424
Views
14
CrossRef citations to date
0
Altmetric
Review

Challenges in the treatment of Alzheimer’s disease: recent progress and treatment strategies of pharmaceuticals targeting notable pathological factors

&
Pages 623-652 | Received 07 Jan 2019, Accepted 17 May 2019, Published online: 29 May 2019

References

  • Sevigny J, Chiao P, Bussiere T, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016;537:50–56.
  • Winblad B, Amouyel P, Andrieu S, et al. Defeating Alzheimer’s disease and other dementias: a priority for European science and society. Lancet Neurol. 2016;15:455–532.
  • Godyn J, Jonczyk J, Panek D, et al. Therapeutic strategies for Alzheimer’s disease in clinical trials. Pharmacol Rep. 2016;68:127–138.
  • Siemers ER, Sundell KL, Carlson C, et al. Phase 3 solanezumab trials: secondary outcomes in mild Alzheimer’s disease patients. Alzheimers Dement. 2016;12:110–120.
  • Dubios B, Hampel H, Feldman HH, et al. Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 2016;12:292–323.
  • Anand R, Gill KD, Mahdi AA. Therapeutics of Alzheimer’s disease: past, present and future. Neuropharmacology. 2014;76:27–50.
  • Panza F, Lozupone M, Solfrizzi V, et al. BACE inhibitors in clinical development for the treatment of Alzheimer’s disease. Expert Rev Neurother. 2018;18:847–857.
  • Calsolaro V, Edison P. Neuroinflammation in Alzheimer’s disease: current evidence and future directions. Alzheimers Dement. 2016;12:719–732.
  • Strooper BD, Karran E. The cellular phase of Alzheimer’s disease. Cell. 2016;164:603–615.
  • Zhang TT, Li W, Meng G, et al. Strategies for transporting nanoparticles across the blood-brain barrier. Biomater Sci. 2016;4:219–229.
  • Liu Y, An S, Li J, et al. Brain-targeted co-delivery of therapeutic gene and peptide by multifunctional nanoparticles in Alzheimer’s disease mice. Biomaterials. 2016;80:33–45.
  • Chen ZL, Huang M, Wang XR, et al. Transferrin-modified liposome promotes α-mangostin to penetrate the blood-brain barrier. Nanomed Nanotech Biol Med. 2016;12:421–430.
  • Vedagiri A, Thangarajan S. Mitigating effect of chrysin loaded solid lipid nanoparticles against amyloid β25–35 induced oxidative stress in rat hippocampal region: an efficient formulation approach for Alzheimer’s disease. Neuropeptides. 2016;58:111–125.
  • Loureiro JA, Gomes B, Fricker G, et al. Cellular uptake of PLGA nanoparticles targeted with anti-amyloid and anti-transferrin receptor antibodies for Alzheimer’s disease treatment. Colloids Surf B. 2016;145:8–13.
  • Kuo YC, Lin CY, Li JS, et al. Wheat germ agglutinin-conjugated liposomes incorporated with cardiolipin to improve neuronal survival in Alzheimer’s disease treatment. Int J Nanomed. 2017;12:1757–1774.
  • Kuo YC, Tsao CW. Neuroprotection against apoptosis of SK-N-MC cells using RMP-7- and lactoferrin-grafted liposomes carrying quercetin. Int J Nanomed. 2017;12:2857–2869.
  • Tamaru M, Akita H, Nakatani T, et al. Application of apolipoprotein E-modified liposomal nanoparticles as a carrier for delivering DNA and nucleic acid in the brain. Int J Nanomed. 2014;9:4267–4276.
  • Tamaru M, Akita H, Kajimoto K, et al. An apolipoprotein E modified liposomal nanoparticle: ligand dependent efficiency as a siRNA delivery carrier for mouse-derived brain endothelial cells. Int J Pharm. 2014;465:77–82.
  • Poirier J, Miron J, Picard C, et al. Apolipoprotein E and lipid homeostasis in the etiology and treatment of sporadic Alzheimer’s disease. Neurobiol Aging. 2014;35:S3–10.
  • Kuo YC, Lin CY. Targeting delivery of liposomes with conjugated p-aminophenyl-α-d-manno-pyranoside and apolipoprotein E for inhibiting neuronal degeneration insulted with β-amyloid peptide. J Drug Target. 2015;23:147–158.
  • Kuo YC, Lee YJ. Rescuing cholinergic neurons from apoptotic degeneration by targeting of serotonin modulator- and apolipoprotein E-conjugated liposomes to hippocampus. Int J Nanomed. 2016;11:6809–6824.
  • Kuo YC, Rajesh R. A critical overview of therapeutic strategy and advancement for Alzheimer’s disease treatment. J Taiwan Inst Chem Eng. 2016;77:92–105.
  • 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;370:322–333.
  • 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;370:311–321.
  • 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.
  • 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.
  • Lovestone S, Boada M, Dubois B, et al. A phase II trial of tideglusib in Alzheimer’s disease. J Alzheimers Dis. 2015;45:75–88.
  • Davtyan H, Ghochikyan A, Petrushina I, et al. Immunogenicity, efficacy, safety, and mechanism of action of epitope vaccine (Lu AF20513) for Alzheimer’s disease: prelude to a clinical trial. J Neurosci. 2013;33:4923–4934.
  • Lee ST, Chu K, Park JE, et al. Erythropoietin improves memory function with reducing endothelial dysfunction and amyloid-beta burden in Alzheimer’s disease models. J Neurochem. 2012;120:115–124.
  • Burstein AH, Dunn I, Soeder T, et al. Azeliragon phase 2b survival analysis supports beneficial effects on delaying time to cognitive deterioration in patients with mild Alzheimer’s disease. Alzheimers Dement. 2016;12:352.
  • Timmers M, Broeck BV, Ramael S, et al. Profiling the dynamics of CSF and plasma Aβ reduction after treatment with JNJ-54861911, a potent oral BACE inhibitor. Alzheimers Dement. 2016;2:202–212.
  • Sakamoto K, Matsuki S, Matsuguma K, et al. BACE1 Inhibitor lanabecestat (AZD3293) in a phase 1 study of healthy Japanese subjects: pharmacokinetics and effects on plasma and cerebrospinal fluid Aβ peptides. J Clin Pharmacol. 2017;57:1460–1471.
  • Hawkes N. Merck ends trial of potential Alzheimer’s drug verubecestat. Bmj. 2017;356:j845.
  • Lai RYK, Darpo B, Dayal S, et al. Elenbecestat, a novel oral BACE inhibitor, has no clinically meaningful effect on QTC interval up to a supratherapeutic dose of 200 mg. Alzheimers Dement. 2017;13:250–251.
  • Ulfer M, Rouzade-Dominguez ML, Huledal G, et al. Results from a 3-month study in healthy subjects aged 60 years with the BACE inhibitor CNP520. Alzheimers Dement. 2017;7:256.
  • Sun X, Cheng WD, Wang YD. β-amyloid: the key peptide in the pathogenesis of Alzheimer’s disease. Front Pharmacol. 2015;6:1–9.
  • Karran E, Hardy J. A critique of the drug discovery and phase 3 clinical programs targeting the amyloid hypothesis for Alzheimer disease. Ann Neurol. 2014;76:185–205.
  • Sevalle J, Amoyel A, Robert P, et al. Aminopeptidase A contributes to the N-terminal truncation of amyloid beta-peptide. J Neurochem. 2009;109:248–256.
  • Schonherr C, Bein J, Isbert S, et al. Generation of aggregation prone N-terminally truncated amyloid β peptides by meprin β depends on the sequence specificity at the cleavage site. ‎Mol Neurodegener. 2016;11:1–16.
