Drug Development for Alzheimer’s Disease: Microglia Induced Neuroinflammation as a Target?
Abstract
:1. Introduction
2. Cause of Alzheimer’s Disease
3. Microglia Induced Neuroinflammation in Alzheimer’s Disease
4. Drugs for Alzheimer’s Disease Treatment
4.1. Current Drugs
4.2. Drugs Targeting Amyloidogenic Route
4.2.1. α-Secretase Enhancer
4.2.2. β-Secretase Inhibitor
4.2.3. γ-Secretase Inhibitor
4.2.4. Aβ Aggregation Inhibitor
4.2.5. Aβ Vaccines
4.2.6. Aβ Antibodies
4.3. Drugs Targeting Tau Protein
4.4. Drugs Targeting Inflammation
4.4.1. Non-Steroidal Anti-Inflammatory Drugs
4.4.2. Other Drugs Targeting Inflammation
5. Conclusive Remarks
Author Contributions
Funding
Conflicts of Interest
References
- Burns, A.; Iliffe, S. Alzheimer’s disease. BMJ 2009, 338, b158. [Google Scholar] [CrossRef] [PubMed]
- Mendez, M.F. Early-onset Alzheimer’s disease: Nonamnestic subtypes and type 2 AD. Arch. Med. Res. 2012, 43, 677–685. [Google Scholar] [CrossRef] [PubMed]
- Alzheimer’s Association. 2016 Alzheimer’s disease facts and figures. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2016, 12, 459–509. [Google Scholar] [CrossRef]
- Mielke, M.M.; Vemuri, P.; Rocca, W.A. Clinical epidemiology of Alzheimer’s disease: Assessing sex and gender differences. Clin. Epidemiol. 2014, 6, 37–48. [Google Scholar] [CrossRef] [PubMed]
- Hebert, L.E.; Weuve, J.; Scherr, P.A.; Evans, D.A. Alzheimer disease in the United States (2010-2050) estimated using the 2010 census. Neurology 2013, 80, 1778–1783. [Google Scholar] [CrossRef] [Green Version]
- Tom, S.E.; Hubbard, R.A.; Crane, P.K.; Haneuse, S.J.; Bowen, J.; McCormick, W.C.; McCurry, S.; Larson, E.B. Characterization of dementia and Alzheimer’s disease in an older population: Updated incidence and life expectancy with and without dementia. Am. J. Public Health 2015, 105, 408–413. [Google Scholar] [CrossRef]
- Bonomo, S.M.; Rigamonti, A.E.; Giunta, M.; Galimberti, D.; Guaita, A.; Gagliano, M.G.; Muller, E.E.; Cella, S.G. Menopausal transition: A possible risk factor for brain pathologic events. Neurobiol. Aging 2009, 30, 71–80. [Google Scholar] [CrossRef]
- Pike, C.J.; Carroll, J.C.; Rosario, E.R.; Barron, A.M. Protective actions of sex steroid hormones in Alzheimer’s disease. Front. Neuroendocrinol. 2009, 30, 239–258. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, D.P.; Woolfrey, K.M.; Penzes, P. Insights into rapid modulation of neuroplasticity by brain estrogens. Pharmacol. Rev. 2013, 65, 1318–1350. [Google Scholar] [CrossRef]
- Stern, Y. Cognitive reserve in ageing and Alzheimer’s disease. Lancet Neurol. 2012, 11, 1006–1012. [Google Scholar] [CrossRef]
- Rocca, W.A.; Mielke, M.M.; Vemuri, P.; Miller, V.M. Sex and gender differences in the causes of dementia: A narrative review. Maturitas 2014, 79, 196–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berchtold, N.C.; Cotman, C.W. Evolution in the conceptualization of dementia and Alzheimer’s disease: Greco-Roman period to the 1960s. Neurobiol. Aging 1998, 19, 173–189. [Google Scholar] [CrossRef]
