Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Drug repositioning and repurposing for Alzheimer disease

Abstract

Drug repositioning and repurposing can enhance traditional drug development efforts and could accelerate the identification of new treatments for individuals with Alzheimer disease (AD) dementia and mild cognitive impairment. Transcriptional profiling offers a new and highly efficient approach to the identification of novel candidates for repositioning and repurposing. In the future, novel AD transcriptional signatures from cells isolated at early stages of disease, or from human neurons or microglia that carry mutations that increase the risk of AD, might be used as probes to identify additional candidate drugs. Phase II trials assessing repurposed agents must consider the best target population for a specific candidate therapy as well as the mechanism of action of the treatment. In this Review, we highlight promising compounds to prioritize for clinical trials in individuals with AD, and discuss the value of Delphi consensus methodology and evidence-based reviews to inform this prioritization process. We also describe emerging work, focusing on the potential value of transcript signatures as a cost-effective approach to the identification of novel candidates for repositioning.

Key points

  • Drug repositioning and repurposing offers a valuable alternative route for the identification of effective disease-modifying treatments for Alzheimer disease (AD).

  • The Delphi method can be used to bring together the opinion of multiple experts to suggest candidates for repurposing.

  • An expert Delphi consensus published in 2012 prioritized five compounds for repurposing as treatments for AD, of which glucagon-like peptide analogues remain high priority candidates.

  • A Delphi consensus involving the authors of this Review was conducted in 2018–2019 and identified the ROCK inhibitor fasudil, the cholinesterase inhibitor phenserine and antiviral treatments such as valaciclovir as high priority candidates for trials in individuals with AD.

  • The prioritization of these compounds was supported by strong packages of preclinical data, most of which include evidence from a number of different preclinical models.

  • Transcriptional screening approaches offer a novel means of identifying potential treatment candidates by targeting AD-associated transcriptional profiles.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Similar content being viewed by others

References

  1. Alzheimer’s Disease International. World Alzheimer Report 2016 https://www.alz.co.uk/research/world-report-2016 (2016).

  2. Petersen, R. C. Mild cognitive impairment. Continuum 22, 404–418 (2016).

    PubMed  Google Scholar 

  3. Jack, C. R. et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 12, 207–216 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Khan, A. et al. Emerging treatments for Alzheimer’s disease for non-amyloid and non-tau targets. Expert. Rev. Neurother. 17, 683–695 (2017).

    CAS  PubMed  Google Scholar 

  5. Morris, G. P., Clark, I. A. & Vissel, B. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer’s disease. Acta Neuropathol. Commun. 2, 135 (2014).

    PubMed  PubMed Central  Google Scholar 

  6. Honig, L. S. et al. Trial of solanezumab for mild dementia due to Alzheimer’s disease. N. Engl. J. Med. 378, 321–330 (2018).

    CAS  PubMed  Google Scholar 

  7. Egan, M. F. et al. Randomized trial of verubecestat for prodromal Alzheimer’s disease. N. Engl. J. Med. 380, 1408–1420 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Ballard, C. & Murphy, C. Clinical trial pipelines for Alzheimer’s disease pose challenges for future effective treatment and therapies [abstract P4-017]. Alzheimers Dement. 14, P1439 (2018).

    Google Scholar 

  9. Corbett, A. et al. Drug repositioning for Alzheimer’s disease. Nat. Rev. Drug Discov. 11, 833–846 (2012).

    CAS  PubMed  Google Scholar 

  10. Hawkes, N. Pfizer abandons research into Alzheimer’s and Parkinson’s diseases. BMJ 360, k122 (2018).

    PubMed  Google Scholar 

  11. Alzheimer’s Disease Neuroimaging Initiative. More ADNI results 2: it’s all about power AlzForum https://www.alzforum.org/news/research-news/more-adni-results-2-its-all-about-power (2008).

  12. Biogen. Aducanumab update. Investors.biogen.com https://investors.biogen.com/static-files/5a31a1e3-4fbb-4165-921a-f0ccb1d64b65 (2019).

  13. Loera-Valencia, R. et al. Current and emerging avenues for Alzheimer’s disease drug targets. J. Intern. Med. 286, 398–437 (2019).

    CAS  PubMed  Google Scholar 

  14. Langedijk, J. et al. Drug repositioning and repurposing: terminology and definitions in literature. Drug Discov. Today 20, 1027–1034 (2015).

    PubMed  Google Scholar 

  15. Stewart, A. K. Medicine. How thalidomide works against cancer. Science 343, 256–257 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Hubsher, G., Haider, M. & Okun, M. S. Amantadine: the journey from fighting flu to treating Parkinson disease. Neurology 78, 1096–1099 (2012).