  • Leissring MA, Lu A, Condron MM, et al. Kinetics of amyloid beta-protein degradation determined by novel fluorescence- and fluorescence polarization-based assays. J Biol Chem. 2003;278:37314–37320.
  • Liao MC, Ahmed M, Smith SO, et al. Degradation of amyloid beta protein by purified myelin basic protein. J Biol Chem. 2009;284:28917–28925.
  • Hu J, Igarashi A, Kamata M, et al. Angiotensin-converting enzyme degrades Alzheimer amyloid beta-peptide (A beta); retards A beta aggregation, deposition, fibril formation; and inhibits cytotoxicity. J Biol Chem. 2001;276:47863–47868.
  • Nostrand WEV, Porter M. Plasmin cleavage of the amyloid beta-protein: alteration of secondary structure and stimulation of tissue plasminogen activator activity. Biochemistry. 1999;38:11570–11576.
  • Baranello RJ, Bharani KL, Padmaraju V, et al. Amyloid-beta protein clearance and degradation (ABCD) pathways and their role in Alzheimer’s disease. Curr Alzheimer Res. 2015;12:32–46.
  • Cai Z, Hussain MD, Yan LJ. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int J Neurosci. 2014;124:307–321.
  • Mosher KI, Wyss-Coray T. Microglial dysfunction in brain aging and Alzheimer’s disease. Biochem Pharmacol. 2014;88:594–604.
  • Wang Y, Cella M, Mallinson K, et al. TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell. 2015;160:1061–1071.
  • Yeh FL, Wang Y, Tom I, et al. TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of amyloid-beta by microglia. Neuron. 2016;16:328–340.
  • Griciu A, Serrano-Pozo A, Parrado AR, et al. Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron. 2013;78:631–643.
  • Killick R, Ribe EM, Al-Shawi R, et al. Clusterin regulates β-amyloid toxicity via Dickkopf-1-driven induction of the wnt-PCP-JNK pathway. Mol Psychiatry. 2014;19:88–98.
  • Hamelin L, Lagarde J, Dorothee G, et al. Early and protective microglial activation in Alzheimer’s disease: a prospective study using 18F-DPA-714 PET imaging. Brain. 2016;139:1252–1264.
  • Zhao Z, Sagare AP, Ma Q, et al. Central role for PICALM in amyloid-β blood-brain barrier transcytosis and clearance. Nature Neurosci. 2015;18:978–987.
  • Hane F, Leonenko Z. Effect of metals on kinetic pathways of amyloid-β aggregation. Biomolecules. 2014;4:101–116.
  • Faller P, Hureau C, Penna GL. Metal ions and intrinsically disordered proteins and peptides: from Cu/Zn amyloid-β to general principles. Acc Chem Res. 2014;47:2252–2259.
  • Notarachille G, Arnesano F, Calo V, et al. Heavy metals toxicity: effect of cadmium ions on amyloid beta protein 1–42. Possible implications for Alzheimer’s disease. Biometals. 2014;27:371–388.
  • Pimplikar SW, Nixon RA, Robakis NK, et al. Amyloid-independent mechanisms in Alzheimer’s disease pathogenesis. J Neurosci. 2010;30:14946–14954.
  • Kumar S, Wirths O, Stuber K, et al. Phosphorylation of the amyloid β-peptide at Ser26 stabilizes oligomeric assembly and increases neurotoxicity. Acta Neuropathol. 2016;131:525–537.
  • Umeda T, Tomiyama T, Sakama N, et al. Intraneuronal amyloid beta oligomers cause cell death via endoplasmic reticulum stress, endosomal/lysosomal leakage, and mitochondrial dysfunction in vivo. J Neurosci Res. 2011;89:1031–1042.
  • Cheng B, Gong H, Xiao H, et al. Inhibiting toxic aggregation of amyloidogenic proteins: a therapeutic strategy for protein misfolding diseases. Biochim Biophys Acta Gen Subj. 2013;1830:4860–4871.
  • Ghosh AK, Osswald HL. BACE1 (β-secretase) inhibitors for the treatment of Alzheimer’s disease. Chem Soc Rev. 2014;43:6765–6813.
  • Yan R, Vassar R. Targeting the β secretase BACE1 for Alzheimer’s disease therapy. Lancet Neurol. 2014;13:319–329.
  • Chen Y, Huang X, Zhang Y, et al. Alzheimer’s β-secretase (BACE1) regulates the cAMP/PKA/CREB pathway independently of β-amyloid. J Neurosci. 2012;32:11390–11395.
  • Devi L, Ohno M. 7,8-dihydroxyflavone, a small-molecule TrkB agonist, reverses memory deficits and BACE1 elevation in a mouse model of Alzheimer’s disease. Neuropsychopharmacology. 2012;37:434–444.
  • Chen CH, Zhou W, Liu S, et al. Increased NF-kB signalling up-regulates BACE1 expression and its therapeutic potential in Alzheimer’s disease. Int J Neuropsychopharmacol. 2012;15:77–90.
  • Marwarh G, Raza S, Prasanthi JRP, et al. Gadd153 and NF-kB crosstalk regulates 27-hydroxycholesterol-induced increase in BACE1 and β-amyloid production in human neuroblastoma SH-SY5Y cells. PLOS One. 2013;8:e70773.
  • Prabhu Y, Burgos PV, Schindler C, et al. Adaptor protein 2-mediated endocytosis of the β-secretase BACE1 is dispensable for amyloid precursor protein processing. Mol Biol Cell. 2012;23:2339–2351.
  • Cumming JN, Smith EM, Wang L, et al. Structure based design of iminohydantoin BACE1 inhibitors: identification of an orally available, centrally active BACE1 inhibitor. Bioorg Med Chem Lett. 2012;22:2444–2449.
  • Stamford AW, Wu Y, Scott JD, et al. Discovery of an orally available, brain penetrant BACE1 inhibitor that affords robust CNS Aβ reduction. ACS Med Chem Lett. 2012;3:897–902.
  • Hilpert H, Guba W, Woltering TJ, et al. β-secretase (BACE1) inhibitors with high in vivo efficacy suitable for clinical evaluation in Alzheimer’s disease. J Med Chem. 2013;56:3980–3995.
  • Yonezawa S, Yamakawa H, Muto C, et al. Conformational restriction approach to BACE1 inhibitors II: SAR study of the isocytosine derivatives fixed with a cis-cyclopropane ring. Bioorg Med Chem Lett. 2013;23:2912–2915.
  • Edraki N, Firuzi O, Foroumadi A, et al. Phenylimino-2H-chromen-3-carboxamide derivatives as novel small molecule inhibitors of β-secretase (BACE1). Bioorg Med Chem Lett. 2013;21:2396–2412.
  • Hettich MM, Matthes F, Ryan DP, et al. The anti-diabetic drug metformin reduces BACE1 protein level by interfering with the MID1 complex. PLOS One. 2014;9:e102420.
  • Thomas AA, Hunt KW, Volgraf M, et al. Discovery of 7-tetrahydropyran-2-yl chromans: β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors that reduce amyloid β-protein (Aβ) in the central nervous system. J Med Chem. 2014;57:878–902.
  • Dineen TA, Chen K, Cheng AC, et al. Inhibitors of β-site amyloid precursor protein cleaving enzyme (BACE1): identification of (S)-7-(2-fluoropyridin-3-yl)-3-((3-methyloxetan-3-yl)ethynyl)-5′H-spiro[chromeno[2,3-b]pyridine-5,4′-oxazol]-2′-amine (AMG-8718). J Med Chem. 2014;57:9811–9831.
