem cell therapy for Alzheimer’s disease. Curr Alzheimer Res 2007; 4: 370–7.CrossRefGoogle ScholarPubMed
Greig, NH, Sambamurti, K, Yu, QS, et al. An overview of phenserine tartrate, a novel acetylcholinesterase inhibitor for the treatment of Alzheimer’s disease. Curr Alzheimer Res 2005; 2: 281–90.Google Scholar
Schneider, LS, Lahiri, DK. The perils of Alzheimer’s drug development. Curr Alzheimer Res 2009; 6: 77–8.Google Scholar
Powell-Doherty, RD, Abbott, ARN, Nelson, LA, Bertke, AS. Amyloid-β and p-tau anti-threat response to herpes simplex virus 1 infection in primary adult murine hippocampal neurons. J Virol 2020; 94: e01874–19.Google Scholar
Wozniak, MA, Frost, AL, Preston, CM, Itzhaki, RF. Antivirals reduce the formation of key Alzheimer’s disease molecules in cell cultures acutely infected with herpes simplex virus type 1. PLoS One 2011; 6: e25152.Google Scholar
Tzeng, NS, Chung, CH, Lin, FH, et al. Anti-herpetic medications and reduced risk of dementia in patients with herpes simplex virus infections: a nationwide, population based cohort study in Taiwan. Neurotherapeutics 2008; 15: 417–29.Google Scholar
Chen, VC, Wu, SI, Huang, KY, et al. Herpes zoster and dementia: a nationwide population-based cohort study. J Clin Psychiatry 2018; 79: 16m11312;DOI: http://doi.org/10.4088/JCP.16m11312.Google Scholar
Bae, S, Yun, SC, Kim, MC, et al. Association of herpes zoster with dementia and effect of antiviral therapy on dementia: a population-based cohort study. Eur Arch Psychiatry Clin Neurosci 2020;DOI: http://doi.org/10.1007/s00406-020-01157-4.CrossRefGoogle Scholar
Xu, W, Yang, Y, Yuan, G, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, reduces Alzheimer disease-associated tau hyperphosphorylation in the hippocampus of rats with type 2 diabetes. J Investig Med 2015; 63: 267–72.Google Scholar
Perry, T, Lahiri, DK, Sambamurti, K, et al. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. J Neurosci Res 2003; 72: 603–12.Google Scholar
Takach, O, Gill, TB, Silverman, MA. Modulation of insulin signaling rescues BDNF transport defects independent of tau in amyloid-β oligomer-treated hippocampal neurons. Neurobiol Aging 2015; 36: 1378–82.CrossRefGoogle ScholarPubMed
Wang, X, Wang, L, Xu, Y, et al. Intranasal administration of exendin-4 antagonizes Aβ31-35-induced disruption of circadian rhythm and impairment of learning and memory. Aging Clin Exp Res 2016; 28: 1259–66.Google Scholar
Solmaz, V, Çınar, BP, Yiğittürk, G, et al. Exenatide reduces TNF-α expression and improves hippocampal neuron numbers and memory in streptozotocin treated rats. Eur J Pharmacol 2015; 765: 482–7.Google Scholar
Bomba, M, Ciavardelli, D, Silvestri, E, et al. Exenatide promotes cognitive enhancement and positive brain metabolic changes in PS1-KI mice but has no effects in 3×Tg-AD animals. Cell Death Dis 2013; 4: e612.CrossRefGoogle Scholar
Long-Smith, CM, Manning, S, McClean, PL, et al. The diabetes drug liraglutide ameliorates aberrant insulin receptor localisation and signalling in parallel with decreasing both amyloid-β plaque and glial pathology in a mouse model of Alzheimer’s disease. Neuromol Med 2013; 15: 102–14.Google Scholar
Qi, L, Ke, L, Liu, X, et al. Subcutaneous administration of liraglutide ameliorates learning and memory impairment by modulating tau hyperphosphorylation via the glycogen synthase kinase-3β pathway in an amyloid β protein induced Alzheimer disease mouse model. Eur J Pharmacol 2016; 15: 2332.CrossRefGoogle Scholar
Gejl, M, Brock, B, Egefjord, L, et al. Blood–brain glucose transfer in Alzheimer’s disease: effect of GLP-1 analog treatment. Sci Rep 2017; 7: 17490.CrossRefGoogle ScholarPubMed
Edison, P, Femminella, G, Holmes, C, et al. Evaluation of liraglutide in treatment for Alzheimer’s disease. Clinical Trials in Alzheimer’s Disease (CTAD) Congress, November 4–7, 2020.Google Scholar
Mullins, RJ, Mustapic, M, Chia, CW, et al. A pilot study of exenatide actions in Alzheimer’s disease. Curr Alzheimer Res 2019; 16: 741–52.Google Scholar
Gerstein, HC, Colhoun, HM, Dagenais, GR, et al Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019; 394: 121–30.Google ScholarPubMed
Ballard, C, Nørgaard, CH, Friedrich, S, et al. Liraglutide and semaglutide: pooled post-hoc analysis to evaluate risk of dementia in patients with type 2 diabetes. Alzheimer’s Association International Conference, 2020.CrossRefGoogle Scholar
Lamb, J, Crawford, ED, Peck, D, et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 2006; 313: 1929–35.Google Scholar
Subramanian, A, Narayan, R, Corsello, SM, et al. A next generation connectivity map: L1000 platform and the first 1,000,000 profiles. Cell 2017; 171: 1437–52.Google Scholar
Williams, G. SPIEDw: a searchable platform-independent expression database web tool. BMC Genomics 2013; 14: 765.CrossRefGoogle ScholarPubMed
Williams, G, Gatt, A, Clarke, E, et al. Drug repurposing for Alzheimer’s disease based on transcriptional profiling of human iPSC-derived cortical neurons. Transl Psychiatry 2019; 9: 220.CrossRefGoogle ScholarPubMed
Bertram, L, Tanzi, RE. Alzheimer disease risk genes: 29 and counting. Nat Rev Neurol 2019; 15: 191–2.Google Scholar
Rothstein, JD, Patel, S, Regan, MR, et al. Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature 2005; 433: 73–7.Google Scholar
Cudkowicz, ME, Titus, S, Kearney, M, et al. Safety and efficacy of ceftriaxone for amyotrophic lateral sclerosis: a multi-stage, randomised, double-blind, placebo-controlled trial. Lancet Neurol 2014; 13: 1083–91.Google Scholar
Singleton, AB, Farrer, M, Johnson, J, et al. Alpha-synuclein locus triplication causes Parkinson’s disease. Science 2003; 302: 841.CrossRefGoogle ScholarPubMed
Mittal, S, Bjørnevik, K, Im, DS, et al. β2-Adrenoreceptor is a regulator of the α-synuclein gene driving risk of Parkinson’s disease. Science 2017; 357: 891–8.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×