    CAS  PubMed  Google Scholar 

  17. Scott, T. J., O’Connor, A. C., Link, A. N. & Beaulieu, T. J. Economic analysis of opportunities to accelerate Alzheimer’s disease research and development. Ann. N. Y. Acad. Sci. 1313, 17–34 (2014).

    PubMed  PubMed Central  Google Scholar 

  18. Judge, A. et al. Protective effect of antirheumatic drugs on dementia in rheumatoid arthritis patients. Alzheimers Dement. 3, 612–621 (2017).

    Google Scholar 

  19. Seyb, K. I. et al. Identification of small molecule inhibitors of β-amyloid cytotoxicity through a cell-based high-throughput screening platform. J. Biomol. Screen. 13, 870–878 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Williams, G. et al. Drug repurposing for Alzheimer’s disease based on transcriptional profiling of human iPSC-derived cortical neurons. Transl Psychiatry. 9, 220 (2019).

    PubMed  PubMed Central  Google Scholar 

  21. Howard, R. et al. Minocycline at 2 different dosages vs placebo for patients with mild Alzheimer disease: a randomized clinical trial. JAMA Neurol. 77, 164–174 (2020).

    PubMed  Google Scholar 

  22. Lawlor, B. et al. NILVAD Study Group. Nilvadipine in mild to moderate Alzheimer disease: a randomised controlled trial. PLoS Med. 15, e1002660 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Kehoe, P. G. et al. Results of the reducing pathology in Alzheimer’s disease through angiotensin targeting (RADAR) trial [abstract LB3]. J. Prev. Alzheimers Dis. 6, S31–S32 (2019).

    Google Scholar 

  24. McKhann, G. et al. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 34, 939–944 (1984).

    CAS  PubMed  Google Scholar 

  25. Birks, J. S. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Databast Syst. Rev. 1, CD005593 (2006).

    Google Scholar 

  26. Howard, R. et al. Determining the minimum clinically important differences for outcomes in the DOMINO trial. Int. J. Geriatr. Psychiatry 26, 812–817 (2011).

    PubMed  Google Scholar 

  27. Kume, K. et al. Effects of telmisartan on cognition and regional cerebral blood flow in hypertensive patients with Alzheimer’s disease. Geriatr. Gerontol. Int. 12, 207–214 (2012).

    PubMed  Google Scholar 

  28. Kehoe, P. G., Miners, J. S. & Love, S. Angiotensins in Alzheimer’s disease — friend or foe? Trends Neurosci. 32, 619–628 (1997).

    Google Scholar 

  29. Kehoe, P. G. & Passmore, P. A. The renin–angiotensin system and antihypertensive drugs in Alzheimer’s disease: current standing of the angiotensin hypothesis? J. Alzheimers Dis. 30, S251–S269 (2012).

    PubMed  Google Scholar 

  30. Wright, J. W. & Harding, J. W. Brain renin-angiotensin — a new look at an old system. Prog. Neurobiol. 95, 49–67 (2011).

    CAS  PubMed  Google Scholar 

  31. Culman, J., Blume, A., Gohlke, P. & Unger, T. The renin-angiotensin system in the brain: possible therapeutic implications for AT1-receptor blockers. J. Hum. Hypertens. 16, S64–S70 (2002).

    CAS  PubMed  Google Scholar 

  32. Wang, J. et al. Valsartan lowers brain β-amyloid protein levels and improves spatial learning in a mouse model of Alzheimer disease. J. Clin. Invest. 117, 3393–3402 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Mogi, M. et al. Telmisartan prevented cognitive decline partly due to PPAR-γ activation. Biochem. Biophys. Res. Commun. 375, 446–449 (2008).

    CAS  PubMed  Google Scholar 

  34. Papadopoulos, P., Tong, X. K., Imboden, H. & Hamel, E. Losartan improves cerebrovascular function in a mouse model of Alzheimer’s disease with combined overproduction of amyloid-β and transforming growth factor-β1. J. Cereb. Blood Flow. Metab. 37, 1959–1970 (2017).

    CAS  PubMed  Google Scholar 

  35. Royea, J., Zhang, L., Tong, X. K. & Hamel, E. Angiotensin IV receptors mediate the cognitive and cerebrovascular benefits of losartan in a mouse model of Alzheimer’s disease. J. Neurosci. 37, 5562–5573 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Ongali, B. et al. Angiotensin II type 1 receptor blocker losartan prevents and rescues cerebrovascular, neuropathological and cognitive deficits in an Alzheimer’s disease model. Neurobiol. Dis. 68, 126–136 (2014).

    CAS  PubMed  Google Scholar 

  37. Danielyan, L. et al. Protective effects of intranasal losartan in the APP/PS1 transgenic mouse model of Alzheimer disease. Rejuvenation Res. 13, 195–201 (2010).

    CAS  PubMed  Google Scholar 

  38. Liu, X. et al. Losartan-induced hypotension leads to tau hyperphosphorylation and memory deficit. J. Alzheimers Dis. 40, 419–427 (2014).

    CAS  PubMed  Google Scholar 

  39. Tian, M., Zhu, D., Xie, W. & Shi, J. Central angiotensin II-induced Alzheimer-like tau phosphorylation in normal rat brains. FEBS Lett. 586, 3737–3745 (2012).

    CAS  PubMed  Google Scholar 

  40. Zhu, D. et al. Central angiotensin II stimulation promotes β amyloid production in Sprague Dawley rats. PLoS ONE 6, e16037 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Anderson, C. et al. Renin-angiotensin system blockade and cognitive function in patients at high risk of cardiovascular disease: analysis of data from the ONTARGET and TRANSCEND studies. Lancet Neurol. 10, 43–53 (2011).

    CAS  PubMed  Google Scholar 

  42. Lithell, H. et al. The study on cognition and prognosis in the elderly (SCOPE): principal results of a randomized double-blind intervention trial. J. Hypertens. 21, 875–886 (2003).

    CAS  PubMed  Google Scholar 

  43. Livingston, G. et al. Dementia: prevention, intervention, and care. Lancet 390, 2673–2734 (2017).

    PubMed  Google Scholar 

  44. Williamson, J. D. et al. Effect of intensive vs standard blood pressure control on probable dementia: a randomized clinical trial. JAMA 321, 553–561 (2019).

    PubMed  PubMed Central  Google Scholar 

  45. US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/ct2/show/NCT02471833 (2019).

  46. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02085265 (2020).

  47. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02646982 (2020).

  48. Schrijvers, E. M. C. et al. Insulin metabolism and the risk of Alzheimer disease — the Rotterdam study. Neurology 75, 1982–1987 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Perry, T. et al. Glucagon-like peptide-1 decreases endogenous amyloid-β peptide (Aβ) levels and protects hippocampal neurons from death induced by Aβ and iron. J. Neurosci. Res. 72, 603–612 (2003).

    CAS  PubMed  Google Scholar 

  50. Holscher, C. in Vitamins and Hormones: Incretins and Insulin Secretion (ed. Litwack, G.) 331–354 (Academic Press, 2010).

  51. Li, L. et al. (Val8) glucagon-like peptide-1 prevents tau hyperphosphorylation, impairment of spatial learning and ultra-structural cellular damage induced by streptozotocin in rat brains. Eur. J. Pharmacol. 674, 280–286 (2012).

    CAS  PubMed  Google Scholar 

  52. Perry, T. et al. A novel neutrotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells. J. Pharmacol. Exp. Ther. 300, 958–966 (2002).

    CAS  PubMed  Google Scholar 

  53. Wang, X. H. et al. Val8-glucagon-like peptide-1 protects against Aβ1-40-induced impairment of hippocampal late-phase long-term potentiation and spatial learning in rats. Neuroscience 170, 1239–1248 (2010).

    CAS  PubMed  Google Scholar 

  54. Kastin, A. J., Akerstrom, V. & Pan, W. Interactions of glucagon-like peptide-1 (GLP-1) with the blood–brain barrier. J. Mol. Neurosci. 18, 7–14 (2002).

    CAS  PubMed  Google Scholar 

  55. McClean, P. L., Parthsarathy, V., Faivre, E. & Holscher, C. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J. Neurosci. 31, 6587–6594 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Gengler, S., McClean, P. L., McCurtin, R., Gault, V. A. & Hölscher, C. Val(8)GLP-1 rescues synaptic plasticity and reduces dense core plaques in APP/PS1 mice. Neurobiol. Aging 33, 265–276 (2012).

    CAS  PubMed  Google Scholar 

  57. Hamilton, A., Patterson, S., Porter, D., Gault, V. A. & Holscher, C. Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain. J. Neurosci. Res. 89, 481–489 (2011).

    CAS  PubMed  Google Scholar 

  58. McClean, P. L., Jalewa, J. & Hölscher, C. Prophylactic liraglutide treatment prevents amyloid plaque deposition, chronic inflammation and memory impairment in APP/PS1 mice. Behav. Brain Res. 293, 96–106 (2015).

    CAS  PubMed  Google Scholar 

  59. Qi, L. 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. 783, 23–32 (2016).

    CAS  PubMed  Google Scholar 

  60. Hansen, H. H. et al. The GLP-1 receptor agonist liraglutide improves memory function and increases hippocampal CA1 neuronal numbers in a senescence-accelerated mouse model of Alzheimer’s disease. J. Alzheimers Dis. 46, 877–888 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Xiong, H. et al. The neuroprotection of liraglutide on Alzheimer-like learning and memory impairment by modulating the hyperphosphorylation of tau and neurofilament proteins and insulin signaling pathways in mice. J. Alzheimers Dis. 37, 623–635 (2013).

    PubMed  Google Scholar 

  62. Cukierman-Yaffe, T. et al. Effect of dulaglutide on cognitive impairment in type 2 diabetes: an exploratory analysis of the REWIND trial. Lancet Neurol. 19, 582–590 (2020).

    CAS  PubMed  Google Scholar 

  63. Gejl, M. et al. Blood-brain glucose transfer in Alzheimer’s disease: effect of GLP-1 analog treatment. Sci. Rep. 7, 17490 (2017).

    PubMed  PubMed Central  Google Scholar 

  64. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01843075 (2019).

  65. Mullins, R. J. et al. A pilot study of exenatide actions in Alzheimer’s disease. Curr. Alzheimer Res. 16, 741–752 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Ono-Saito, N., Niki, I. & Hidaka, H. H-series protein kinase inhibitors and potential clinical applications. Pharmacol. Ther. 82, 123–131 (1999).

    CAS  PubMed  Google Scholar 

  67. Sellers, K. J. et al. Amyloid β synaptotoxicity is Wnt-PCP dependent and blocked by fasudil. Alzheimers Dement. 14, 306–317 (2018).

    PubMed  PubMed Central  Google Scholar 

  68. Huentelman, M. J. et al. Peripheral delivery of a ROCK inhibitor improves learning and working memory. Behav. Neurosci. 123, 218–223 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Couch, B. A., DeMarco, G. J., Gourley, S. L. & Koleske, A. J. Increased dendrite branching in AβPP/PS1 mice and elongation of dendrite arbors by fasudil administration. J. Alzheimers Dis. 20, 1003–1008 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Song, Y., Chen, X., Wang, L. Y., Gao, W. & Zhu, M. J. Rho kinase inhibitor fasudil protects against β-amyloid-induced hippocampal neurodegeneration in rats. CNS Neurosci. Ther. 19, 603–610 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Killick, R. et al. Clusterin regulates β-amyloid toxicity via Dickkopf-1-driven induction of the wnt-PCP-JNK pathway. Mol. Psychiatry 19, 88–98 (2014).

    CAS  PubMed  Google Scholar 

  72. Elliott, C. et al. A role for APP in Wnt signalling links synapse loss with β-amyloid production. Transl Psychiatry 8, 179 (2018).

    PubMed  PubMed Central  Google Scholar 

  73. Killick, R., Ballard, C., Aarsland, D. & Sultana J. Fasudil for the treatment of Alzheimer’s disease. Preprint at OSF https://osf.io/3e4y6 (2020).

  74. Fukumoto, Y. et al. Double-blind, placebo-controlled clinical trial with a rho-kinase inhibitor in pulmonary arterial hypertension. Circ. J. 77, 2619–2625 (2013).

    CAS  PubMed  Google Scholar 

  75. Yan, B. et al. Curative effect of fasudil injection combined with nimodipine on Alzheimer disease of elderly patients. J. Clin. Med. Pract. 14, 36 (2011).

    Google Scholar 

  76. Yu, J. Z. et al. Multitarget therapeutic effect of fasudil in APP/PS1 transgenic mice. CNS Neurol. Disord. Drug Targets 16, 199–209 (2017).

    CAS  PubMed  Google Scholar 

  77. Hou, Y. et al. Changes in hippocampal synapses and learning-memory abilities in a streptozotocin-treated rat model and intervention by using fasudil hydrochloride. Neuroscience 200, 120–129 (2012).

    CAS  PubMed  Google Scholar 

  78. Winblad, B. et al. Phenserine efficacy in Alzheimer’s disease. J. Alzheimers Dis. 22, 1201–1208 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Becker, R. E. et al. Phenserine and inhibiting pre-programmed cell death: in pursuit of a novel intervention for Alzheimer’s disease. Curr. Alzheimer Res. 15, 883–891 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Chang, C.-F. et al. (−)-Phenserine inhibits neuronal apoptosis following ischemia/reperfusion injury. Brain Res. 1677, 118–128 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Tweedie, D. et al. Cognitive impairments induced by concussive mild traumatic brain injury in mouse are ameliorated by treatment with phenserine via multiple non-cholinergic and cholinergic mechanisms. PLoS ONE 11, e0156493 (2016).

    PubMed  PubMed Central  Google Scholar 

  82. Lecca, D. et al. (−)-Phenserine and the prevention of pre-programmed cell death and neuroinflammation in mild traumatic brain injury and Alzheimer’s disease challenged mice. Neurobiol. Dis. 130, 104528 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Sugaya, K. et al. Practical issues in stem cell therapy for Alzheimer’s disease. Curr. Alzheimer Res. 4, 370–377 (2007).

    CAS  PubMed  Google Scholar 

  84. Marutle, A. et al. Modulation of human neural stem cell differentiation in Alzheimer (APP23) transgenic mice by phenserine. Proc. Natl Acad. Sci. USA 104, 12506–12511 (2007).

    CAS  PubMed  Google Scholar 

  85. Lahiri, D. K. et al. Differential effects of two hexahydropyrroloindole carbamate-based anticholinesterase drugs on the amyloid beta protein pathway involved in Alzheimer’s disease. Neuromolecular Med. 9, 157–168 (2007).

    CAS  PubMed  Google Scholar 

  86. Lahiri, D. K. et al. The experimental Alzheimer’s disease drug posiphen [(+)-phenserine] lowers amyloid-β peptide levels in cell culture and mice. J. Pharmacol. Exp. Ther. 320, 386–396 (2007).

    CAS  PubMed  Google Scholar 

  87. Haroutunian, V. et al. Pharmacological modulation of Alzheimer’s β-amyloid precursor protein levels in the CSF of rats with forebrain cholinergic system lesions. Brain Res. Mol. Brain Res. 46, 161–168 (1997).

    CAS  PubMed  Google Scholar 

  88. Patel, N. et al. Phenserine, a novel acetylcholinesterase inhibitor, attenuates impaired learning of rats in a 14-unit T-maze induced by blockade of the N-methyl-D-aspartate receptor. Neuroreport 9, 171–176 (1998).

    CAS  PubMed  Google Scholar 

  89. Kadir, A. et al. Effect of phenserine treatment on brain functional activity and amyloid in Alzheimer’s disease. Ann. Neurol. 63, 621–631 (2008).

    CAS  PubMed  Google Scholar 

  90. Greig, N. H. et al. An overview of phenserine tartrate, a novel acetylcholinesterase inhibitor for the treatment of Alzheimer’s disease. Curr. Alzheimer Res. 2, 281–290 (2005).

    CAS  PubMed  Google Scholar 

  91. Birks, J. S. & Harvey, R. J. Donepezil for dementia due to Alzheimer’s disease. Cochrane Database Syst. Rev. 6, CD001190 (2018).

    PubMed  Google Scholar 

  92. Axonyx. Axonyx announces results of curtailed phase III clinical trials for phenserine in Alzheimer’s disease. Businesswire https://www.businesswire.com/news/home/20050920005633/en/Axonyx-Announces-Results-Curtailed-Phase-III-Clinical (2005).

  93. Schneider, L. S. & Lahiri, D. K. The perils of Alzheimer’s drug development. Curr. Alzheimer Res. 6, 77–78 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Jamieson, G. A., Maitland, N. J., Wilcock, G. K., Craske, J. & Itzhaki, R. F. Latent herpes simplex virus type 1 in normal and Alzheimer’s disease brains. J. Med. Virol. 33, 224–227 (1991).

    CAS  PubMed  Google Scholar 

  95. IItzhaki, R. F. et al. Herpes simplex virus type 1 in brain and risk of Alzheimer’s disease. Lancet 349, 241–244 (1997).

    Google Scholar 

  96. Wozniak, M. A., Frost, A. L. & Itzhaki, R. F. Alzheimer’s disease-specific tau phosphorylation is induced by herpes simplex virus type 1. J. Alzheimers Dis. 16, 341–350 (2009).

    CAS  PubMed  Google Scholar 

  97. Santana, S., Recuero, M., Bullido, M. J., Valdivieso, F. & Aldudo, J. Herpes simplex virus type I induces the accumulation of intracellular β-amyloid in autophagic compartments and the inhibition of the non-amyloidogenic pathway in human neuroblastoma cells. Neurobiol. Aging. 33, 19–33 (2012).

    Google Scholar 

  98. Wozniak, M. A., Itzhaki, R. F., Shipley, S. J. & Dobson, C. B. Herpes simplex virus infection causes cellular β-amyloid accumulation and secretase upregulation. Neurosci. Lett. 429, 95–100 (2007).

    CAS  PubMed  Google Scholar 

  99. Piacentini, R. et al. HSV-1 promotes Ca2+-mediated APP phosphorylation and Aβ accumulation in rat cortical neurons. Neurobiol. Aging. 32, 2323.e13–2323.e26 (2011).

    CAS  Google Scholar 

  100. Wozniak, M. A., Mee, A. P. & Itzhaki, R. F. Herpes simplex virus type 1 DNA is located within Alzheimer’s disease amyloid plaques. J. Pathol. 217, 131–138 (2009).

    CAS  PubMed  Google Scholar 

  101. Lerchundi, R. et al. Tau cleavage at D421 by caspase-3 is induced in neurons and astrocytes infected with herpes simplex virus type 1. J. Alzheimers Dis. 23, 513–520 (2011).

    CAS  PubMed  Google Scholar 

  102. Zambrano, A. et al. Neuronal cytoskeletal dynamic modification and neurodegeneration induced by infection with herpes simplex virus type 1. J. Alzheimers Dis. 14, 259–269 (2008).

    CAS  PubMed  Google Scholar 

  103. Wozniak, M. A., Frost, A. L., Preston, C. M. & Itzhaki, R. F. Antivirals reduce the formation of key Alzheimer’s disease molecules in cell cultures acutely infected with herpes simplex virus type 1. PLoS ONE 6, e25152 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Tzeng, N. S. 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 15, 417–429 (2008).

    Google Scholar 

  105. Chen, V. C. et al. Herpes zoster and dementia: a nationwide population-based cohort study. J. Clin. Psychiatry 79, 16m11312 (2018).

    PubMed  Google Scholar 

  106. Devanand, D. P. et al. Antiviral therapy: valacyclovir treatment of Alzheimer’s disease (VALAD) trial: protocol for a randomised, double-blind,placebo-controlled, treatment trial. BMJ Open 10, e032112 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Sultana, J. & Ballard, C. Disease-modifying agents for rheumatoid arthritis: methotrexate, chloroquine phosphate/proguanil hydrochloride, cyclosporine, cyclophosphamide, hydroxychloroquine sulphate, sodium aurothiomalate, and sulfasalazine as potential repurposing treatments for Alzheimer’s Disease; a comprehensive literature review. Preprint at OSF https://mfr.osf.io/render?url=https%3A%2F%2Fosf.io%2F6g57a%2Fdownload (2020).

  108. Van Gool, W. A. et al. Effect of hydroxychloroquine on progression of dementia in early Alzheimer’s disease: an 18-month randomised, double-blind, placebo-controlled study. Lancet 358, 455–460 (2001).

    PubMed  Google Scholar 

  109. Ali, M. R. et al. Tempol and perindopril protect against lipopolysaccharide-induced cognition impairment and amyloidogenesis by modulating brain-derived neurotropic factor, neuroinflammation and oxido-nitrosative stress. Naunyn Schmiedebergs Arch. Pharmacol. 389, 637–656 (2016).

    PubMed  Google Scholar 

  110. de Oliveira, F. et al. Brain penetrating angiotensin converting enzyme inhibitors and cognitive change in patients with dementia due to Alzheimer’s disease. J. Alzheimers Dis. 42, S321–S324 (2014).

    PubMed  Google Scholar 

  111. Wharton, W. et al. The effects of ramipril in individuals at risk for Alzheimer’s disease: results of a pilot clinical trial. J. Alzheimers Dis. 32, 147–156 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Lamb, J. et al. The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935 (2006).

    CAS  PubMed  Google Scholar 

  113. Subramanian, A. et al. A next generation connectivity map: L1000 platform and the first 1,000,000 profiles. Cell. 171, 1437–1452.e17 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Gatt, A. et al. Alzheimer’s disease progression in the 5×FAD mouse captured with a multiplex gene expression array. J. Alzheimer’s Dis. 72, 1177–1191 (2019).

    CAS  Google Scholar 

  115. Hargis, K. E. & Blalock, E. M. Transcriptional signatures of brain aging and Alzheimer’s disease: what are our rodent models telling us? Behav. Brain Res. 322, 311–328 (2017).

    CAS  PubMed  Google Scholar 

  116. Williams, G. A searchable cross-platform gene expression database reveals connections between drug treatments and disease. BMC Genomics 13, 12 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Williams, G. SPIEDw: a searchable platform-independent expression database web tool. BMC Genomics 14, 765 (2013).

    PubMed  PubMed Central  Google Scholar 

  118. Ballard, C. et al. Identifying novel candidates for re-purposing as potential therapeutic agents for AD. Preprint at bioRxiv https://www.biorxiv.org/content/10.1101/622308v1 (2019).

  119. Bertram, L. & Tanzi, R. E. Alzheimer disease risk genes: 29 and counting. Nat. Rev. Neurol. 15, 191–192 (2019).

    PubMed  Google Scholar 

  120. Maragakis, N. J. & Rothstein, J. D. Glutamate transporters in neurologic disease. Arch. Neurol. 58, 365–370 (2001).

    CAS  PubMed  Google Scholar 

  121. Rothstein, J. D. et al. Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann. Neurol. 38, 73–84 (1995).

    CAS  PubMed  Google Scholar 

  122. Rothstein, J. D. et al. β-Lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature 433, 73–77 (2005).

    CAS  PubMed  Google Scholar 

  123. Yimer, E. M., Hishe, H. Z. & Tuem, K. B. Repurposing of the β-lactam antibiotic, ceftriaxone for neurological disorders: a review. Front. Neurosci. 13, 236 (2019).

    PubMed  PubMed Central  Google Scholar 

  124. Cudkowicz, M. E. et al. Safety and efficacy of ceftriaxone for amyotrophic lateral sclerosis: a multi-stage, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 13, 1083–1091 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Singleton, A. B. et al. α-Synuclein locus triplication causes Parkinson’s disease. Science 302, 841 (2003).

    CAS  PubMed  Google Scholar 

  126. Mittal, S. et al. β2-Adrenoreceptor is a regulator of the α-synuclein gene driving risk of Parkinson’s disease. Science 357, 891–898 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Gronich, N. et al. β2-adrenoceptor agonists and antagonists and risk of Parkinson’s disease. Mov. Disord. 33, 1465–1471 (2018).

    CAS  PubMed  Google Scholar 

  128. Searles Nielsen, S. et al. β2-adrenoreceptor medications and risk of Parkinson disease. Ann. Neurol. 84, 683–693 (2018).

    CAS  PubMed  Google Scholar 

  129. Boshoff, E. L., Fletcher, E. J. R. & Duty, S. Fibroblast growth factor 20 is protective towards dopaminergic neurons in vivo in a paracrine manner. Neuropharmacology 137, 156–163 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Niu, J. et al. Efficient treatment of Parkinson’s disease using ultrasonography-guided rhFGF20 proteoliposomes. Drug Deliv. 25, 1560–1569 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Weissmiller, A. M. & Wu, C. Current advances in using neurotrophic factors to treat neurodegenerative disorders. Transl Neurodegener. 1, 14 (2012).

    PubMed  PubMed Central  Google Scholar 

  132. Ohmachi, S. et al. FGF-20, a novel neurotrophic factor, preferentially expressed in the substantia nigra pars compacta of rat brain. Biochem. Biophys. Res. Commun. 277, 355–360 (2000).

    CAS  PubMed  Google Scholar 

  133. Fletcher, E. J. R. et al. Targeted repositioning identifies drugs that increase fibroblast growth factor 20 production and protect against 6-hydroxydopamine-induced nigral cell loss in rats. Sci. Rep. 9, 8336 (2019).

    PubMed  PubMed Central  Google Scholar 

  134. Murdoch, D. & Plosker, G. L. Triflusal: a review of its use in cerebral infarction and myocardial infarction, and as thromboprophylaxis in atrial fibrillation. Drugs 66, 671–692 (2006).

    CAS  PubMed  Google Scholar 

  135. Kim, S. W. et al. Neuroprotective effect of triflusal and its main metabolite, 2-hydroxy-4-trifluoromethylbenzoic acid (HTB), in the postischemic brain. Neurosci. Lett. 643, 59–64 (2017).

    CAS  PubMed  Google Scholar 

  136. Vicari, R. M. et al. Efficacy and safety of fasudil in patients with stable angina: a double-blind, placebo-controlled, phase 2 trial. J. Am. Coll. Cardiol. 46, 1803–1811 (2005).

    CAS  PubMed  Google Scholar 

  137. 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. 19, 428–438 (1996).

    CAS  PubMed  Google Scholar 

  138. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02997982 (2020).

  139. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03282916 (2020).

  140. Nitsch, R. M., Deng, M., Tennis, M., Schoenfeld, D. & Growdon, J. H. The selective muscarinic M1 agonist AF102B decreases levels of total Abeta in cerebrospinal fluid of patients with Alzheimer’s disease. Ann. Neurol. 48, 913–918 (2000).

    CAS  PubMed  Google Scholar 

  141. AbdAlla, S., El Hakim, A., Abdelbaset, A., Elfaramawy, Y. & Quitterer, U. Inhibition of ACE retards tau hyperphosphorylation and signs of neuronal degeneration in aged rats subjected to chronic mild stress. Biomed. Res. Int. 2015, 917156 (2015).

    PubMed  PubMed Central  Google Scholar 

  142. AbdAlla, S., Langer, A., Fu, X. & Quitterer, U. ACE inhibition with captopril retards the development of signs of neurodegeneration in an animal model of Alzheimer’s disease. Int. J. Mol. Sci. 14, 16917–16942 (2013).

    PubMed  PubMed Central  Google Scholar 

  143. Sultana, J. & Ballard, C. ACE inhibitors: captopril, ramipril, lisinopril, perindopril as potential repurposing treatments for Alzheimer’s disease; a comprehensive literature review. Preprint at OSF https://mfr.osf.io/render?url=https%3A%2F%2Fosf.io%2Fz9uc4%2Fdownload (2020).

Download references

Acknowledgements

The authors thank J. Pickett from the Alzheimer’s Society (UK), C. Routledge from Alzheimer Research UK and H. Fillit from the Alzheimer’s Drug Discovery Foundation for their invaluable contributions to the 2018–2019 Delphi consensus. We also thank the Wellcome Trust for supporting much of the work pertaining to transcript signatures presented in the current paper. J.C. is supported by KMA, NIGMS grant P20GM109025, NINDS grant U01NS093334, and NIA grant R01AG053798.

Review criteria

Searches were performed in EMBASE, PsycINFO, MEDLINE and Cochrane databases for papers published after 1960. Search terms were as follows: generic class OR specific drug names OR any known alternative name (obtained from the electronic Medicines Compendium and the British National Formulary) AND dement* OR Alzheim* OR mild cognitive impairmen* OR neuropsych* test* OR cognitive func*.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Clive Ballard.

Ethics declarations

Competing interests

C.B. reports grants and personal fees from Acadia Pharmaceutical Company and H. Lundbeck, as well as personal fees from AARP, Biogen, Eli Lilly, Novo Nordisk, Otsuka, Roche and Synexus, outside the submitted work. D.A. has received research support and/or honoraria from Astra-Zeneca, GE Health, H. Lundbeck and Novartis Pharmaceuticals, and served as a paid consultant for Biogen, Eisai, H. Lundbeck, Heptares and Mentis Cura. J.C. reports grants from National Institute of General Medical Sciences Centers of Biomedical Research Excellence (grant no. P20GM109025), during the conduct of the study; personal fees from Acadia, Actinogen, AgeneBio, Alkahest, Alzheimer Drug Discovery Foundation, Alzheon, Avanir, Axsome, Biogen, Cassava, Cerecin, Cerevel, Cognoptix, Cortexyme, EIP Pharma, Eisai, Foresight, Green Valley, Grifols, Hisun, Karuna, Nutricia, Otsuka, reMYND, Resverlogix, Roche, Samumed, Samus Therapeutics, Third Rock, Signant Health, Sunovion, Suven, and United Neuroscience pharmaceutical and assessment companies; other from ADAMAS, BioAsis, MedAvante, QR Pharma, United Neuroscience, other from Keep Memory Alive; outside the submitted work. J.C. has copyright for the Neuropsychiatric Inventory with royalties paid. J.O’B. reports personal fees from Axon, Eisai, GE Healthcare, Roche and TauRx; grants and personal fees from Avid Technology and Eli Lilly; grants from Alliance Medical and Merck, outside the submitted work. J.L.M. reports personal fees from Alector, Alergan, Eisai, Genentech, Oryzon, Green Valley, H. Lundbeck, Janssen, Eli Lilly, MSD, Novartis and Raman Health; personal fees and other from Biogen, outside the submitted work. All other authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ballard, C., Aarsland, D., Cummings, J. et al. Drug repositioning and repurposing for Alzheimer disease. Nat Rev Neurol 16, 661–673 (2020). https://doi.org/10.1038/s41582-020-0397-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41582-020-0397-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing