535
Views
22
CrossRef citations to date
0
Altmetric
Review

Preclinical to phase II amyloid beta (Aβ) peptide modulators under investigation for Alzheimer’s disease

&
Pages 579-592 | Received 04 Aug 2016, Accepted 28 Mar 2017, Published online: 24 Apr 2017

References

  • Sheng M, Sabatini BL, Sudhof TC. Synapses and Alzheimer’s disease. Cold Spring Harb Perspect Biol. 2012;4:1–18.
  • Selkoe DJ. Preventing Alzheimer’s disease. Science. 2012;337:1488–1492.
  • Citron M. Alzheimer’s disease: strategies for disease modification. Nat Rev Drug Discov. 2010;9:387–398.
  • Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol (Berl). 1991;82:239–259.
  • Herrmann N, Chau SA, Kircanski I, et al. Current and emerging drug treatment options for Alzheimer’s disease: a systematic review. Drugs. 2011;71:2031–2065.
  • Alzheimer A. Über eine eigenartige Erkrankung der Hirnrinde. Am J Geriatr Psychiatry. 1907;64:146–148.
  • Lesne S, Koh MT, Kotilinek L, et al. A specific amyloid-beta protein assembly in the brain impairs memory. Nature. 2006;440:352–357.
  • Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256:184–185.
  • Cappai R, Barnham KJ. Delineating the mechanism of Alzheimer’s disease A beta peptide neurotoxicity. Neurochem Res. 2008;33:526–532.
  • Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356.
  • Barghorn S, Nimmrich V, Striebinger A, et al. Globular amyloid beta-peptide oligomer - a homogenous and stable neuropathological protein in Alzheimer’s disease. J Neurochem. 2005;95:834–847.
  • Lacor PN, Buniel MC, Furlow PW, et al. Abeta oligomer-induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer’s disease. J Neurosci. 2007;27:796–807.
  • Wilcox KC, Lacor PN, Pitt J, et al. Abeta oligomer-induced synapse degeneration in Alzheimer’s disease. Cell Mol Neurobiol. 2011;31:939–948.
  • Sokolow S, Luu SH, Nandy K, et al. Preferential accumulation of amyloid-beta in presynaptic glutamatergic terminals (VGluT1 and VGluT2) in Alzheimer’s disease cortex. Neurobiol Dis. 2011;45:381–387.
  • Kummer MP, Heneka MT. Truncated and modified amyloid-beta species. Alzheimer’s Research & Therapy. 2014;6:28.
  • Kummer MP, Hermes M, Delekarte A, et al. Nitration of Tyrosine 10 critically enhances amyloid beta aggregation and plaque formation. Neuron. 2011;71:833–844.
  • Schilling S, Lauber T, Schaupp M, et al. On the seeding and oligomerization of pGlu-amyloid peptides (in vitro). Biochemistry. 2006;45:12393–12399.
  • Russo C, Violani E, Salis S, et al. Pyroglutamate-modified amyloid beta-peptides–AbetaN3(pE)–strongly affect cultured neuron and astrocyte survival. J Neurochem. 2002;82:1480–1489.
  • Selkoe DJ. Soluble oligomers of the amyloid beta-protein impair synaptic plasticity and behavior. Behav Brain Res. 2008;192:106–113.
  • De Felice FG, Velasco PT, Lambert MP, et al. Abeta oligomers induce neuronal oxidative stress through an N-methyl-D-aspartate receptor-dependent mechanism that is blocked by the Alzheimer drug memantine. J Biol Chem. 2007;282:11590–11601.
  • Alberdi E, Sanchez-Gomez MV, Cavaliere F, et al. Amyloid beta oligomers induce Ca2+ dysregulation and neuronal death through activation of ionotropic glutamate receptors. Cell Calcium. 2010;47:264–272.
  • Bieschke J, Herbst M, Wiglenda T, et al. Small-molecule conversion of toxic oligomers to nontoxic b-sheet‚Äìrich amyloid fibrils. Nat Chem Biol 2011;8:93–101.
  • Selkoe DJ. Resolving controversies on the path to Alzheimer’s therapeutics. Nat Med. 2011;17:1060–1065.
  • Christensen DD. Alzheimer’s disease: progress in the development of anti-amyloid disease-modifying therapies. CNS Spectrums. 2007;12(113–116):119–123.
  • Masters CL, Cappai R, Barnham KJ, et al. Molecular mechanisms for Alzheimer’s disease: implications for neuroimaging and therapeutics. J Neurochem. 2006;97:1700–1725.
  • Durham TB, Shepherd TA. Progress toward the discovery and development of efficacious BACE inhibitors. Curr Opin Drug Discov Devel. 2006;9:776–791.
  • Imbimbo BP. Therapeutic potential of gamma-secretase inhibitors and modulators. Curr Top Med Chem. 2008;8:54–61.
  • Evin G, Sernee MF, Masters CL. Inhibition of gamma-secretase as a therapeutic intervention for Alzheimer’s disease: prospects, limitations and strategies. CNS Drugs. 2006;20:351–372.
  • Olson RE, Albright CF. Recent progress in the medicinal chemistry of gamma-secretase inhibitors. Curr Top Med Chem. 2008;8:17–33.
  • Walker LC, Rosen RF. Alzheimer therapeutics-what after the cholinesterase inhibitors? Age Ageing. 2006;35:332–335.
  • Dominguez D, Tournoy J, Hartmann D, et al. Phenotypic and biochemical analyses of BACE1- and BACE2-deficient mice. J Biol Chem. 2005;280:30797–30806.
  • Selkoe D, Kopan R. Notch and Presenilin: regulated intramembrane proteolysis links development and degeneration. Annu Rev Neurosci. 2003;26:565–597.
  • Puzzo D, Privitera L, Leznik E, et al. Picomolar amyloid-beta positively modulates synaptic plasticity and memory in hippocampus. J Neurosci. 2008;28:14537–14545.
  • Abramov E, Dolev I, Fogel H, et al. Amyloid-beta as a positive endogenous regulator of release probability at hippocampal synapses. Nat Neurosci. 2009;12:1567–1576.
  • Giuffrida ML, Caraci F, Pignataro B, et al. Beta-amyloid monomers are neuroprotective. J Neurosci. 2009;29:10582–10587.
  • Kumar DK, Choi SH, Washicosky KJ, et al. Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med. 2016;8:340ra372.
  • Wolfe MS. gamma-Secretase in biology and medicine. Semin Cell Dev Biol. 2009;20:219–224.
  • Henley DB, May PC, Dean RA, et al. Development of semagacestat (LY450139), a functional gamma-secretase inhibitor, for the treatment of Alzheimer’s disease. Expert Opin Pharmacother. 2009;10:1657–1664.
  • Bateman RJ, Siemers ER, Mawuenyega KG, et al. A gamma-secretase inhibitor decreases amyloid-beta production in the central nervous system. Ann Neurol. 2009;66:48–54.
  • Fleisher AS, Raman R, Siemers ER, et al. Phase 2 safety trial targeting amyloid beta production with a gamma-secretase inhibitor in Alzheimer disease. Arch Neurol. 2008;65:1031–1038.
  • Ross J, Sharma S, Winston J, et al. CHF5074 reduces biomarkers of neuroinflammation in patients with mild cognitive impairment: a 12-week, double-blind, placebo-controlled study. Curr Alzheimer Res. 2013;10:742–753.
  • Rogers K, Felsenstein KM, Hrdlicka L, et al. Modulation of gamma-secretase by EVP-0015962 reduces amyloid deposition and behavioral deficits in Tg2576 mice. Mol Neurodegener. 2012;7:61.
  • Citron M, Teplow DB, Selkoe DJ. Generation of amyloid beta protein from its precursor is sequence specific. Neuron. 1995;14:661–670.
  • Evin G, Lessene G, Wilkins S. BACE inhibitors as potential drugs for the treatment of Alzheimer’s disease: focus on bioactivity. Recent Pat CNS Drug Discov. 2011;6:91–106.
  • Probst G, Xu YZ. Small-molecule BACE1 inhibitors: a patent literature review (2006 – 2011). Expert opinion on therapeutic patents. 2012;22:511–540.
  • Evin G. Future therapeutics in Alzheimer’s disease: development status of BACE inhibitors. BioDrugs. 2016;30:173–194.
  • Yan R, Vassar R. Targeting the beta secretase BACE1 for Alzheimer’s disease therapy. Lancet Neurol. 2014;13:319–329.
  • Menting KW, Claassen JA. beta-secretase inhibitor; a promising novel therapeutic drug in Alzheimer’s disease. Front Aging Neurosci. 2014;6:165.
  • Amijee H, Scopes DI. The quest for small molecules as amyloid inhibiting therapies for Alzheimer’s disease. J Alzheimers Dis. 2009;17:33–47.
  • De Strooper B, Chavez Gutierrez L. Learning by failing: ideas and concepts to tackle gamma-secretases in Alzheimer’s disease and beyond. Annu Rev Pharmacol Toxicol. 2015;55:419–437.
  • Neumann U, Rueeger H, Machauer R, et al. A novel BACE inhibitor NB-360 shows a superior pharmacological profile and robust reduction of amyloid-beta and neuroinflammation in APP transgenic mice. Mol Neurodegener. 2015;10:44.
  • Shimshek DR, Jacobson LH, Kolly C, et al. Pharmacological BACE1 and BACE2 inhibition induces hair depigmentation by inhibiting PMEL17 processing in mice. Scientific Reports. 2016;6:21917.
  • Filser S, Ovsepian SV, Masana M, et al. Pharmacological inhibition of BACE1 impairs synaptic plasticity and cognitive functions. Biol Psychiatry. 2015;77:729–739.
  • Zhu K, Xiang XFilser S, et al., Beta-site amyloid precursor protein cleaving enzyme 1 inhibition impairs synaptic plasticity via seizure protein 6. Biol Psychiatry. 2016.
  • Nisha CM, Kumar A, Nair P, et al. Molecular docking and in silico ADMET study reveals acylguanidine 7a as a potential inhibitor of beta-secretase. Adv Bioinformatics. 2016;2016:9258578.
  • Atwal JK, Chen Y, Chiu C, et al. A therapeutic antibody targeting BACE1 inhibits amyloid-beta production in vivo. Sci Transl Med. 2011;3:84ra43.
  • Yu YJ, Zhang Y, Kenrick M, et al. Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Sci Transl Med. 2011;3:84ra44.
  • Wang W, Liu Y, Lazarus RA. Allosteric inhibition of BACE1 by an exosite-binding antibody. Curr Opin Struct Biol. 2013;23:797–805.
  • Saftig P, Hetman M, Schmahl W, et al. Mice deficient for the lysosomal proteinase cathepsin D exhibit progressive atrophy of the intestinal mucosa and profound destruction of lymphoid cells. EMBO Journal. 1995;14:3599–3608.
  • May PC, Willis BA, Lowe SL, et al. The potent BACE1 inhibitor LY2886721 elicits robust central Abeta pharmacodynamic responses in mice, dogs, and humans. J Neurosci. 2015;35:1199–1210.
  • Lai R, Albala B, Kaplow JM, et al. First-in-human study of E2609, a novel BACE1 inhibitor, demonstrates prolonged reductions in plasma beta-amyloid levels after single dosing. Alzheimers Dement. 2012;8:P96.
  • Lai R, Albala B, Kaplow JM, et al. Novel BACE1 inhibitor E2609 reduces plasma and CSF amyloid in health subjects after 14 days oral administration. The 11th International Conference On Alzheimer’s & Parkinson’s Diseases, Florence, Italy., (2013).
  • Timmers M, Broeck BV, Slemmon J, Profiling the dynamics of CSF and plasma Abeta reduction with JNJ-54861911, an oral BACE inhibitor. 12th international conference on Alzheimer’s and Parkinson’s diseases and related neurological disorders (AD/PD), (2015).
  • Luo X, Yan R. Inhibition of BACE1 for therapeutic use in Alzheimer’s disease. Int J Clin Exp Pathol. 2010;3:618–628.
  • Forman M, Tseng J, Palcza J, et al. The Novel BACE Inhibitor MK-8931 Dramatically Lowers CSF Aβ Peptides in Healthy Subjects: Results from a Rising Single Dose Study. Neurology. 2012;78:PL02.004.
  • Forman M, Kleijn H-J, 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:P139.
  • Postina R, Schroeder A, Dewachter I, et al. A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J Clin Invest. 2004;113:1456–1464.
  • Vingtdeux V, Marambaud P. Identification and biology of alpha-secretase. J.Neurochem. 2012;120 Suppl 1:34–45.
  • Desire L, Marcade M, Peillon H, et al. Clinical trials of EHT 0202, a neuroprotective and procognitive alpha-secretase stimulator for Alzheimer’s disease. Alzheimer’s & Dementia. 2009;5:P255–P256.
  • Vellas B, Sol O, Snyder PJ, et al. EHT0202 in Alzheimer’s disease: a 3-month, randomized, placebo-controlled, double-blind study. Curr Alzheimer Res. 2011;8:203–212.
  • Nikolaev A, McLaughlin T, O’Leary DD, et al. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature. 2009;457:981–989.
  • Galvan V, Gorostiza OF, Banwait S, et al. Reversal of Alzheimer’s-like pathology and behavior in human APP transgenic mice by mutation of Asp664. Proc Natl Acad Sci U S A. 2006;103:7130–7135.
  • Harris JA, Devidze N, Halabisky B, et al. Many neuronal and behavioral impairments in transgenic mouse models of Alzheimer’s disease are independent of caspase cleavage of the amyloid precursor protein. J Neurosci. 2010;30:372–381.
  • Lahiri DK, Chen D, Maloney B, et al. The experimental Alzheimer’s disease drug posiphen [(+)-phenserine] lowers amyloid-beta peptide levels in cell culture and mice. J Pharmacol Exp Ther. 2007;320:386–396.
  • Maccecchini ML, Chang MY, Pan C, et al. Posiphen as a candidate drug to lower CSF amyloid precursor protein, amyloid-beta peptide and tau levels: target engagement, tolerability and pharmacokinetics in humans. J Neurol Neurosurg Psychiatry. 2012;83:894–902.
  • Heneka MT, Landreth GE. PPARs in the brain. Biochim Biophys Acta. 2007;1771:1031–1045.
  • Combs CK, Johnson DE, Karlo JC, et al. Inflammatory mechanisms in Alzheimer’s disease: inhibition of beta-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARgamma agonists. J Neurosci. 2000;20:558–567.
  • Kim EJ, Kwon KJ, Park JY, et al. Effects of peroxisome proliferator-activated receptor agonists on LPS-induced neuronal death in mixed cortical neurons: associated with iNOS and COX-2. Brain Res. 2002;941:1–10.
  • Luna-Medina R, Cortes-Canteli M, Alonso M, et al. Regulation of inflammatory response in neural cells in vitro by thiadiazolidinones derivatives through peroxisome proliferator-activated receptor gamma activation. J Biol Chem. 2005;280:21453–21462.
  • Pedersen WA, McMillan PJ, Kulstad JJ, et al. Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Exp Neurol. 2006;199:265–273.
  • Escribano L, Simon AM, Perez-Mediavilla A, et al. Rosiglitazone reverses memory decline and hippocampal glucocorticoid receptor down-regulation in an Alzheimer’s disease mouse model. Biochem Biophys Res Commun. 2009;379:406–410.
  • O’Reilly JA, Lynch M. Rosiglitazone improves spatial memory and decreases insoluble Abeta(1-42) in APP/PS1 mice. J Neuroimmune Pharmacol. 2012;7:140–144.
  • Xu S, Liu G, Bao X, et al. Rosiglitazone prevents amyloid-beta oligomer-induced impairment of synapse formation and plasticity via increasing dendrite and spine mitochondrial number. J Alzheimers Dis. 2014;39:239–251.
  • Xu S, Guan Q, Wang C, et al. Rosiglitazone prevents the memory deficits induced by amyloid-beta oligomers via inhibition of inflammatory responses. Neurosci Lett. 2014;578:7–11.
  • Yu Y, Li X, Blanchard J, et al. Insulin sensitizers improve learning and attenuate tau hyperphosphorylation and neuroinflammation in 3xTg-AD mice. J Neural Transm. 2015;122:593–606.
  • Tzimopoulou S, Cunningham VJ, Nichols TE, et al. Multi-center randomized proof-of-concept clinical trial applying [(1)(8)F]FDG-PET for evaluation of metabolic therapy with rosiglitazone XR in mild to moderate Alzheimer’s’ disease. J Alzheimers Dis. 2010;22:1241–1256.
  • Watson GS, Cholerton BA, Reger MA, et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. Am J Geriatr Psychiatry. 2005;13:950–958.
  • Risner ME, Saunders AM, Altman JF, et al. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer’s disease. Pharmacogenomics J. 2006;6:246–254.
  • Harrington C, Sawchak S, Chiang C, et al. Rosiglitazone does not improve cognition or global function when used as adjunctive therapy to AChE inhibitors in mild-to-moderate Alzheimer’s disease: two phase 3 studies. Curr Alzheimer Res. 2011;8:592–606.
  • Gold M, Alderton C, Zvartau-Hind M, et al. Rosiglitazone monotherapy in mild-to-moderate Alzheimer’s disease: results from a randomized, double-blind, placebo-controlled phase III study. Dement Geriatr Cogn Disord. 2010;30:131–146.
  • Lunemann JD, Nimmerjahn F, Dalakas MC. Intravenous immunoglobulin in neurology–mode of action and clinical efficacy. Nat Rev Neurol. 2015;11:80–89.
  • Loeffler DA. Should development of Alzheimer’s disease-specific intravenous immunoglobulin be considered? J Neuroinflammation. 2014;11:198.
  • Gong Y, Chang L, Viola KL, et al. Alzheimer’s disease-affected brain: presence of oligomeric Abeta ligands (ADDLs) suggests a molecular basis for reversible memory loss. Proc Natl Acad Sci U S A. 2003;100:10417–10422.
  • Lee EB, Leng LZ, Zhang B, et al. Targeting amyloid-beta peptide (Abeta) oligomers by passive immunization with a conformation-selective monoclonal antibody improves learning and memory in Abeta precursor protein (APP) transgenic mice. J Biol Chem. 2006;281:4292–4299.
  • Hillen H, Barghorn S, Striebinger A, et al. Generation and therapeutic efficacy of highly oligomer-specific beta-amyloid antibodies. J Neurosci. 2010;30:10369–10379.
  • Zago W, Buttini M, Comery TA, et al. Neutralization of soluble, synaptotoxic amyloid beta species by antibodies is epitope specific. J Neurosci. 2012;32:2696–2702.
  • De Felice FG, Wu D, Lambert MP, et al. Alzheimer’s disease-type neuronal tau hyperphosphorylation induced by Abeta oligomers. Neurobiol Aging. 2008;29:1334–1347.
  • Shughrue PJ, Acton PJ, Breese RS, et al. Anti-ADDL antibodies differentially block oligomer binding to hippocampal neurons. Neurobiol Aging. 2010;31:189–202.
  • Zameer A, Kasturirangan S, Emadi S, et al. Anti-oligomeric Abeta single-chain variable domain antibody blocks Abeta-induced toxicity against human neuroblastoma cells. J Mol Biol. 2008;384:917–928.
  • Venkataramani V, Wirths O, Budka H, et al. Antibody 9D5 recognizes oligomeric pyroglutamate amyloid-beta in a fraction of amyloid-beta deposits in Alzheimer’s disease without cross-reactivity with other protein aggregates. J Alzheimers Dis. 2012;29:361–371.
  • Tucker S, Moller C, Tegerstedt K, et al. The murine version of BAN2401 (mAb158) selectively reduces amyloid-beta protofibrils in brain and cerebrospinal fluid of tg-ArcSwe mice. J Alzheimers Dis. 2015;43:575–588.
  • Glabe CG. Structural classification of toxic amyloid oligomers. J Biol Chem. 2008;283:29639–29643.
  • Lambert MP, Barlow AK, Chromy BA, et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A. 1998;95:6448–6453.
  • Lacor PN, Buniel MC, Chang L, et al. Synaptic targeting by Alzheimer’s-related amyloid beta oligomers. J Neurosci. 2004;24:10191–10200.
  • Bontempi, B., Silva, Alcino J. (Eds.) Memories: Molecules and CircuitsResearch and Perspectives in Neurosciences. 2007;155–179.
  • Krafft GA, Klein WL. ADDLs and the signaling web that leads to Alzheimer’s disease. Neuropharmacology. 2010;59:230–242.
  • Crews L, Masliah E. Molecular mechanisms of neurodegeneration in Alzheimer’s disease. Hum Mol Genet. 2010;19:R12–R20.
  • Shankar GM, Li S, Mehta TH, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14:837–842.
  • Townsend M, Mehta T, Selkoe DJ. Soluble Abeta inhibits specific signal transduction cascades common to the insulin receptor pathway. J Biol Chem. 2007;282:33305–33312.
  • Klein WL, Krafft GA, Finch CE. Targeting small Abeta oligomers: the solution to an Alzheimer’s disease conundrum? Trends Neurosci. 2001;24:219–224.
  • Dinamarca MC, Rios JA, Inestrosa NC. Postsynaptic receptors for amyloid-beta oligomers as mediators of neuronal damage in Alzheimer’s disease. Front Physiol. 2012;3:464.
  • Koffie RM, Hyman BT, Spires-Jones TL. Alzheimer’s disease: synapses gone cold. Mol Neurodegener. 2011;6:63.
  • Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8:101–112.
  • Cerpa W, Dinamarca MC, Inestrosa NC. Structure-function implications in Alzheimer’s disease: effect of Abeta oligomers at central synapses. Curr Alzheimer Res. 2008;5:233–243.
  • Walsh DM, Klyubin I, Fadeeva JV, et al. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–539.
  • Rammes G, Hasenjager A, Sroka-Saidi K, et al. Therapeutic significance of NR2B-containing NMDA receptors and mGluR5 metabotropic glutamate receptors in mediating the synaptotoxic effects of beta-amyloid oligomers on long-term potentiation (LTP) in murine hippocampal slices. Neuropharmacology. 2011;60:982–990.
  • Hsieh H, Boehm J, Sato C, et al. AMPAR removal underlies Abeta-induced synaptic depression and dendritic spine loss. Neuron. 2006;52:831–843.
  • Chang L, Bakhos L, Wang Z, et al. Femtomole immunodetection of synthetic and endogenous amyloid-beta oligomers and its application to Alzheimer’s disease drug candidate screening. J Mol Neurosci. 2003;20:305–313.
  • Westerman MA, Cooper-Blacketer D, Mariash A, et al. The relationship between Abeta and memory in the Tg2576 mouse model of Alzheimer’s disease. J Neurosci. 2002;22:1858–1867.
  • Ashe KH. Mechanisms of memory loss in Abeta and tau mouse models. Biochem Soc Trans. 2005;33:591–594.
  • Selkoe DJ. Alzheimer’s disease is a synaptic failure. Science. 2002;298:789–791.
  • Catalano SM, Dodson EC, Henze DA, et al. The role of amyloid-beta derived diffusible ligands (ADDLs) in Alzheimer’s disease. Curr Top Med Chem. 2006;6:597–608.
  • Walsh DM, Selkoe DJ. A beta oligomers - a decade of discovery. J Neurochem. 2007;101:1172–1184.
  • Benilova I, Karran E, De Strooper B. The toxic Abeta oligomer and Alzheimer’s disease: an emperor in need of clothes. Nat Neurosci. 2012;15:349–357.
  • Brouillette J, Caillierez R, Zommer N, et al. Neurotoxicity and memory deficits induced by soluble low-molecular-weight amyloid-beta1-42 oligomers are revealed in vivo by using a novel animal model. J Neurosci. 2012;32:7852–7861.
  • Larson ME, Lesne SE. Soluble Abeta oligomer production and toxicity. J Neurochem. 2012;120(Suppl 1):125–139.
  • Parsons CG, Ruitenberg M, Freitag CE, et al. MRZ-99030 - A novel modulator of Abeta aggregation: I - Mechanism of action (MoA) underlying the potential neuroprotective treatment of Alzheimer’s disease, glaucoma and age-related macular degeneration (AMD). Neuropharmacology. 2015;92:158–169.
  • Rammes G, Gravius A, Ruitenberg M, et al. MRZ-99030 - A novel modulator of Abeta aggregation: II - Reversal of Abeta oligomer-induced deficits in long-term potentiation (LTP) and cognitive performance in rats and mice. Neuropharmacology. 2015;92:170–182.
  • Gervais F, Paquette J, Morissette C, et al. Targeting soluble Abeta peptide with Tramiprosate for the treatment of brain amyloidosis. Neurobiol Aging. 2007;28:537–547.
  • Aisen PS, Saumier D, Briand R, et al. A Phase II study targeting amyloid-beta with 3APS in mild-to-moderate Alzheimer disease. Neurology. 2006;67:1757–1763.
  • Gauthier S, Aisen PS, Ferris SH, et al. Effect of tramiprosate in patients with mild-to-moderate Alzheimer’s disease: exploratory analyses of the MRI sub-group of the Alphase study. J Nutr Health Aging. 2009;13:550–557.
  • 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.
  • Adlard PA, Cherny RA, Finkelstein DI, et al. Rapid restoration of cognition in Alzheimer’s transgenic mice with 8-hydroxy quinoline analogs is associated with decreased interstitial Aβ. Neuron. 2008;59:43–55.
  • Sampson EL, Jenagaratnam L, McShane R. Metal protein attenuating compounds for the treatment of Alzheimer’s dementia. Cochrane Database Syst Rev. 2014;Cd005380. doi: 10.1002/14651858.CD005380.pub5.
  • Mandel SA, Amit T, Weinreb O, et al. Simultaneous manipulation of multiple brain targets by green tea catechins: a potential neuroprotective strategy for Alzheimer and Parkinson diseases. CNS Neurosci Ther. 2008;14:352–365.
  • Look GC, Jerecic J, Cherbavaz DB, et al. Discovery of ADDL–targeting small molecule drugs for Alzheimer’s disease. Curr Alzheimer Res. 2007;4:562–567.
  • Frydman-Marom A, Rechter M, Shefler I, et al. Cognitive-performance recovery of Alzheimer’s disease model mice by modulation of early soluble amyloidal assemblies. Angew Chem Int Ed Engl. 2009;48:1981–1986.
  • Lai ZW, Petrera A, Schilling O. Protein amino-terminal modifications and proteomic approaches for N-terminal profiling. Curr Opin Chem Biol. 2015;24:71–79.
  • Jawhar S, Wirths T, Bayer A. Pyroglutamate amyloid-beta (Aβ): a hatchet man in Alzheimer disease. J Biol Chem. 2011;286:38825–38832.
  • Frost JL, Le KX, Cynis H, et al. Pyroglutamate-3 amyloid-β deposition in the brains of humans, non-human primates, canines, and Alzheimer disease-like transgenic mouse models. Am J Pathol. 2013;183:369–381.
  • Cynis H, Schilling S, Bodnar M, et al. Inhibition of glutaminyl cyclase alters pyroglutamate formation in mammalian cells. Biochim Biophys Acta. 2006;1764:1618–1625.
  • Schilling S, Zeitschel U, Hoffmann T, et al. Glutaminyl cyclase inhibition attenuates pyroglutamate Aβ and Alzheimer’s disease-like pathology. Nat Med. 2008;14:1106–1111.
  • Hofling C, Indrischek H, Hopcke T, et al. Mouse strain and brain region-specific expression of the glutaminyl cyclases QC and isoQC. Int J Dev Neurosci. 2014;36:64–73.

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.