412
Views
13
CrossRef citations to date
0
Altmetric
Perspective

Should drug discovery scientists still embrace the amyloid hypothesis for Alzheimer’s disease or should they be looking elsewhere?

, &
Pages 1241-1251 | Received 02 Apr 2020, Accepted 06 Jul 2020, Published online: 20 Jul 2020

References

  • Brothers HM, Gosztyla ML, Robinson SR. The physiological roles of amyloid-β peptide hint at new ways to treat Alzheimer’s disease. Front Aging Neurosci. 2018;10:118.
  • Biundo F, Del Prete D, Zhang H, et al. A role for tau in learning, memory and synaptic plasticity. Sci Rep. 2018;8:3184.
  • Gomez-Arboledas A, Davila JC, Sanchez-Mejias E, et al. Phagocytic clearance of presynaptic dystrophies by reactive astrocytes in Alzheimer’s disease. Glia. 2018;66:637–653.
  • Apostolova LG, Risacher SL, Duran T, et al. Associations of the top 20 Alzheimer disease risk variants with brain amyloidosis. JAMA Neurol. 2018;75:328–341.
  • Cummings J, Lee G, Ritter A, et al. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Dement. 2019;5:272–293.
  • Jonsson T, Atwal JK, Steinberg S, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;488:96–99.
  • Yamazaki Y, Zhao N, Caulfield TR, et al. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol. 2019;15:501–518.
  • Musiek ES, Holtzman DM. Three dimensions of the amyloid hypothesis: time, space, and “wingmen”. Nat Neurosci. 2015;18:800–806.
  • 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.
  • 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.
  • Mattsson-Carlgren N, Andersson E, Janelidze S, et al. Aβ deposition is associated with increases in soluble and phosphorylated tau that precede a positive tau PET in Alzheimer’s disease. Sci Adv. 2020;6:eaaz2387.
  • Vogel JW, Iturria-Medina Y, Strandberg OT, et al. Spread of pathological tau proteins through communicating neurons in human Alzheimer’s disease. Nat Commun. 2020;11:2612.
  • Gauthier S, Feldman HH, Schneider LS, et al. Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer’s disease: a randomised, controlled, double-blind, parallel-arm, phase 3 trial. Lancet. 2016;388:2873–2884.
  • Wilcock GK, Gauthier S, Frisoni GB, et al. Potential of low dose leuco-methylthioninium bis(hydromethanesulphonate) (LMTM) monotherapy for treatment of mild Alzheimer’s disease: cohort analysis as modified primary outcome in a phase 3 clinical trial. J Alzheimers Dis. 2018;61:435–457.
  • Schelter BO, Shiells H, Baddeley TC, et al. Concentration-dependent activity of hydromethylthionine on cognitive decline and brain atrophy in mild to moderate Alzheimer’s disease. J Alzheimers Dis. 2019;72:931–946.
  • Shiells H, Schelter BO, Bentham P, et al. Concentration-dependent activity of hydromethylthionine on clinical decline and brain atrophy in a randomized controlled trial in behavioral variant frontotemporal dementia. J Alzheimers Dis. 2020;75:501–519.
  • Hoskin JL, Sabbagh MN, Al-Hasan Y, et al. Tau immunotherapies for Alzheimer’s disease. Expert Opin Investig Drugs. 2019;28:545–554.
  • Panza F, Logroscino G. Anti-tau vaccine in Alzheimer’s disease: a tentative step. Lancet Neurol. 2017;16:99–100.
  • Boxer AL, Qureshi I, Ahlijanian M, et al. Safety of the tau-directed monoclonal antibody BIIB092 in progressive supranuclear palsy: a randomised, placebo-controlled, multiple ascending dose phase 1b trial. Lancet Neurol. 2019;18:549–558.
  • Jackson M Biogen’s tau antibody fails in PSP, but Alzheimer’s studies continue. Scrip; 2019 Dec 13.
  • Carroll J AbbVie scraps an anti-tau study, and that may foretell more big trouble for a beleaguered Biogen. Endpoint News; 2019 Jul 26.
  • DeVos SL, Miller RL, Schoch KM, et al. Tau reduction prevents neuronal loss and reverses pathological tau deposition and seeding in mice with tauopathy. Sci Transl Med. 2017;9:pii: eaag0481.
  • Reiman EM, Arboleda-Velasquez JF, Quiroz YT, et al. Exceptionally low likelihood of Alzheimer’s dementia in APOE2 homozygotes from a 5,000-person neuropathological study. Nat Commun. 2020;11:667.
  • Therriault J, Benedet AL, Pascoal TA, et al. Association of apolipoprotein E ε4 with medial temporal tau independent of amyloid-β. JAMA Neurol. 2019 Dec 20. DOI:10.1001/jamaneurol.2019.4421. [Epub ahead of print].
  • Arboleda-Velasquez JF, Lopera F, O’Hare M, et al. Resistance to autosomal dominant Alzheimer’s disease in an APOE3 Christchurch homozygote: a case report. Nat Med. 2019;25:1680–1683.
  • Verghese PB, Castellano JM, Garai K, et al. ApoE influences amyloid-β (Aβ) clearance despite minimal apoE/Aβ association in physiological conditions. Proc Natl Acad Sci USA. 2013;110:E1807–16.
  • Liu CC, Zhao N, Fu Y, et al. ApoE4 accelerates early seeding of amyloid pathology. Neuron. 2017;96:1024–1032.
  • 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;96:1013–1023.
  • Shi Y, Manis M, Long J, et al. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J Exp Med. 2019;216:2546–2561.
  • Kim J, Eltorai AE, Jiang H, et al. Anti-apoE immunotherapy inhibits amyloid accumulation in a transgenic mouse model of Aβ amyloidosis. J Exp Med. 2012;209:2149–2156.
  • Liao F, Li A, Xiong M, et al. Targeting of nonlipidated, aggregated apoE with antibodies inhibits amyloid accumulation. J Clin Invest. 2018;128:2144–2155.
  • Rosenberg JB, Kaplitt MG, De BP, et al. AAVrh.10-mediated APOE2 central nervous system gene therapy for APOE4-associated Alzheimer’s disease. Hum Gene Ther Clin Dev. 2018;29:24–47.
  • Corlier F, Hafzalla G, Faskowitz J, et al. Systemic inflammation as a predictor of brain aging: contributions of physical activity, metabolic risk, and genetic risk. Neuroimage. 2018;172:118–129.
  • Warren KN, Beason-Held LL, Carlson O, et al. Elevated markers of inflammation are associated with longitudinal changes in brain function in older adults. J Gerontol A Biol Sci Med Sci. 2018;73:770–778.
  • Marsland AL, Gianaros PJ, Kuan DC, et al. Brain morphology links systemic inflammation to cognitive function in midlife adults. Brain Behav Immun. 2015;48:195–204.
  • Imbimbo BP. An update on the efficacy of Non-steroidal anti-inflammatory drugs in Alzheimer’s disease. Expert Opin Investig Drugs. 2009;18:1147–1168.
  • Hampel H, Caraci F, Cuello CA. A path toward precision medicine for neuroinflammatory mechanisms in Alzheimer’s disease. Front Immunol. 2020;11:456.
  • Chetty A, Sharda A, Warburton R, et al. A purinergic P2Y6 receptor agonist prodrug modulates airway inflammation, remodeling, and hyperreactivity in a mouse model of asthma. J Asthma Allergy. 2018;11:159–171.
  • Steed PM, Tansey MG, Zalevsky J, et al. Inactivation of TNF signaling by rationally designed dominant-negative TNF variants. Science. 2003;301:1895–1898.
  • Yli-Karjanmaa M, Larsen KS, Fenger CD, et al. TNF deficiency causes alterations in the spatial organization of neurogenic zones and alters the number of microglia and neurons in the cerebral cortex. Brain Behav Immun. 2019;82:279–297.
  • Sama DM, Mohmmad Abdul H, Furman JL, et al. Inhibition of soluble tumor necrosis factor ameliorates synaptic alterations and Ca2+ dysregulation in aged rats. PLoS One. 2012;7:e38170.
  • Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19:477–489.
  • Kelley N, Jeltema D, Duan Y, et al. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20:pii: E3328.
  • Kirkland JL, Tchkonia T, Zhu Y, et al. The clinical potential of senolytic drugs. J Am Geriatr Soc. 2017;65:2297–2301.
  • Wissler Gerdes EO, Zhu Y, Tchkonia T, et al. Discovery, development, and future application of senolytics: theories and predictions. Febs J. 2020 Feb 29;287:2418–2427. [Epub ahead of print].
  • Biber K, Moller T, Boddeke E, et al. Central nervous system myeloid cells as drug targets: current status and translational challenges. Nat Rev Drug Discov. 2016;15:110–124.
  • Brennan FH, Lee JD, Ruitenberg MJ, et al. Therapeutic targeting of complement to modify disease course and improve outcomes in neurological conditions. Semin Immunol. 2016;28:292–308.
  • Imbimbo BP, Ippati S, Ceravolo F, et al. Perspective: is therapeutic plasma exchange a viable option for treating Alzheimer’s disease? Alzheimers Dement;2020. DOI:10.1002/trc2.12004. in press.
  • Middeldorp J, Lehallier B, Villeda SA, et al. Preclinical assessment of young blood plasma for Alzheimer disease. JAMA Neurol. 2016;73:1325–1333.
  • Lopez OL. AMBAR (Alzheimer’s management by albumin replacement) phase IIb/III results. J Prev Alzheimers Dis. 2018;5(Suppl 1):S8.
  • Rubinsztein DC, Marino G, Kroemer G. Autophagy and aging. Cell. 2011;146:682–695.
  • Boyle PA, Yang J, Yu L, et al. Varied effects of age-related neuropathologies on the trajectory of late life cognitive decline. Brain. 2017;140:804–812.
  • Lonskaya I, Hebron ML, Desforges NM, et al. Nilotinib-induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J Mol Med. 2014;92:373–386.
  • Turner RS, Hebron ML, Lawler A, et al. Nilotinib effects on safety, tolerability, and biomarkers in Alzheimer’s disease. Ann Neurol. 2020;88:183–194.
  • Grimm A, Eckert A. Brain aging and neurodegeneration: from a mitochondrial point of view. J Neurochem. 2017;143:418–431.
  • Swerdlow RH. Mitochondria and mitochondrial cascades in Alzheimer’s disease. J Alzheimers Dis. 2018;62:1403–1416.
  • Galasko DR, Peskind E, Clark CM, et al. Antioxidants for Alzheimer disease: a randomized clinical trial with cerebrospinal fluid biomarker measures. Arch Neurol. 2012;69:836–841.
  • Atamna H, Nguyen A, Schultz C, et al. Methylene blue delays cellular senescence and enhances key mitochondrial biochemical pathways. Faseb J. 2008;22:703–712.
  • Atamna H, Mackey J, Dhahbi JM. Mitochondrial pharmacology: electron transport chain bypass as strategies to treat mitochondrial dysfunction. Biofactors. 2012;38:158–166.
  • Congdon EE, Wu JW, Myeku N, et al. Methylthioninium chloride (Methylene Blue) induces autophagy and attenuates tauopathy in vitro and in vivo. Autophagy. 2012;8:609–622.
  • Zhang L, Zhang S, Maezawa I, et al. Modulation of mitochondrial complex I activity averts cognitive decline in multiple animal models of familial Alzheimer’s disease. EBioMedicine. 2015;2:294–305.
  • Reddy PH, Manczak M, Kandimalla R. Mitochondria-targeted small molecule SS31: a potential candidate for the treatment of Alzheimer’s disease. Hum Mol Genet. 2017;26:1483–1496.
  • Sebastiao I, Candeias E, Santos MS, et al. Insulin as a bridge between type 2 diabetes and Alzheimer disease: how anti-diabetics could be a solution for dementia. Front Endocrinol. 2014;5:110.
  • McIntosh EC, Nation DA. Alzheimer’s disease neuroimaging initiative. Importance of treatment status in links between type 2 diabetes and Alzheimer’s disease. Diabetes Care. 2019;42:972–979.
  • Kuehn BM. In Alzheimer research, glucose metabolism moves to center stage. JAMA. 2020 Jan 8. DOI:10.1001/jama.2019.20939. [Epub ahead of print].
  • Toledo JB, Arnold SE, Raible K, et al. Contribution of cerebrovascular disease in autopsy confirmed neurodegenerative disease cases in the National Alzheimer’s coordinating centre. Brain. 2013;136:2697–2706.
  • Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci. 2011;12:723–738.
  • Brunet A, Berger SL. Epigenetics of aging and aging-related disease. J Gerontol A Biol Sci Med Sci. 2014;69(suppl 1):S17–S20.
  • Delgado-Morales R, Agis-Balboa RC, Esteller M, et al. Epigenetic mechanisms during ageing and neurogenesis as novel therapeutic avenues in human brain disorders. Clin Epigenetics. 2017;9:67.
  • Liu X, Jiao B, Shen L. The epigenetics of Alzheimer’s disease: factors and therapeutic implications. Front Genet. 2018;9:579.
  • Maes T, Molinero C, Antonijoan RM, et al. First-in-human phase I results show safety, tolerability and brain penetrance of ORY-2001, an epigenetic drug targeting LSD1 and MAO-B. Alzheimers Dement. 2017;13:P1573–P1574.
  • Morrison JH, Baxter MG. The ageing cortical synapse: hallmarks and implications for cognitive decline. Nat Rev Neurosci. 2012;13:240–250.
  • Scheff SW, Price DA, Schmitt FA, et al. Hippocampal synaptic loss in early Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging. 2006;27:1372–1384.
  • van Deijk AF, Broersen LM, Verkuyl JM, et al. High content analysis of hippocampal neuron-astrocyte co-cultures shows a positive effect of fortasyn connect on neuronal survival and postsynaptic maturation. Front Neurosci. 2017;11:440.
  • Soininen H, Solomon A, Visser PJ, et al. 24-month intervention with a specific multinutrient in people with prodromal Alzheimer’s disease (LipiDiDiet): a randomised, double-blind, controlled trial. Lancet Neurol. 2017;16:965–975.
  • Jiang C, Li G, Huang P, et al. The gut microbiota and Alzheimer’s disease. J Alzheimers Dis. 2017;58:1–15.
  • Vojdani A, Vojdani E, Saidara E, et al. Reaction of amyloid-β peptide antibody with different infectious agents involved in Alzheimer’s disease. J Alzheimers Dis. 2018;63:847–860.
  • Panza F, Lozupone M, Solfrizzi V, et al. Time to test antibacterial therapy in Alzheimer’s disease. Brain. 2019b;142:2905–2929.
  • Morris JC. Editorial: is now the time for combination therapies for Alzheimer disease? J Prev Alzheimers Dis. 2019;6:153–154.
  • Cuadrado-Tejedor M, Ricobaraza AL, Torrijo R, et al. Phenylbutyrate is a multifaceted drug that exerts neuroprotective effects and reverses the Alzheimer s disease like phenotype of a commonly used mouse model. Curr Pharm Des. 2013;19:5076–5084.
  • Dionisio PA, Amaral JD, Ribeiro MF, et al. Amyloid β pathology is attenuated by tauroursodeoxycholic acid treatment in APP/PS1 mice after disease onset. Neurobiol Aging. 2015;36:228–240.
  • Roche provides topline results from investigator-led phase II/III trial with gantenerumab in rare inherited form of Alzheimer’s disease. cited 2020 May 18 Available from: ttps://www.roche.com/media/releases/med-cor-2020-02-10.htm
  • Mattsson N, Cullen N, Andreasson U, et al. Association between longitudinal plasma neurofilament light and neurodegeneration in patients with Alzheimer disease. JAMA Neurol. 2019;76:791–799.
  • Amadoro G, Latina V, Corsetti V, et al. N-terminal tau truncation in the pathogenesis of Alzheimer’s Disease (AD): developing a novel diagnostic and therapeutic approach. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165584.
  • Corsetti V, Borreca A, Latina V, et al. Passive immunotherapy for N-truncated tau ameliorates the cognitive deficits in two mouse Alzheimer’s disease models. Brain Commun. 2020 Apr 6;2. DOI:10.1093/braincomms/fcca039.
  • Vasanthakumar A, Davis JW, Idler K, et al. Harnessing peripheral DNA methylation differences in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) to reveal novel biomarkers of disease. Clin Epigenetics. 2020;12:84.

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.