- Zhang, B.; Gaiteri, C.; Bodea, L.G.; Wang, Z.; McElwee, J.; Podtelezhnikov, A.A.; Zhang, C.; Xie, T.; Tran, L.; Dobrin, R.; et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 2013, 153, 707–720. [Google Scholar] [CrossRef]
- Carreiras, M.C.; Mendes, E.; Perry, M.J.; Francisco, A.P.; Marco-Contelles, J. The multifactorial nature of Alzheimer’s disease for developing potential therapeutics. Curr. Top. Med. Chem. 2013, 13, 1745–1770. [Google Scholar] [CrossRef] [PubMed]
- Arendt, T. Synaptic degeneration in Alzheimer’s disease. Acta Neuropathol. 2009, 118, 167–179. [Google Scholar] [CrossRef] [PubMed]
- Blennow, K.; de Leon, M.J.; Zetterberg, H. Alzheimer’s disease. Lancet 2006, 368, 387–403. [Google Scholar] [CrossRef]
- Campion, D.; Dumanchin, C.; Hannequin, D.; Dubois, B.; Belliard, S.; Puel, M.; Thomas-Anterion, C.; Michon, A.; Martin, C.; Charbonnier, F.; et al. Early-onset autosomal dominant Alzheimer disease: Prevalence, genetic heterogeneity, and mutation spectrum. Am. J. Hum. Genet. 1999, 65, 664–670. [Google Scholar] [CrossRef]
- Nussbaum, R.L. Genome-wide association studies, Alzheimer disease, and understudied populations. JAMA 2013, 309, 1527–1528. [Google Scholar] [CrossRef]
- Mahley, R.W.; Weisgraber, K.H.; Huang, Y. Apolipoprotein E4: A causative factor and therapeutic target in neuropathology, including Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2006, 103, 5644–5651. [Google Scholar] [CrossRef]
- Neu, S.C.; Pa, J.; Kukull, W.; Beekly, D.; Kuzma, A.; Gangadharan, P.; Wang, L.S.; Romero, K.; Arneric, S.P.; Redolfi, A.; et al. Apolipoprotein E Genotype and Sex Risk Factors for Alzheimer Disease: A Meta-analysis. JAMA Neurol. 2017, 74, 1178–1189. [Google Scholar] [CrossRef]
- Jonsson, T.; Stefansson, H.; Steinberg, S.; Jonsdottir, I.; Jonsson, P.V.; Snaedal, J.; Bjornsson, S.; Huttenlocher, J.; Levey, A.I.; Lah, J.J.; et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. N. Engl. J. Med. 2013, 368, 107–116. [Google Scholar] [CrossRef] [PubMed]
- Guerreiro, R.; Wojtas, A.; Bras, J.; Carrasquillo, M.; Rogaeva, E.; Majounie, E.; Cruchaga, C.; Sassi, C.; Kauwe, J.S.; Younkin, S.; et al. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med. 2013, 368, 117–127. [Google Scholar] [CrossRef] [PubMed]
- Abduljaleel, Z.; Al-Allaf, F.A.; Khan, W.; Athar, M.; Shahzad, N.; Taher, M.M.; Elrobh, M.; Alanazi, M.S.; El-Huneidi, W. Evidence of trem2 variant associated with triple risk of Alzheimer’s disease. PLoS ONE 2014, 9, e92648. [Google Scholar] [CrossRef] [PubMed]
- Jin, S.C.; Benitez, B.A.; Karch, C.M.; Cooper, B.; Skorupa, T.; Carrell, D.; Norton, J.B.; Hsu, S.; Harari, O.; Cai, Y.; et al. Coding variants in TREM2 increase risk for Alzheimer’s disease. Hum. Mol. Genet. 2014, 23, 5838–5846. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.; Liu, W.; Wang, X. TREM2 variants and risk of Alzheimer’s disease: A meta-analysis. Neurol. Sci. 2015, 36, 1881–1888. [Google Scholar] [CrossRef] [PubMed]
- Bezprozvanny, I.; Mattson, M.P. Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease. Trends Neurosci. 2008, 31, 454–463. [Google Scholar] [CrossRef] [PubMed]
- Bezprozvanny, I. