1,422
Views
47
CrossRef citations to date
0
Altmetric
Review

Emerging drugs to reduce abnormal β-amyloid protein in Alzheimer’s disease patients

, , , , , , , , , , , , , & show all
Pages 377-391 | Received 24 Jun 2016, Accepted 22 Sep 2016, Published online: 06 Oct 2016

References

  • Alzheimer’s Association. Changing the trajectory of Alzheimer’s disease. cited 2016 Sep 15]. Available from: http://www.alz.org/alzheimers_disease_trajectory.asp
  • Schneider LS, Mangialasche F, Andreasen N, et al. Clinical trials and late-stage drug development for Alzheimer’s disease: an appraisal from 1984 to 2014. J Intern Med. 2014;275(3):251–283.
  • Panza F, Logroscino G, Imbimbo BP, et al. Is there still any hope for amyloid-based immunotherapy for Alzheimer’s disease? Curr Opin Psychiatry. 2014;27(2):128–137.
  • Villemagne VL, Pike KE, Chételat G, et al. Longitudinal assessment of Aβ and cognition in aging and Alzheimer disease. Ann Neurol. 2011;69(1):181–192.
  • Johnson KA, Minoshima S, Bohnen N, et al.; Alzheimer’s Association; Society of Nuclear Medicine and Molecular Imaging; Amyloid Imaging Taskforce. Appropriate use criteria for amyloid PET: a report of the Amyloid imaging task force, the society of nuclear medicine and molecular imaging, and the Alzheimer’s Association. Alzheimers Dement. 2013; 9(1):e1–16.
  • Tan CC, Yu JT, Wang HF, et al. Efficacy and safety of donepezil, galantamine, rivastigmine, and memantine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2014;41(2):615–631.
  • Dubois B, Feldman HH, Jacova C, et al. Research criteria for the diagnosis of Alzheimer’s disease: revising the NINCDS-ADRDA criteria. Lancet Neurol. 2007;6(8):734–746.
  • Dubois B, Feldman HH, Jacova C, et al. Revising the definition of Alzheimer’s disease: a newlexicon. Lancet Neurol. 2010;9(11):1118–1127.
  • Geldmacher DS, Provenzano G, McRae T, et al. Donepezil is associated with delayed nursing home placement in patients with Alzheimer’s disease. J Am Geriatr Soc. 2003;51(7):937–944.
  • Wimo A, Winblad B, Stoffler A, et al. Resource utilisation and cost analysis of memantine in patients with moderate to severe Alzheimer’s disease. Pharmacoeconomics. 2003;21(5):327–340.
  • Lopez OL, Becker JT, Wisniewski S, et al. Cholinesterase inhibitor treatment alters the natural history of Alzheimer’ s disease. J Neurol Neurosurg Psychiatry. 2002;72(3):310–314.
  • Lopez OL, Becker JT, Wahed AS, et al. Long-term effects of the concomitant use of memantine with cholinesterase inhibition in Alzheimer disease. J Neurol Neurosurg Psychiatry. 2009;80(6):600–607.
  • Gasper MC, Ott BR, Lapane KL. Is Donepezil therapy associated with reduced mortality in nursing home residents with dementia? Am J Geriatr Pharmacother. 2005;3(1):1–7.
  • Zhu CW, Livote EE, Scarmeas N, et al. Long-term associations between cholinesterase inhibitors and memantine use and health outcomes among patients with Alzheimer’s disease. Alzheimers Dement. 2013;9(6):733–740.
  • Bartus RT, Dean RL 3rd, Beer B, et al. The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982;217:408–414.
  • Schneider LS. Alzheimer disease pharmacologic treatment and treatment research. Continuum (Minneap Minn). 2013;19:339–357.
  • Alzheimer’s Association. 2015 Alzheimer’s disease facts and figures. Alzheimers Dement. 2015;11(3):332–384.
  • IMS Health. White paper IMS disease insight. Alfinito P. Innovative therapies innovative therapies for Alzheimer’s disease. New treatments will drive significant market. [ cited 2016 Sep 15]. Available from: http://www.pharma-iq.com/pre-clinical-discovery-and-development/white-papers/ims-disease-insights-%E2%80%93-alzheimer%E2%80%99s-disease
  • Qian X, Hamad B, Dias-Lalcaca G. The Alzheimer disease market. Nat Rev Drug Discov. 2015;14(10):675–676.
  • Medivation. Pfizer and Medivation announce results from two phase 3 studies in dimebon (latrepirdine*) Alzheimer’s disease clinical development program. cited 2016 Sep 15]. Available from: http://press.pfizer.com/press-release/pfizer-and-medivation-announce-results-two-phase-3-studies-dimebon-latrepirdine-alzhei
  • Quinn JF, Raman R, Thomas RG, et al. Docosahexaenoic acid supplementation and cognitive decline in Alzheimer disease: a randomized trial. JAMA. 2010;304(17):1903–1911.
  • Sampson EL, Jenagaratnam L, McShane R. Metal protein attenuating compounds for the treatment of Alzheimer’s dementia. Cochrane Database Syst Rev. 2014;2:CD005380.
  • Mohs RC, Shiovitz TM, Tariot PN, et al. Atomoxetine augmentation of cholinesterase inhibitor therapy in patients with Alzheimer disease: 6-month, randomized, double-blind, placebo-controlled, parallel-trial study. Am J Geriatr Psychiatry. 2009;17(9):752–759.
  • Yun H-M, Rhim H. The serotonin-6 receptor as a novel therapeutic target. Exp Neurobiol. 2011;20(4):159–168.
  • Grove RA, Harrington CM, Mahler A, et al. A randomized, double-blind, placebo-controlled, 16-week study of the H3 receptor antagonist, GSK239512 as a monotherapy in subjects with mild-to-moderate Alzheimer’s disease. Curr Alzheimer Res. 2014;11(1):47–58.
  • Walter J, Kaether C, Steiner H, et al. The cell biology of Alzheimer’s disease: uncovering the secrets of secretases. Curr Opin Neurobiol. 2001;11:585–590.
  • Frisardi V, Solfrizzi V, Imbimbo BP, et al. Towards disease-modifying treatment of Alzheimer’s disease: drugs targeting beta-amyloid. Curr Alzheimer Res. 2010;7(1):40–55.
  • Seripa D, Solfrizzi V, Imbimbo BP, et al. Tau-directed approaches for the treatment of Alzheimer’s disease: focus on leuco-methylthioninium. Expert Rev Neurother. 2016;16(3):259–277.
  • Jack CR Jr., Albert MS, Knopman DS, et al. Introduction to the recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):257–262.
  • Jack CR Jr., Knopman DS, Jagust WJ, et al. Tracking pathophysiological processes inAlzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 2013;12(2):207–216.
  • 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(1):4–28.
  • D’Onofrio G, Panza F, Frisardi V, et al. Advances in the identification of γ-secretase inhibitors for the treatment of Alzheimer’s disease. Expert Opin Drug Discov. 2012;7(1):19–37.
  • Lannfelt L, Blennow K, Zetterberg H, et al. Safety, efficacy, and biomarker findings of PBT2 in targeting Abeta as a modifying therapy for Alzheimer’s disease: a phase IIa, double-blind, randomised, placebo-controlled trial. Lancet Neurol. 2008;7(9):779–786.
  • Salloway S, Sperling R, Keren R, et al. A phase 2 randomized trial of ELND005, scylloinositol, in mild to moderate Alzheimer disease. Neurology. 2011;77(13):1253–1262.
  • Huang Y, Mucke L. Alzheimer mechanisms and therapeutic strategies. Cell. 2012;148(6):1204–1222.
  • Bu X-L, Jiao S-S, Lian Y, et al. Perspectives on the tertiary prevention strategy for Alzheimer’s Disease. Curr Alzheimer Res. 2016;13(3):307–316.
  • Panza F, Solfrizzi V, Imbimbo BP, et al. Amyloid-directed monoclonal antibodies for the treatment of Alzheimer’s disease: the point of no return? Expert Opin Biol Ther. 2014;14(10):1465–1476.
  • Wischik CM, Harrington CR, Storey JM. Tau-aggregation inhibitor therapy for Alzheimer’s disease. Biochem Pharmacol. 2014;88(4):529–539.
  • Heneka MT, Carson MJ, El Khoury J, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405.
  • Rogers J, Kirby LC, Hempelman SR, et al. Clinical trial of indomethacin in Alzheimer’s disease. Neurology. 1993;43(8):1609–1611.
  • De Jong D, Jansen R, Hoefnagels W, et al. No effect of one-year treatment with indomethacin on Alzheimer’s disease progression: a randomized controlled trial. PLoS One. 2008;3(1):e1475.
  • Imbimbo BP, Solfrizzi V, Panza F. Are NSAIDs useful to treat Alzheimer’s disease or mild cognitive impairment? Front Aging Neurosci. 2010;2:1–14.
  • Martin BK, Szekely C, Brandt J, et al.; and the ADAPT Research Group. Cognitive function over time in the Alzheimer’s disease anti-inflammatory prevention trial (ADAPT): results of a randomized, controlled trial of naproxen and celecoxib. Arch Neurol. 2008;65(7):896–905.
  • Alzheimer’s Disease Anti-inflammatory Prevention Trial Research Group. Results of a follow-up study to the randomized Alzheimer’s Disease Anti-infl ammatory Prevention Trial (ADAPT). Alzheimers Dement. 2013;9(6):714–723.
  • Hori Y, Takeda S, Cho H, et al. A Food and Drug Administration-approved asthma therapeutic agent impacts amyloid β in the brain in a transgenic model of Alzheimer disease. J Biol Chem. 2015;290(4):1966–1978.
  • Weggen S, Eriksen JL, Das P, et al. A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity. Nature. 2001;414(6860):212–216.
  • In ‘T Veld BA, Launer LJ, Hoes AW, et al. NSAIDs and incident Alzheimer’s disease. The Rotterdam Study. Neurobiol Aging. 1998;19(6):607–611.
  • Vlad SC, Miller DR, Kowall NW, et al. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology. 2008;70(19):1672–1677.
  • 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):5709–5714.
  • Lleó A, Berezovska O, Herl L, et al. Nonsteroidal anti-inflammatory drugs lower Abeta42 and change presenilin 1 conformation. Nat Med. 2004;10(10):1065–1066.
  • Pasqualetti P, Bonomini C, Dal Forno G, et al. A randomized controlled study on effects of ibuprofen on cognitive progression of Alzheimer’s disease. Aging Clin Exp Res. 2009;21(2):102–110.
  • Jaturapatporn D, Isaac MG, McCleery J, et al. Aspirin, steroidal and non-steroidal anti-inflammatory drugs for the treatment of Alzheimer’s disease. Cochrane Database Syst Rev. 2012;2:CD006378.
  • Akaishi T, Morimoto T, Shibao M, et al. Structural requirements for the flavonoid fisetin in inhibiting fibril formation of amyloid beta protein. Neurosci Lett. 2008;444(3):280–285.
  • Ushikubo H, Watanabe S, Tanimoto Y, et al. 3,3ʹ,4ʹ,5,5ʹ-Pentahydroxyflavone is a potent inhibitor of amyloid β fibril formation. Neurosci Lett. 2012;513(1):51–56.
  • Bierhaus A, Humpert PM, Morcos M, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med. 2005;83(11):876–886.
  • Schmidt AM, Hori O, Brett J, et al. Cellular receptors for advanced glycation end products: implications for induction of oxidant stress and cellular dysfunction in the pathogenesis of vascular lesions. Arterioscler Thromb. 1994;14(10):1521–1528.
  • Yan SD, Chen X, Fu J, et al. RAGE and amyloid-beta peptide neurotoxicity in Alzheimer’s disease. Nature. 1996;382(6593):685–691.
  • Luth HJ, Ogunlade V, Kuhla B, et al. Age-and stage-dependent accumulation of advanced glycation end products in intracellular deposits in normal and Alzheimer’s disease brains. Cereb Cortex. 2005;15(2):211–220.
  • Sato T, Shimogaito N, Wu X, et al. Toxic advanced glycation end products (TAGE) theory in Alzheimer’s disease. Am J Alzheimers Dis Other Demen. 2006;21(3):197–208.
  • Lue LF, Yan SD, Stern DM, et al. Preventing activation of receptor for advanced glycation endproducts in Alzheimer’s disease. Curr Drug Targets CNS Neurol Disord. 2005;4(3):249–266.
  • Rocken C, Kientsch-Engel R, Mansfeld S, et al. Advanced glycation end products and receptor for advanced glycation end products in AA amyloidosis. Am J Pathol. 2003;162(4):1213–1220.
  • Sabbagh MN, Agro A, Bell J, et al. PF-04494700, an oral inhibitor of receptor for advanced glycation end products (RAGE), in Alzheimer disease. Alzheimer Dis Assoc Disord. 2011;25(3):206–212.
  • Vassar R, Bennett BD, Babu-Khan S, et al. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 1999;286(5440):735–741.
  • Yan R, Bienkowski MJ, Shuck ME, et al. Membrane-anchored aspartyl protease with Alzheimer’s disease beta-secretase activity. Nature. 1999;402(6761):533–537.
  • Bennett BD, Babu-Khan S, Loeloff R, et al. Expression analysis of BACE2 in brain and peripheral tissues. J Biol Chem. 2000;275(27):20647–20651.
  • Farzan M, Schnitzler CE, Vasilieva N, et al. BACE2, a β-secretase homolog, cleaves at the β site and within the amyloid-β region of the amyloid-b precursor protein. Proc Natl Acad Sci USA. 2000;97(17):9712–9717.
  • Yan R, Vassar R. Targeting the β secretase BACE1 for Alzheimer’s disease therapy. Lancet Neurol. 2014;13(3):319–329.
  • Willem M, Garratt AN, Novak B, et al. Control of peripheral nerve myelination by the beta-secretase BACE1. Science. 2006;314(5799):664–666.
  • Hu X, Hicks CW, He W, et al. Bace1 modulates myelination in the central and peripheral nervous system. Nat Neurosci. 2006;9(12):1520–1525.
  • Vassar R. BACE1 inhibitor drugs in clinical trials for Alzheimer’s disease. Alzheimers Res Ther. 2014;6(9):89.
  • Salzer JL. Axonal regulation of Schwann cell ensheathment and myelination. J Peripher Nerv Syst. 2012;17(suppl 3):14–19.
  • Southan C, Hancock JM. A tale of two drug targets: the evolutionary history of BACE1 and BACE2. Front Genet. 2013;4:293.
  • Esterházy D, Stützer I, Wang H, et al. Bace2 is a β cell-enriched protease that regulates pancreatic β cell function and mass. Cell Metab. 2011;14(3):365–377.
  • Forman M, Palcza J, Tseng J, et al. The novel BACE inhibitor MK-8931 dramatically lowers cerebrospinal fluid Aβ peptides in health subjects following single- and multiple-dose administration. Alzheimers Dement. 2012;8(4):P704.
  • Forman M, Kleijn H, Dockendorf M, et al. The novel BACE inhibitor MK-8931 dramatically lowers CSF beta-amyloid in patients with mild-to-moderate Alzheimer’s disease. Alzheimers Dement. 2013;9(4):P139.
  • Lemere CA. Immunotherapy for Alzheimer’s disease: hoops and hurdles. Mol Neurodegener. 2013;8:36.
  • Bard F, Cannon C, Barbour R, et al. Peripherally administered antibodies against amyloid β-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat Med. 2000;6(8):916–919.
  • Morgan D. Mechanisms of A beta plaque clearance following passive A beta immunization. Neurodegener Dis. 2005;2(5):261–266.
  • Panza F, Frisardi V, Imbimbo BP, et al. Anti-β-amyloid immunotherapy for Alzheimer’s disease: focus on bapineuzumab. Curr Alzheimer Res. 2011;8(8):808–817.
  • Imbimbo BP, Ottonello S, Frisardi V, et al. Solanezumab for the treatment of mild-to-moderate Alzheimer’s disease. Expert Rev Clin Immunol. 2012;8(2):135–149.
  • Tayeb HO, Murray ED, Price BH, et al. Bapineuzumab and solanezumab for Alzheimer’s disease: is the ‘amyloid cascade hypothesis’ still alive? Expert Opin Biol Ther. 2013;13(7):1075–1084.
  • Salloway S, Sperling R, Fox NC, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer’s disease. N Engl J Med. 2014;370(4):322–333.
  • Doody RS, Thomas RG, Farlow M, et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer’s disease. N Engl J Med. 2014;370(4):311–321.
  • Siemers ER, Friedrich S, Dean RA, et al. Safety and changes in plasma and cerebrospinal fluid amyloid-beta after a single administration of an amyloid-beta monoclonal antibody in subjects with Alzheimer disease. Clin Neuropharmacol. 2010;33(2):67–73.
  • Farlow M, Arnold SE, Van Dyck CH, et al. Safety and biomarker effects of solanezumab in patients with Alzheimer’s disease. Alzheimers Dement. 2012;8(4):261–271.