  • Epstein O, Bryan MC, Cheng AC, et al. Lead optimization and modulation of hERG activity in a series of aminooxazoline xanthene β-site amyloid precursor protein cleaving enzyme (BACE1) inhibitors. J Med Chem. 2014;57:9796–97810.
  • Lu X, Deng Y, Yu D, et al. Histone acetyltransferase p300 mediates histone acetylation of PS1 and BACE1 in a cellular model of Alzheimer’s disease. PLOS One. 2014;9:e103067.
  • Marwarha G, Raza S, Meiers C, et al. Leptin attenuates BACE1 expression and amyloid-β genesis via the activation of SIRT1 signaling pathway. Biochim Biophys Acta. 2014;1842:1587–1595.
  • Adwan L, Subaiea GM, Zawia NH. Tolfenamic acid downregulates BACE1 and protects against lead-induced upregulation of Alzheimer’s disease related biomarkers. Neuropharmacology. 2014;79:596–602.
  • Wang Q, Xiao B, Cui S, et al. Triptolide treatment reduces Alzheimer’s disease (AD)-like pathology through inhibition of BACE1 in a transgenic mouse model of AD. Dis Models Mech. 2014;7:1385–1395.
  • Wang R, Chen S, Liu Y, et al. All-trans-retinoic acid reduces BACE1 expression under inflammatory conditions via modulation of nuclear factor κB (NFκB) signaling. J Biol Chem. 2015;37:22532–22542.
  • Zheng N, Yuan P, Li C, et al. Luteolin reduces BACE1 expression through NF-κB and estrogen receptor mediated pathways in HEK293 and SH-SY5Y cells. J Alzheimers Dis. 2015;45:659–671.
  • Filser S, Ovsepian SV, Masana M, et al. Pharmacological inhibition of BACE1 impairs synaptic plasticity and cognitive functions. Biol Psychiatry. 2015;77:729–739.
  • Orejana L, Barros-Minones L, Jordan J, et al. Sildenafil decreases BACE1 and cathepsin B levels and reduces APP amyloidogenic processing in the SAMP8 mouse. J Gerontol A Biol Sci Med Sci. 2015;70:675–685.
  • Qi C, Bao J, Wang J, et al. Asperterpenes A and B, two unprecedented meroterpenoids from Aspergillus terreus with BACE1 inhibitory activities. Chem Sci. 2016;7:6563–6572.
  • Zhang JS, Zhou SF, Wang Q, et al. Gastrodin suppresses BACE1 expression under oxidative stress condition via inhibition of the PKR/eIF2a pathway in Alzheimer’s disease. Neuroscience. 2016;325:1–9.
  • Chon SH, Yang EJ, Lee T, et al. β-Secretase (BACE1) inhibitory and neuroprotective effects of p-terphenyls from polyozellus multiplex. Food Funct. 2016;7:3834–3842.
  • Butler CR, Ogilvie K, Martinez-Alsina L, et al. Aminomethyl-derived beta secretase (BACE1) inhibitors: engaging Gly230 without an anilide functionality. J Med Chem. 2017;60:386–402.
  • Fang M, Wang J, Zhang X, et al. The miR-124 regulates the expression of BACE1/β-secretase correlated with cell death in Alzheimer’s disease. Toxicol Lett. 2012;209:94–105.
  • Zhu HC, Wang LM, Wang M, et al. MicroRNA-195 downregulates Alzheimer’s disease amyloid-β production by targeting BACE1. Brain Res Bull. 2012;88:596–601.
  • Liu L, Martin R, Chan C. Palmitate-activated astrocytes via serine palmitoyltransferase increase BACE1 in primary neurons by sphingomyelinases. Neurobiol Aging. 2013;34:540–550.
  • Yun SM, Cho SJ, Song JC, et al. SUMO1 modulates Aβ generation via BACE1 accumulation. Neurobiol Aging. 2013;34:650–662.
  • Chlebek J, Simone AD, Hostalkova A, et al. Application of BACE1 immobilized enzyme reactor for the characterization of multifunctional alkaloids from Corydalis cava (Fumariaceae) as Alzheimer’s disease targets. Fitoterapia. 2016;109:241–247.
  • Youn K, Jun M. In vitro BACE1 inhibitory activity of geraniin and corilagin from geranium thunbergii. Planta Med. 2013;79:1038–1042.
  • Youn K, Jeong WS, Jun M. β-secretase (BACE1) inhibitory property of loganin isolated from Corni fructus. Nat Prod Res. 2013;27:1471–1474.
  • Veerappan K, Natarajan S, Subramaniyam S, et al. Identification of BACE1 inhibitors from Panax ginseng saponins-an Insilco approach. Comput Biol Med. 2013;43:1037–1044.
  • Yu F, Zhang Y, Chuang DM. Lithium reduces BACE1 overexpression, beta amyloid accumulation, and spatial learning deficits in mice with traumatic brain injury. J Neurotrauma. 2012;29:2432–2451.
  • Li S, Liu Z, Ji F, et al. Delivery of quantum dot-siRNA nanoplexes in SK-N-SH cells for BACE1 gene silencing and intracellular imaging. Mol Ther Nucleic Acids. 2012;1:e20.
  • Kennedy ME, Stamford AW, Chen X, et al. The BACE1 inhibitor verubecestat (MK-8931) reduces CNS β-amyloid in animal models and in Alzheimer’s disease patients. Sci Transl Med. 2016;8:363ra150.
  • Woltering TJ, Wostl W, Hilpert H, et al. BACE1 inhibitors: a head group scan on a series of amides. Bioorg Med Chem Lett. 2013;23:4239–4243.
  • Jeppsson F, Eketjall S, Janson J, et al. Discovery of AZD3839, a potent and selective bace1 inhibitor clinical candidate for the treatment of Alzheimer disease. J Biol Chem. 2012;287:41245–41257.
  • May PC, Willis BA, Lowe SL, et al. The potent BACE1 inhibitor LY2886721 elicits robust central Aβ pharmacodynamic responses in mice, dogs, and humans. J Neurosci. 2015;35:1199–1210.
  • Eketjall S, Janson J, Kaspersson K, et al. AZD3293: a novel, orally active BACE1 inhibitor with high potency and permeability and markedly slow off-rate kinetics. J Alzheimers Dis. 2016;50:1109–1123.
  • Golde TE, Koo EH, Felsensteinv KM, et al. γ-Secretase inhibitors and modulators. Biochim Biophys Acta Biomembr. 2013;1828:2898–2907.
  • Mitani Y, Yarimizu J, Saita K, et al. Differential effects between γ-secretase inhibitors and modulators on cognitive function in amyloid precursor protein-transgenic and nontransgenic mice. J Neurosci. 2012;32:2037–2050.
  • 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.
  • Pozdnyakov N, Murrey HE, Crump CJ, et al. γ-secretase modulator (GSM) photoaffinity probes reveal distinct allosteric binding sites on presenilin. J Biol Chem. 2013;288:9710–9720.
  • Bischoff F, Berthelot D, Cleyn MD, et al. Design and synthesis of a novel series of bicyclic heterocycles as potent γ-secretase modulators. J Med Chem. 2012;55:9089–9106.
  • Rogers K, Felsenstein KM, Hrdlicka L, et al. Modulation of γ-secretase by EVP-0015962 reduces amyloid deposition and behavioral deficits in Tg2576 mice. ‎Mol Neurodegener. 2012;7:61.
