2,892
Views
27
CrossRef citations to date
0
Altmetric
Editorial

Do BACE inhibitor failures in Alzheimer patients challenge the amyloid hypothesis of the disease?

ORCID Icon, , &
Pages 599-602 | Received 31 Jan 2019, Accepted 17 May 2019, Published online: 27 May 2019

References

  • Panza F, Lozupone M, Logroscino G, et al. A critical appraisal of amyloid-β-targeting therapies for Alzheimer disease. Nat Rev Neurol. 2019;15:73–88.
  • Egan MF, Kost J, Tariot PN, et al. Randomized trial of verubecestat for mild-to-moderate Alzheimer’s disease. N Engl J Med. 2018;378:1691–1703.
  • Cummings J. APECS trial of the BACE1 inhibitor verubecestat for prodromal Alzheimer’s disease. J Prev Alzheimers Dis. 2018;5:S1.
  • Sur C, Kost J, Scott D, et al. BACE inhibition by verubecestat produces a rapid, non-progressive reduction in brain and hippocampal volume in Alzheimer’s disease. J Prev Alzheimers Dis. 2018;5:S18 OC13.
  • Update on Janssen’s BACE Inhibitor Program. Janssen’s press release. [cited 2019 May 5]. Available from: https://www.janssen.com/update-janssens-bace-inhibitor-program
  • Bowman Rogers M Bump in the road or disaster? BACE inhibitors Worsen Cognition. Alzheimer Res Forum. 2018 November;2.
  • Update on Phase III clinical trials of lanabecestat for Alzheimer’s disease. AstraZeneca’s press release. [cited 2019 May 5]. Available from: https://www.astrazeneca.com/media-centre/press-releases/2018/update-on-phaseiii-clinical-trials-of-lanabecestat-for-alzheimers-disease-12062018.html
  • Lo AC, Evans CD, Mancini M, et al. Results from the Phase 2 NAVIGATE-AD clinical trial evaluating LY3202626 BACE inhibitor in patients with mild Alzheimer’s disease dementia. J Prev Alzheimers Dis. 2018;5:S37 LB1.
  • Lynch SY, Kaplow J, Zhao J, et al. Elenbecestat in MCI-to-Moderate Alzheimer’s disease: safety and effectiveness as measured by amyloid PET and the ADCOMS clinical endpoints. J Prev Alzheimers Dis. 2018;5:S11–S12OC5.
  • Zhu K, Xiang X, Filser S, et al. Beta-site amyloid precursor protein cleaving enzyme 1 inhibition impairs synaptic plasticity via Seizure Protein 6. Biol Psychiatry. 2018;83:428–437.
  • Hitt B, Riordan SM, Kukreja L, et al. β-Site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1)-deficient mice exhibit a close homolog of L1 (CHL1) loss-of-function phenotype involving axon guidance defects. J Biol Chem. 2012;287:38408–38425.
  • Hu X, Fan Q, Hou H, et al. Neurological dysfunctions associated with altered BACE1-dependent Neuregulin-1 signaling. J Neurochem. 2016;136:234–249.
  • Vassar R, Kuhn PH, Haass C, et al. Function, therapeutic potential and cell biology of BACE proteases: current status and future prospects. J Neurochem. 2014;130:4–28.
  • Filser S, Ovsepian SV, Masana M, et al. Pharmacological inhibition of BACE1 impairs synaptic plasticity and cognitive functions. Biol Psychiatry. 2015;77:729–739.
  • Doody RS, Raman R, Farlow M, et al. A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med. 2013;369:341–350.
  • Bateman RJ, Siemers ER, Mawuenyega KG, et al. A γ-secretase inhibitor decreases amyloid-β production in the central nervous system. Ann Neurol. 2009;66:48–54.
  • Coric V, Salloway S, van Dyck CH, et al. Targeting prodromal Alzheimer disease with avagacestat: A randomized clinical trial. JAMA Neurol. 2015;72:1324–1333.
  • Coric V, van Dyck CH, Salloway S, et al. Safety and tolerability of the γ-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Arch Neurol. 2012;69:1430–1440.
  • Green RC, Schneider LS, Amato DA, et al. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. Jama. 2009;302:2557–2564.
  • Vandenberghe R, Riviere ME, Caputo A, et al. Active Aβ immunotherapy CAD106 in Alzheimer’s disease: A phase 2b study. Alzheimers Dement. 2016;3:10–22.
  • Schneeberger A, Hendrix S, Mandler M, et al. Results from a Phase II study to assess the clinical and immunological activity of AFFITOPE® AD02 in patients with early Alzheimer’s disease. J Prev Alzheimers Dis. 2015;2:103–114.
  • Salloway S, Sperling R, Keren R, et al. A phase 2 randomized trial of ELND005, scyllo-inositol, in mild to moderate Alzheimer disease. Neurology. 2011;77:1253–1262.
  • Bishop GM, Robinson SR. Physiological roles of amyloid-β and implications for its removal in Alzheimer’s disease. Drugs Aging. 2004;21:621–630.
  • Puzzo D, Gulisano W, Arancio O, et al. The keystone of Alzheimer pathogenesis might be sought in Aβ physiology. Neuroscience. 2015;307:26–36.
  • Kamenetz F, Tomita T, Hsieh H, et al. APP processing and synaptic function. Neuron. 2003;37:925–937.
  • Esteban JA. Living with the enemy: a physiological role for the β-amyloid peptide. Trends Neurosci. 2004;27:1–3.
  • Parihar MS, Brewer GJ. Amyloid-β as a modulator of synaptic plasticity. J Alzheimers Dis. 2010;22:741–763.
  • Puzzo D, Privitera L, Fa’ M, et al. Endogenous amyloid-β is necessary for hippocampal synaptic plasticity and memory. Ann Neurol. 2011;69:819–830.
  • Lawrence JL, Tong M, Alfulaij N, et al. Regulation of presynaptic Ca2+, synaptic plasticity and contextual fear conditioning by a N-terminal β-amyloid fragment. J Neurosci. 2014;34:14210–14218.
  • Palmeri A, Ricciarelli R, Gulisano W, et al. Amyloid-β peptide is needed for cGMP-induced long-term potentiation and memory. J Neurosci. 2017;37:6926–6937.
  • Abramov E, Dolev I, Fogel H, et al. Amyloid-β as a positive endogenous regulator of release probability at hippocampal synapses. Nat Neurosci. 2009;12:1567–1576.
  • Morley JE, Farr SA, Banks WA, et al. A physiological role for amyloid-β protein: enhancement of learning and memory. J Alzheimers Dis. 2010;19:441–449.
  • López-Toledano MA, Shelanski ML. Neurogenic effect of β-amyloid peptide in the development of neural stem cells. J Neurosci. 2004;24:5439–5444.
  • Plant LD, Boyle JP, Smith IF, et al. The production of amyloid β peptide is a critical requirement for the viability of central neurons. J Neurosci. 2003;23:5531–5535.
  • Sun L, Zhou R, Yang G, et al. Analysis of 138 pathogenic mutations in presenilin-1 on the in vitro production of Aβ42 and Aβ40 peptides by γ-secretase. Proc Natl Acad Sci U S A. 2017;114:E476–85.
  • Kepp KP. Alzheimer’s disease due to loss of function: A new synthesis of the available data. Prog Neurobiol. 2016;143:36–60.
  • Lumsden AL, Rogers JT, Majd S, et al. Dysregulation of neuronal iron homeostasis as an alternative unifying effect of mutations causing familial Alzheimer’s disease. Front Neurosci. 2018;12:533.
  • Morris GP, Clark IA, Vissel B. Questions concerning the role of amyloid-β in the definition, aetiology and diagnosis of Alzheimer’s disease. Acta Neuropathol. 2018;136:663–689.
  • Gulisano W, Maugeri D, Baltrons MA, et al. Role of amyloid-β and tau proteins in Alzheimer’s disease: confuting the amyloid cascade. J Alzheimers Dis. 2018;64:S611–31.
  • Haas JG, Lathe R. Microbes and Alzheimer’s disease: new findings call for a paradigm change. Trends Neurosci. 2018;41:570–573.
  • Panza F, Lozupone M, Seripa D, et al. Amyloid-β immunotherapy for alzheimer disease: is it now a long shot? Ann Neurol. 2019;85:303–315.
  • Kivipelto M, Mangialasche F, Ngandu T. Lifestyle interventions to prevent cognitive impairment, dementia and Alzheimer disease. Nat Rev Neurol. 2018;14:653–656.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.