  • Sperling RA, Aisen PS, Beckett LA, et al. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):280–292.
  • Donohue MC, Sperling RA, Salmon DP, et al. Australian imaging, biomarkers, and lifestyle flagship study of ageing, Alzheimer’s disease neuroimaging initiative, Alzheimer’s Disease Cooperative Study. The preclinical Alzheimer cognitive composite: measuring amyloid-related decline. JAMA Neurol. 2014;71(8):961–970.
  • Sperling RA, Rentz DM, Johnson KA, et al. The A4 study: stopping AD before symptoms begin? Sci Transl Med. 2014;6(228):228fs13.
  • Jack CR Jr, Knopman DS, Weigand SD, et al. An operational approach to NIA–AA criteria for preclinical Alzheimer’s disease. Ann Neurol. 2011;71(6):765–775.
  • Panza F, Solfrizzi V, Imbimbo BP, et al. Efficacy and safety studies of gantenerumab in patients with Alzheimer’s disease. Expert Rev Neurother. 2014;14(9):973–986.
  • Bohrmann B, Baumann K, Benz J, et al. Gantenerumab: a novel human anti-Aβ antibody demonstrates sustained cerebral amyloid-β binding and elicits cell-mediated removal of human amyloid-β. J Alzheimers Dis. 2012;28(1):49–69.
  • Ostrowitzki S, Deptula D, Thurfjell L, et al. Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Arch Neurol. 2012;69(2):198–207.
  • Rinne JO, Brooks DJ, Rossor MN, et al. 11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients with Alzheimer’s disease treated with bapineuzumab: a phase 2, double-blind, placebo-controlled, ascending-dose study. Lancet Neurol. 2010;9(4):363–372.
  • Hoffmann-La Roche Media Release. Roche provides update on gantenerumab development programme. [ cited 2016 Sep 15]. http://www.roche.com/media/store/releases/med-cor-2014-12-19b.htm
  • Moreth J, Mavoungou C, Schindowski K. Passive anti-amyloid immunotherapy inAlzheimer’s disease: what are the most promising targets? Immun Ageing. 2013;10(1):18.
  • Sevigny J, Chiao P, Bussière T, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–56.
  • 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(11):1430–1440.
  • Doody RS, Raman R, Siemers E, et al. A phase 2 trial of semagacestat for treatment of Alzheimer’s disease. N Engl J Med. 2013;369(4):341–350.
  • Panza F, Solfrizzi V, Imbimbo BP, et al. Amyloid-based immunotherapy for Alzheimer’s disease in the time of prevention trials: the way forward. Expert Rev Clin Immunol. 2014;10(3):405–419.
  • Mills SM, Mallmann J, Santacruz AM, et al. Preclinical trials in autosomal dominant AD: implementation of the DIAN-TU trial. Rev Neurol (Paris). 2013;169(10):737–743.
  • Reiman EM, Langbaum JB, Fleisher AS, et al. Alzheimer’s Prevention Initiative: a plan to accelerate the evaluation of presymptomatic treatments. J Alzheimers Dis. 2011;26(Suppl 3):321–329.
  • Sperling RA, Jack CR Jr, Aisen PS. Testing the right target and right drug at the right stage. Sci Transl Med. 2011;3(111):111cm33.
  • Solomon A, Mangialasche F, Richard E, et al. Advances in the prevention of Alzheimer’s disease and dementia. J Intern Med. 2014;275(3):229–250.
  • Andrieu S, Coley N, Lovestone S, et al. Prevention of sporadic Alzheimer’s disease: lessons learned from clinical trials and future directions. Lancet Neurol. 2015;14(9):926–944.
  • Wang J, Tan L, Yu J-T. Prevention trials in Alzheimer’s disease: current status and future perspectives. J Alzheimers Dis. 2016;50(4):927–945.
  • Soldan A, Pettigrew C, Cai Q, et al.; BIOCARD Research Team. Hypothetical preclinical alzheimer disease groups and longitudinal cognitive change. JAMA Neurol. 2016;73(6):698–705.
  • Morris GP, Clark IA, Vissel B. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer’s disease. Acta Neuropathol Commun. 2014;2:135.
  • Musiek ES, Holtzman DM. Three dimensions of the amyloid hypothesis: time, space and ‘wingmen’. Nat Neurosci. 2015;18(6):800–806.
  • Bishop GM, Robinson SR. Physiological roles of amyloid-beta and implications for its removal in Alzheimer’s disease. Drugs Aging. 2004;21(10):621–630.
  • Abrahamson EE, Ikonomovic MD, Dixon CE, et al. Simvastatin therapy prevents brain trauma-induced increases in beta-amyloid peptide levels. Ann Neurol. 2009;66(3):407–414.
  • Johnson V, Stewart W, Smith DH. Traumatic brain injury and amyloid-β pathology: a link to Alzheimer’s disease? Nat Rev Neurosci. 2010;11(5):361–370.
  • Dong Y, Zhang G, Zhang B, et al. The common inhalational anesthetic sevoflurane induces apoptosis and increases beta-amyloid protein levels. Arch Neurol. 2009;66(5):620–631.
  • Perucho J1, Rubio I, Casarejos MJ, et al. Anesthesia with isoflurane increases amyloid pathology in mice models of Alzheimer’s disease. J Alzheimers Dis. 2010;19(4):1245–1257.
  • Dal Bianco A, Bradl M, Frischer J, et al. Multiple sclerosis and Alzheimer’s disease. Ann Neurol. 2008;63(2):174–183.
  • Steinacker P, Fang L, Kuhle J, et al. Soluble beta-amyloid precursor protein is related to disease progression in amyotrophic lateral sclerosis. PLoS One. 2011;6(8):e23600.
  • Gudmundsson P, Skoog I, Waern M, et al. The relationship between cerebrospinal fluid biomarkers and depression in elderly women. Am J Geriatr Psychiatry. 2007;15(10):832–838.
  • Sun X, Steffens DC, Au R, et al. Amyloid-associated depression: a prodromal depression of Alzheimer disease? Arch Gen Psychiatry. 2008;65(5):542–550.
  • Castellani RJ, Lee H-G, Zhu X, et al. Alzheimer disease pathology as a host response. J Neuropathol Exp Neurol. 2008;67(6):523–531.
  • Struble RG, Ala T, Patrylo PR, et al. Is brain amyloid production a cause or a result of dementia of the Alzheimer’s type? J Alzheimers Dis. 2010;22(2):393–399.
  • Toyn J. What lessons can be learned from failed Alzheimer’s disease trials? Expert Rev Clin Pharmacol. 2015;8(3):267–269.
  • Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8(6):595–608.
  • Karran E, De Strooper B. The amyloid cascade hypothesis: are we poised for success or failure? J Neurochem. Forthcoming 2016. [Epub ahead of print].
  • Ngandu T, Lehtisalo J, Solomon A, et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. Lancet. 2015;385(9984):2255–2263.
  • Lesne` S, Koh MT, Kotilinek L, et al. A specific amyloid-β protein assembly in the brain impairs memory. Nature. 2006;440(7082):352–357.
  • Koffie RM, Meyer-Luehmann M, Hashimoto T, et al. Oligomeric amyloid β associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques. Proc Natl Acad Sci USA. 2009;106(10):4012–4017.
  • Lee EB, Leng LZ, Zhang B, et al. Targeting amyloid-β peptide (Aβ) oligomers by passive immunization with a conformation-selective monoclonal antibody improves learning and memory in Aβ precursor protein (APP) transgenic mice. J Biol Chem. 2006;281(7):4292–4299.
  • Lambert MP, Velasco PT, Chang L, et al. Monoclonal antibodies that target pathological assemblies of Aβ. J Neurochem. 2007;100(1):23–35.
  • Frost JL, Liu B, Kleinschmidt M, et al. Passive immunization against pyroglutamate-3 amyloid-β reduces plaque burden in Alzheimer’s-like transgenic mice: a pilot study. Neurodegenerative Dis. 2012;10(1–4):265–270.
  • 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(12):2149–2156.
  • DeMattos RB, Lu J, Tang Y, et al. A plaque-specific antibody clears existing β-amyloid plaques in Alzheimer’s disease mice. Neuron. 2012;76(5):908–920.
  • Morales-Corraliza J, Schmidt SD, Mazzella MJ, et al. Immunization targeting a minor plaque constituent clears β-amyloid and rescues behavioral deficits in an Alzheimer’s disease mouse model. Neurobiol Aging. 2013;34(1):137–145.

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.