  • Findeis MA, Schroeder F, McKee TD, et al. Discovery of a novel pharmacological and structural class of gamma secretase modulators derived from the extract of Actaea racemosa. ACS Chem Neurosci. 2012;3:941–951.
  • Jung JI, Ladd TB, Kukar T, et al. Steroids as γ-secretase modulators. Faseb J. 2013;27:3775–3785.
  • Yu Y, Logovinsky V, Schuck E, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of the novel γ-secretase modulator, E2212, in healthy human subjects. J Clin Pharmacol. 2014;54:528–536.
  • Loureiro RM, Dumin JA, McKee TD, et al. Efficacy of SPI-1865, a novel gamma-secretase modulator, in multiple rodent models. Alzheimers Res Ther. 2013;5:19.
  • Chena JJ, Qiana W, Biswasa K, et al. Discovery of 2-methylpyridine-based biaryl amides as γ-secretase modulators for the treatment of Alzheimer’s disease. Bioorg Med Chem Lett. 2013;23:6447–6454.
  • Mitani Y, Akashiba H, Saita K, et al. Pharmacological characterization of the novel γ-secretase modulator AS2715348, a potential therapy for Alzheimer’s disease, in rodents and nonhuman primates. Neuropharmacology. 2014;79:412–419.
  • Pettersson M, Johnson DS, Subramanyam C, et al. Design, synthesis, and pharmacological evaluation of a novel series of pyridopyrazine-1,6-dione γ-secretase modulators. J Med Chem. 2014;57:1046–1062.
  • Soares HD, Gasior M, Toyn JH, et al. The γ-secretase modulator, BMS-932481, modulates Aβ peptides in the plasma and cerebrospinal fluid of healthy volunteers. J Pharmacol Exp Ther. 2016;358:138–150.
  • Blain JF, Bursavich MG, Freeman EA, et al. Characterization of FRM-36143 as a new γ-secretase modulator for the potential treatment of familial Alzheimer’s disease. Alzheimers Res Ther. 2016;8:34.
  • Pettersson M, Johnson DS, Humphrey JM, et al. Design of pyridopyrazine-1,6-dione γ-secretase modulators that align potency, MDR efflux ratio, and metabolic stability. ACS Med Chem Lett. 2015;6:596–601.
  • Takai T, Hoashi Y, Tomata Y, et al. Discovery of novel 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridine derivatives as γ-secretase modulators. Bioorg Med Chem Lett. 2015;25:4245–4249.
  • Pohland M, Hagl S, Pellowska M, et al. MH84: a novel γ-secretase modulator/PPARγ agonist- improves mitochondrial dysfunction in a cellular model of Alzheimer’s disease. Neurochem Res. 2016;14:231–232.
  • Toyn JH, Boy KM, Raybon J, et al. Robust translation of γ-secretase modulator pharmacology across preclinical species and human subjects. J Pharmacol Exp Ther. 2016;358:125–137.
  • Takai T, Koike T, Honda E, et al. Design and synthesis of piperazine derivatives as a novel class of γ-secretase modulators that selectively lower Aβ42 production. Bioorg Med Chem. 2015;23:1923–1934.
  • Jung JI, Price AR, Ladd TB, et al. Cholestenoic acid, an endogenous cholesterol metabolite, is a potent γ-secretase modulator. ‎Mol Neurodegener. 2015;10:29.
  • Scannevin RH, Chollate S, Brennan MS, et al. BIIB042, a novel γ-secretase modulator, reduces amyloidogenic Aβ isoforms in primates and rodents and plaque pathology in a mouse model of Alzheimer’s disease. Neuropharmacology. 2016;103:57–68.
  • Rynearson KD, Buckle RN, Barnes KD, et al. Design and synthesis of aminothiazole modulators of the gamma-secretase enzyme. Bioorg Med Chem Lett. 2016;26:3928–3937.
  • Pettersson M, Johnson DS, Rankic DA, et al. Discovery of cyclopropyl chromane-derived pyridopyrazine-1,6-dione γ-secretase modulators with robust central efficacy. Med Chem Commun. 2017;8:730–743.
  • Takai T, Koike T, Nakamura M, et al. Discovery of novel 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyridine derivatives as γ-secretase modulators (Part 2). Bioorg Med Chem. 2016;24:3192–31206.
  • Shi J, Zuev D, Xu L, et al. Design and optimization of tricyclic gamma-secretase modulators. Bioorg Med Chem Lett. 2016;26:1498–1502.
  • Kounnasa MZ, Lane-Donovanb C, Nowakowskic DW, et al. NGP 555, a γ-secretase modulator, lowers the amyloid biomarker, Aβ42, in cerebrospinal fluid while preventing Alzheimer’s disease cognitive decline in rodents. Alzheimers Dement. 2017;3:65–73.
  • Rammes G, Gravius A, Ruitenberg M, et al. MRZ-99030 – a novel modulator of Aβ aggregation: II – reversal of Aβ oligomer-induced deficits in long-term potentiation (LTP) and cognitive performance in rats and mice. Neuropharmacology. 2015;92:170–182.
  • Borgegard T, Jureus A, Olsson F, et al. First and second generation γ-secretase modulators (GSMs) modulate amyloid-β (Aβ) peptide production through different mechanisms. J Biol Chem. 2012;287:11810–11819.
  • Crump CJ, Johnson DS, Li YM. Development and mechanism of γ-secretase modulators for Alzheimer’s disease. Biochemistry. 2013;52:3197–3216.
  • Jumpertz T, Rennhack A, Ness J, et al. Presenilin is the molecular target of acidic γ-secretase modulators in living cells. PLOS One. 2012;7:e30484.
  • Takeo K, Tanimura S, Shinoda T, et al. Allosteric regulation of γ-secretase activity by a phenylimidazole-type γ-secretase modulator. Proc Natl Acad Sci USA. 2014;111:10544–10549.
  • Wagner SL, Zhang C, Cheng S, et al. Soluble γ-secretase modulators selectively inhibit the production of the 42-amino acid amyloid β peptide variant and augment the production of multiple carboxy-truncated amyloid β species. Biochemistry. 2014;53:702–713.
  • Hubbs JL, Fuller NO, Austin WF, et al. Optimization of a natural product-based class of γ-secretase modulators. J Med Chem. 2012;55:9270–9282.
  • Hayashi I, Takatori S, Urano Y, et al. Neutralization of the γ-secretase activity by monoclonal antibody against extracellular domain of nicastrin. Oncogene. 2012;31:787–798.
  • Ferreira ST, Lourenco MV, Oliveira MM, et al. Soluble amyloid-β oligomers as synaptotoxins leading to cognitive impairment in Alzheimer’s disease. Front Cell Neurosci. 2015;9:191.
  • Esparza TJ, Zhao H, Cirrito JR, et al. Amyloid-beta oligomerization in Alzheimer dementia versus high-pathology controls. Ann Neurol. 2013;73:104–119.
  • Takeda S, Hashimoto T, Roe AD, et al. Brain interstitial oligomeric amyloid β increases with age and is resistant to clearance from brain in a mouse model of Alzheimer’s disease. Faseb J. 2013;27:3239–3248.
  • Parajuli B, Sonobe Y, Horiuchi H, et al. Oligomeric amyloid β induces IL-1β processing via production of ROS: implication in Alzheimer’s disease. Cell Death Dis. 2013;4:e975.
  • Mohamed LA, Qosa H, Kaddoumi A. Age-related decline in brain and hepatic clearance of amyloid-beta is rectified by the cholinesterase inhibitors donepezil and rivastigmine in rats. ACS Chem Neurosci. 2015;6:725–736.
  • Deane R, Singh I, Sagare AP, et al. A multimodal RAGE-specific inhibitor reduces amyloid β-mediated brain disorder in a mouse model of Alzheimer disease. J Clin Invest. 2012;122:1377–1392.
  • Um JW, Nygaard HB, Heiss JK, et al. Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci. 2012;15:1227–1235.
  • Frenkel D, Wilkinson K, Zhao L, et al. Scara1 deficiency impairs clearance of soluble amyloid-β by mononuclear phagocytes and accelerates Alzheimer’s-like disease progression. Nat Commun. 2013;4:2030.
  • Yuyama K, Sun H, Mitsutake S, et al. Sphingolipid-modulated exosome secretion promotes clearance of amyloid-β by microglia. J Biol Chem. 2012;287:10977–10989.
  • Michaud JP, Bellavance MA, Prefontaine P, et al. Real-time in vivo imaging reveals the ability of monocytes to clear vascular amyloid beta. Cell Rep. 2013;5:646–653.
  • Zhang X, Wang B, O’Callaghan P, et al. Heparanase overexpression impairs inflammatory response and macrophage-mediated clearance of amyloid-β in murine brain. Acta Neuropathol. 2012;124:465–478.
  • Gontier G, George C, Chaker Z, et al. Blocking IGF signaling in adult neurons alleviates Alzheimer’s disease pathology through amyloid-β clearance. J Neurosci. 2015;35:11500–11513.
  • 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;110:1807–1816.
  • Mulder SD, Veerhuis R, Blankenstein MA, et al. The effect of amyloid associated proteins on the expression of genes involved in amyloid-β clearance by adult human astrocytes. Exp Neurol. 2012;233:373–379.
  • Kanekiyo T, Cirrito JR, Liu CC, et al. Neuronal clearance of amyloid-β by endocytic receptor. J Neurosci. 2013;33:19276–19283.
  • Koffie RM, Hashimoto T, Tai HC, et al. Apolipoprotein E4 effects in Alzheimer’s disease are mediated by synaptotoxic oligomeric amyloid-β. Brain. 2012;235:2155–2168.
  • Castellano JM, Deane R, Gottesdiener AJ, et al. Low-density lipoprotein receptor overexpression enhances the rate of brain-to-blood Aβ clearance in a mouse model of β-amyloidosis. Proc Natl Acad Sci USA. 2012;109:15502–15507.
  • Song Q, Huang M, Yao L, et al. Lipoprotein-based nanoparticles rescue the memory loss of mice with Alzheimer’s disease by accelerating the clearance of amyloid-beta. ACS Nano. 2014;8:2345–2359.
  • Kuo YC, Rajesh R. Targeted delivery of rosmarinic acid across the blood–brain barrier for neuronal rescue using polyacrylamide-chitosan-poly(lactide-co-glycolide) nanoparticles with surface cross-reacting material 197 and apolipoprotein E. Int J Pharm. 2017;528:228–241.
  • Kuo YC, Liu YC. Cardiolipin-incorporated liposomes with surface CRM197 for enhancing neuronal survival against neurotoxicity. Int J Pharm. 2014;473:334–344.
  • Brambilla D, Verpillot R, Droumaguet BL, et al. PEGylated nanoparticles bind to and alter amyloid-beta peptide conformation: toward engineering of functional nanomedicines for Alzheimer’s disease. ACS Nano. 2012;6:5897–5908.
  • Ge JF, Qiao JP, Qi CC, et al. The binding of resveratrol to monomer and fibril amyloid beta. Neurochem Int. 2012;7:1192–1201.
  • Wang Y, Xia Z, Xu JR, et al. α-mangostin, a polyphenolic xanthone derivative from mangosteen, attenuates β-amyloid oligomers-induced neurotoxicity by inhibiting amyloid aggregation. Neuropharmacology. 2012;62:871–881.
  • Hohsfield LA, Humpel C. Intravenous infusion of monocytes isolated from 2-week-old mice enhances clearance of beta-amyloid plaques in an Alzheimer mouse model. PLOS One. 2015;10:e0121930.
  • Dinkins MB, Dasgupta S, Wang G, et al. Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer’s disease. Neurobiol Aging. 2014;35:1792–1800.
  • Xiang X, Werner G, Bohrmann B, et al. TREM2 deficiency reduces the efficacy of immunotherapeutic amyloid clearance. EMBO Mol Med. 2016;8:992–1004.
  • DeMattos RB, Lu J, Tang Y, et al. A plaque-specific antibody clears existing β-amyloid plaques in Alzheimer’s disease mice. Neuron. 2012;76:908–920.
  • Ostrowitzki S, Deptula D, Thurfjell L, et al. Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Arch Neurol. 2012;69:198–207.
  • Humpel C. Organotypic vibrosections from whole brain adult Alzheimer mice (overexpressing amyloid-precursor-protein with the Swedish-Dutch-Iowa mutations) as a model to study clearance of beta-amyloid plaques. Front Aging Neurosci. 2015;7:47.
  • Xiao Q, Yan P, Ma X, et al. Enhancing Astrocytic lysosome biogenesis facilitates Aβ clearance and attenuates amyloid plaque pathogenesis. J Neurosci. 2014;34:9607–9620.
  • Wu J, Bei B, Yang H, et al. Activation of the CB2 receptor system reverses amyloid-induced memory deficiency. Neurobiol Aging. 2013;34:791–804.
  • Fox BM, Beck HP, Roveto PM, et al. A selective prostaglandin E2 receptor subtype 2 (EP2) antagonist increases the macrophage-mediated clearance of amyloid-beta plaques. J Med Chem. 2015;58:5256–5273.
  • Sevigny J, Chiao P, Williams L, et al. Aducanumab (BIIB037), an anti-amyloid beta monoclonal antibody, in patients with prodromal or mild Alzheimer’s disease: interim results of a randomized, double-blind, placebo-controlled, phase 1b study. Alzheimers Dement. 2015;11:P277.
  • Piazza F, Winblad B. Amyloid-related imaging abnormalities (ARIA) in immunotherapy trials for Alzheimer’s disease: need for prognostic biomarkers? J Alzheimers Dis. 2016;52:417–420.
  • Lee LL, Ha HH, Chang YT, et al. Discovery of amyloid‐beta aggregation inhibitors using an engineered assay for intracellular protein folding and solubility. Protein Sci. 2009;18:277–286.
  • Villar-Pique A, Espargaro A, Sabate R, et al. Using bacterial inclusion bodies to screen for amyloid aggregation inhibitors. Microb Cell Fact. 2012;11:55.
  • Li X, Zhang X, Ladiwala ARA, et al. Mechanisms of transthyretin inhibition of β-amyloid aggregation in vitro. J Neurosci. 2013;33:19423–19433.
  • Gil-Bea F, Akterin S, Persson T, et al. Thioredoxin-80 is a product of alpha-secretase cleavage that inhibits amyloid-beta aggregation and is decreased in Alzheimer’s disease brain. EMBO Mol Med. 2012;4:1097–1111.
  • Churches QI, Caine J, Epa VC, et al. Naturally occurring polyphenolic inhibitors of amyloid beta aggregation. Bioorganic Med Chem Lett. 2014;24:3108–3112.
  • Lai AY, McLaurin J. Inhibition of amyloid-beta peptide aggregation rescues the autophagic deficits in the TgCRND8 mouse model of Alzheimer disease. Biochim Biophys Acta Mol Basis Dis. 2012;1822:1629–1637.
  • Luo Y, Vali S, Sun S, et al. Aβ42-binding peptoids as amyloid aggregation inhibitors and detection ligands. ACS Chem Neurosci. 2013;4:952–962.
  • Mansson C, Arosio P, Hussein R, et al. Interaction of the molecular chaperone DNAJB6 with growing amyloid-beta 42 (Aβ42) aggregates leads to sub-stoichiometric inhibition of amyloid formation. J Biol Chem. 2014;289:31066–31076.
  • Yan JW, Li YP, Ye WJ, et al. Design, synthesis and evaluation of isaindigotone derivatives as dual inhibitors for acetylcholinesterase and amyloid beta aggregation. Bioorg Med Chem. 2012;20:2527–2534.
  • Wang SW, Wang YJ, Su YJ, et al. Rutin inhibits β-amyloid aggregation and cytotoxicity, attenuates oxidative stress, and decreases the production of nitric oxide and proinflammatory cytokines. Neurotoxicology. 2012;33:482–490.
  • Medina M, Avila J. New perspectives on the role of tau in Alzheimer’s disease. Implications for therapy. Biochem Pharmacol. 2014;88:540–547.
  • Wischik CM, Harrington CR, Storey JMD. Tau-aggregation inhibitor therapy for Alzheimer’s disease. Biochem Pharmacol. 2014;88:529–539.
  • Chiotis K, Saint-Aubert L, Savitcheva I, et al. Imaging in-vivo tau pathology in Alzheimer’s disease with THK5317 PET in a multimodal paradigm. Eur J Nucl Med Mol Imaging. 2016;43:1686–1699.
  • Pooler AM, Noble W, Hanger DP. A role for tau at the synapse in Alzheimer’s disease pathogenesis. Neuropharmacology. 2014;76:1–8.
  • Wu JW, Hussaini SA, Bastille IM, et al. Neuronal activity enhances tau propagation and tau pathology in vivo. Nat Neurosci. 2016;19:1085–1092.
  • Mondragon-Rodriguez S, Perry G, Luna-Munoz J, et al. Phosphorylation of tau protein at sites Ser396–404 is one of the earliest events in Alzheimer’s disease and down syndrome. Neuropathol Appl Neurobiol. 2014;40:121–135.
  • Yarchoan M, Toledo JB, Lee EB, et al. Abnormal serine phosphorylation of insulin receptor substrate 1 is associated with tau pathology in Alzheimer’s disease and tauopathies. Acta Neuropathol. 2014;128:679–689.
  • Zhang Z, Song M, Liu X, et al. Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer’s disease. Nat Med. 2014;20:1254–1262.
  • Glodzik L, Rusinek H, Pirraglia E, et al. Blood pressure decrease correlates with tau pathology and memory decline in hypertensive elderly. Neurobiol Aging. 2014;35:64–71.
  • Qiu L, Ng G, Tan EK, et al. Chronic cerebral hypoperfusion enhances tau hyperphosphorylation and reduces autophagy in Alzheimer’s disease mice. Sci Rep. 2016;6:23964.
  • Spilsbury A, Miwa S, Attems J, et al. The role of telomerase protein TERT in Alzheimer’s disease and in tau-related pathology in vitro. J Neurosci. 2015;35:1659–1674.
  • Domise M, Didier S, Marinangeli C, et al. AMP-activated protein kinase modulates tau phosphorylation and tau pathology in vivo. Sci Rep. 2016;6:26758.
  • Tian M, Zhu D, Xie W, et al. Central angiotensin II-induced Alzheimer-like tau phosphorylation in normal rat brains. FEBS Lett. 2012;586:3737–3745.
  • Gassowska M, Czapski GA, Pająk B, et al. Extracellular α-synuclein leads to microtubule destabilization via GSK-3β-dependent tau phosphorylation in PC12 cells. PLOS One. 2014;9:e94259.
  • Rueli RH, Torres DJ, Dewing AS, et al. Selenoprotein S reduces endoplasmic reticulum stress-induced phosphorylation of tau: potential role in selenate mitigation of tau pathology. J Alzheimers Dis. 2017;55:749–762.
  • Platt TL, Beckett TL, Kohler K, et al. Obesity, diabetes, and leptin resistance promote tau pathology in a mouse model of disease. Neuroscience. 2016;315:162–174.
  • Vasconcelos B, Stancu IC, Buist A, et al. Heterotypic seeding of tau fibrillization by pre-aggregated Abeta provides potent seeds for prion-like seeding and propagation of tau-pathology in vivo. Acta Neuropathol. 2016;131:549–569.
  • Liu S, Breitbart A, Sun Y, et al. Blocking the apolipoprotein E/amyloid β interaction in triple transgenic mice ameliorates Alzheimer’s disease related amyloid β and tau pathology. J Neurochem. 2014;128:577–591.
  • Salta E, Sierksma A, Eynden EV, et al. miR‐132 loss de‐represses ITPKB and aggravates amyloid and TAU pathology in Alzheimer’s brain. EMBO Mol Med. 2016;8:987–1113.
  • Chen LM, Xiong YS, Kong FL, et al. Neuroglobin attenuates Alzheimer-like tau hyperphosphorylation by activating Akt signaling. J Neurochem. 2012;120:157–164.
  • Chen Z, Chen B, Xu WF, et al. Effects of PTEN inhibition on regulation of tau phosphorylation in an okadaic acid-induced neurodegeneration model. Int J Dev Neurosci. 2012;30:411–419.
  • Fu H, Chen H, Wang C, et al. Flurbiprofen, a cyclooxygenase inhibitor, protects mice from hepatic ischemia/reperfusion injury by inhibiting GSK-3β signaling and mitochondrial permeability transition. Mol Med. 2012;18:1128–1135.
  • Wu J, Li J, Hu H, et al. Rho-Kinase inhibitor, fasudil, prevents neuronal apoptosis via the Akt activation and PTEN inactivation in the ischemic penumbra of rat brain. Cell Mol Neurobiol. 2012;32:1187–1197.
  • Modi KK, Jana A, Ghosh S, et al. A physically-modified saline suppresses neuronal apoptosis, attenuates tau phosphorylation and protects memory in an animal model of Alzheimer’s disease. PLOS One. 2014;12:e0180602.
  • Chen S, An FM, Yin L, et al. Glucagon-like peptide-1 protects hippocampal neurons against advanced glycation end product-induced tau hyperphosphorylation. Neuroscience. 2014;256:137–146.
  • Li L, Zhang ZF, Holscher C, 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. 2012;674:280–286.
  • Ali T, Yoon GH, Shah SA, et al. Osmotin attenuates amyloid beta-induced memory impairment, tau phosphorylation and neurodegeneration in the mouse hippocampus. Sci Rep. 2015;5:11708.
  • Gu MY, Kim J, Yang HO. The neuroprotective effects of justicidin A on amyloid beta25–35-induced neuronal cell death through inhibition and induction of autophagy in SH-SY5Y cells. Neurochem Res. 2016;41:1458–1467.
  • van der Harg JM, Nolle A, Zwart R, et al. The unfolded protein response mediates reversible tau phosphorylation induced by metabolic stress. Cell Death Dis. 2014;5:e1393.
  • Radford H, Moreno JA, Verity N, et al. PERK inhibition prevents tau-mediated neurodegeneration in a mouse model of frontotemporal dementia. Acta Neuropathol. 2015;130:633–642.
  • Absalon S, Kochanek DM, Raghavan V, et al. MiR-26b, upregulated in Alzheimer’s disease, activates cell cycle entry, tau-phosphorylation, and apoptosis in postmitotic neurons. J Neurosci. 2013;33:14645–14659.
  • Caccamo A, Magri A, Medina DX, et al. mTOR regulates tau phosphorylation and degradation: implications for Alzheimer’s disease and other tauopathies. Aging Cell. 2013;12:370–380.
  • Kandimalla R, Manczak M, Fry D, et al. Reduced dynamin-related protein 1 protects against phosphorylated tau-induced mitochondrial dysfunction and synaptic damage in Alzheimer’s disease. Hum Mol Gen. 2016;25:4881–4897.
  • Yang CC, Kuai XX, Li YL, et al. Cornel iridoid glycoside attenuates tau hyperphosphorylation by inhibition of PP2A demethylation. J Evid Based Complementary Altern Med 2013;2013: Article ID 108486.
  • Laurent C, Eddarkaoui S, Derisbourg M, et al. Beneficial effects of caffeine in a transgenic model of Alzheimer’s disease-like tau pathology. Neurobiol Aging. 2014;35:2079–2090.
  • Yang S, Gong Q, Wu Q, et al. Alkaloids enriched extract from Dendrobium nobile Lindl. attenuates tau protein hyperphosphorylation and apoptosis induced by lipopolysaccharide in rat brain. Phytomedicine. 2014;21:712–716.
  • Chen J, Deng X, Liu N, et al. Quercetin attenuates tau hyperphosphorylation and improves cognitive disorder via suppression of ER stress in a manner dependent on AMPK pathway. J Funct Foods. 2016;22:463–476.
  • Wischik CM, Staff RT, Wischik DJ, et al. Tau aggregation inhibitor therapy: an exploratory phase 2 study in mild or moderate Alzheimer’s disease. J Alzheimers Dis. 2015;44:705–720.
  • Akoury E, Pickhardt M, Gajda M, et al. Mechanistic basis of phenothiazine-driven inhibition of tau aggregation. Angew Chem Int Ed Engl. 2013;52:3511–3515.
  • Baddeley TC, McCaffrey J, Storey JMD, et al. Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimer’s disease. J Pharmacol Exp Ther. 2015;352:110–118.
  • Graham DL, Gray AJ, Joyce JA, et al. Increased O-GlcNAcylation reduces pathological tau without affecting its normal phosphorylation in a mouse model of tauopathy. Neuropharmacology. 2014;79:307–313.
  • Cook C, Carlomagno Y, Gendron TF, et al. Acetylation of the KXGS motifs in tau is a critical determinant in modulation of tau aggregation and clearance. Hum Mol Gen. 2014;23:104–116.
  • Wobst HJ, Sharma A, Diamond MI, et al. The green tea polyphenol (–)-epigallocatechin gallate prevents the aggregation of tau protein into toxic oligomers at substoichiometric ratios. FEBS Lett. 2015;589:77–83.
  • Mohamed T, Hoang T, Jelokhani-Niaraki M, et al. Tau-derived-hexapeptide 306VQIVYK311 aggregation inhibitors: nitrocatechol moiety as a pharmacophore in drug design. ACS Chem Neurosci. 2013;4:1559–1570.
  • Wang CK, Northfield SE, Huang YH, et al. Inhibition of tau aggregation using a naturally-occurring cyclic peptide scaffold. Eur J Med Chem. 2016;109:342–349.
  • McEwan WA, Falcon B, Vaysburd M, et al. Cytosolic Fc receptor TRIM21 inhibits seeded tau aggregation. Proc Natl Acad Sci USA. 2017;114:574–579.
  • Asai H, Ikezu S, Tsunoda S, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci. 2015;18:1584–1593.
  • Rao MV, McBrayer MK, Campbell J, et al. Specific calpain inhibition by calpastatin prevents tauopathy and neurodegeneration and restores normal lifespan in tau P301L mice. J Neurosci. 2014;4:9222–9234.
  • McCormick AV, Wheeler JM, Guthrie CR, et al. Dopamine D2 receptor antagonism suppresses tau aggregation and neurotoxicity. Biol Psychiatry. 2013;73:464–471.
  • Han DH, Na HK, Choi WH, et al. Direct cellular delivery of human proteasomes to delay tau aggregation. Nat Commun. 2014;5:5633.
  • Ballatore C, Crowe A, Piscitelli F, et al. Aminothienopyridazine inhibitors of tau aggregation: evaluation of structure–activity relationship leads to selection of candidates with desirable in vivo properties. Bioorg Med Chem. 2012;20:4451–4461.
  • Okuda M, Hijikuro I, Fujita Y, et al. PE859, a novel tau aggregation inhibitor, reduces aggregated tau and prevents onset and progression of neural dysfunction in vivo. PLOS One. 2015;10:e0117511.
  • Santa-Maria I, Diaz-Ruiz C, Ksiezak-Reding H, et al. GSPE interferes with tau aggregation in vivo: implication for treating tauopathy. Neurobiol Aging. 2012;33:2072–2081.
  • Haque MM, Kim D, Yu YH, et al. Inhibition of tau aggregation by a rosamine derivative that blocks tau intermolecular disulfide cross-linking. Amyloid. 2014;21:185–190.
  • Paranjape SR, Chiang YM, Sanchez JF, et al. Inhibition of tau aggregation by three aspergillus nidulans secondary metabolites: 2,ω-dihydroxyemodin, asperthecin, and asperbenzaldehyde. Planta Med. 2014;80:77–85.
  • Yao J, Gao X, Sun W, et al. Molecular hairpin: a possible model for inhibition of tau aggregation by tannic acid. Biochemistry. 2013;52:1893–1902.
  • Okuda M, Hijikuro I, Fujita Y, et al. Design and synthesis of curcumin derivatives as tau and amyloid β dual aggregation inhibitors. Bioorg Med Chem Lett. 2016;26:5024–5028.
  • Frenkel-Pinter M, Tal S, Scherzer-Attali R, et al. Cl-NQTrp alleviates tauopathy symptoms in a model organism through the inhibition of tau aggregation-engendered toxicity. Neurodegener Dis. 2017;17:73–82.
  • Polito VA, Li H, Martini‐Stoica H, et al. Selective clearance of aberrant tau proteins and rescue of neurotoxicity by transcription factor EB. EMBO Mol Med. 2014;6:1105–1114.
  • Polydoro M, de Calignon A, Suarez-Calvet M, et al. Reversal of neurofibrillary tangles and tau-associated phenotype in the rTgTauEC model of early Alzheimer’s disease. J Neurosci. 2013;33:13300–13311.
  • Tennstaedt A, Popsel S, Truebestein L, et al. Human high temperature requirement serine protease a1 (HTRA1) degrades tau protein aggregates. J Biol Chem. 2012;287:20931–20941.
  • Walls KC, Ager RR, Vasilevko V, et al. p-tau immunotherapy reduces soluble and insoluble tau in aged 3xTg-AD mice. Neurosci Lett. 2014;575:96–100.
  • Yanamandra K, Jiang H, Mahan TE, et al. Anti-tau antibody reduces insoluble tau and decreases brain atrophy. Ann Clin Transl Neurol. 2015;2:278–288.
  • Congdon EE, Gu J, Sait HBR, et al. Antibody uptake into neurons occurs primarily via clathrin-dependent Fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance. J Biol Chem. 2013;288:35452–35465.
  • Esteves-Villanueva JO, Trzeciakiewicz H, Loeffler DA, et al. Effects of tau domain-specific antibodies and intravenous immunoglobulin on tau aggregation and aggregate degradation. Biochemistry. 2015;54:293–302.
  • Herrup K. The case for rejecting the amyloid cascade hypothesis. Nat Neurosci. 2015;18:794–799.
  • Dorszewska J, Prendecki M, Oczkowska A, et al. Molecular basis of familial and sporadic Alzheimer’s Disease. Curr Alzheimer Res. 2016;13:952–963.
  • Terry RD. The pathogenesis of Alzheimer disease: an alternative to the amyloid hypothesis. J Neuropathol Exp Neurol. 1996;55:1023–1025.
  • Neve RL, Robakis NK. Alzheimer’s disease: a re-examination of the amyloid hypothesis. Trends Neurosci. 1998;21:15–19.
  • Wes PD, Sayed FA, Bard F, et al. Targeting microglia for the treatment of Alzheimer’s disease. Glia. 2016;64:1710–1732.
  • Itzhaki RF. Herpes simplex virus type 1 and Alzheimer’s disease: increasing evidence for a major role of the virus. Front Aging Neurosci. 2014;11:1–9.
  • Wozniak MA, Mee AP, Itzhaki RF. Herpes simplex virus type 1 DNA is located within Alzheimer’s disease amyloid plaque. J Pathol. 2009;217:131–138.
  • Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8:595–608.
  • Wozniak MA, Frost AL, Preston CM, et al. Antivirals reduce the formation of key Alzheimer’s disease molecules in cell cultures acutely infected with herpes simplex virus type 1. PLoS One. 2011;6:e25152.
  • Cunnane SC, Courchesne‐Loyer A, St-Pierre V, et al. Can ketones compensate for deteriorating brain glucose uptake during aging? Implications for the risk and treatment of Alzheimer’s disease. Ann NY Acad Sci. 2016;1367:12–20.
  • Gejl M, Gjedde A, Egefjord L, et al. In Alzheimer’s disease, 6-month treatment with GLP-1 analog prevents decline of brain glucose metabolism: randomized, placebo-controlled, double-blind clinical trial. Front Aging Neurosci. 2016;8:1–10.
  • Perry T, Lahiri DK, Sambamurti K, et al. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. Neurosci Res. 2003;72:603–612.
  • McClean PL, Gault VA, Harriott P, et al. Glucagon-like peptide-1 analogues enhance synaptic plasticity in the brain: a link between diabetes and Alzheimer’s disease. Eur J Pharmacol. 2010;630:158–162.
  • Xu W, Yang Y, Yuan G, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, reduces Alzheimer disease-associated tau hyperphosphorylation in the hippocampus of rats with type 2 diabetes. J Investig Med. 2015;63:267–272.
  • Hunter K, Holscher C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012;13:33.
  • Holscher C. Central effects of GLP-1: new opportunities for treatments of neurodegenerative diseases. J Endocrinol. 2014;221:T31–T41.
  • Talbot K, Wang HY. The nature, significance, and glucagon-like peptide-1 analog treatment of brain insulin resistance in Alzheimer’s disease. Alzheimers Dement. 2014;10:S12–S25.
  • Klinge PM, Harmening K, Miller MC, et al. Encapsulated native and glucagon-like peptide-1 transfected human mesenchymal stem cells in a transgenic mouse model of Alzheimer’s disease. Neurosci Lett. 2011;497:6–10.
  • Heile A. Clinical transplantation of stem cell therapy in traumatic brain injury: the potential of encapsulated mesenchymal stem cell biodelivery of glucagon-like peptide-1. Dialogues Clin Neurosci. 2011;13:279–286.
  • Chen Y, Zhang J, Zhang B, et al. Targeting insulin signaling for the treatment of Alzheimer’s disease. Curr Top Med Chem. 2016;16:485–492.
  • Kosaraju J, Gali CC, Khatwal RB, et al. Saxagliptin: a dipeptidyl peptidase-4 inhibitor ameliorates streptozotocin induced Alzheimer’s disease. Neuropharmacology. 2013;72:291–300.
  • Kosaraju J, Madhunapantula SRV, Chinni S, et al. Dipeptidyl peptidase-4 inhibition by Pterocarpus marsupium and Eugenia jambolana ameliorates streptozotocin induced Alzheimer’s disease. Behav Brain Res. 2014;267:55–65.
  • Gault VA, Lennox R, Flatt PR. Sitagliptin, a dipeptidyl peptidase-4 inhibitor, improves recognition memory, oxidative stress and hippocampal neurogenesis and upregulates key genes involved in cognitive decline. Diabetes Obes Metab. 2015;17:403–413.
  • Yang D, Nakajo Y, Iihara K, et al. Alogliptin, a dipeptidylpeptidase-4 inhibitor, for patients with diabetes mellitus type 2, induces tolerance to focal cerebral ischemia in non-diabetic, normal mice. Brain Res. 2013;1517:104–113.
  • Holst JJ, Deacon CF. Glucagon-like peptide 1 and inhibitors of dipeptidyl peptidase IV in the treatment of type 2 diabetes mellitus. Curr Opin Pharmacol. 2004;6:589–596.
  • Matteucci E, Giampietro O. Mechanisms of neurodegeration in type 2 diabetes and the neuroprotective potential of dipeptidyl peptidase 4 inhibitors. Curr Med Chem. 2015;22:1573–1581.
  • Mander BA, Winer JR, Jagust WJ, et al. Sleep: a novel mechanistic pathway, biomarker, and treatment target in the pathology of Alzheimer’s disease? Trends Neurosci. 2016;39:552–566.
  • Longo FM, Massa SM. Small-molecule modulation of neurotrophin receptors: a strategy for the treatment of neurological disease. Nat Rev Drug Discov. 2013;12:507–525.
  • Song JH, Yu JT, Tan L. Brain-derived neurotrophic factor in Alzheimer’s disease: risk, mechanisms, and therapy. Mol Neurobiol. 2015;52:1477–1493.
  • Budni J, Bellettini-Santos T, Mina F, et al. The involvement of BDNF, NGF and GDNF in aging and Alzheimer’s disease. Aging Dis. 2015;6:331–341.
  • Honea RA, Cruchaga C, Perea RD, et al. Characterizing the role of brain derived neurotrophic factor genetic variation in Alzheimer’s disease neurodegeneration. PLoS ONE. 2013;8:e76001.
  • Zhang L, Lian Y, Chen Y, et al. Brain-derived neurotrophic factor ameliorates learning deficits in a rat model of Alzheimer’s disease induced by Aβ1–42. PLoS ONE. 2015;10:e0122415.
  • Jiao SS, Shen LL, Zhu C, et al. Brain-derived neurotrophic factor protects against tau-related neurodegeneration of Alzheimer’s disease. Transl Psychiatry. 2016;6:e907.
  • Prior M, Dargusch R, Ehren JL, et al. The neurotrophic compound J147 reverses cognitive impairment in aged Alzheimer’s disease mice. Alzheimers Res Ther. 2013;5:25.
  • Kazim SF, Blanchard J, Dai CL, et al. Disease modifying effect of chronic oral treatment with a neurotrophic peptidergic compound in a triple transgenic mouse model of Alzheimer’s disease. Neurobiol Dis. 2014;17:110–130.
  • Coelho FG, Vital TM, Stein AM, et al. Acute aerobic exercise increases brain-derived neurotrophic factor levels in elderly with Alzheimer’s disease. J Alzheimers Dis. 2014;39:401–408.
  • Angelova A, Angelov B, Drechsler M, et al. Neurotrophin delivery using nanotechnology. Drug Discov Today. 2013;18:1263–1271.
  • Kuo YC, Chen CL, Rajesh R. Optimized liposomes with transactivator of transcription peptide and anti-apoptotic drugs to target hippocampal neurons and prevent tau-hyperphosphorylated neurodegeneration. Acta Biomater. 2019;87:207–222.

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.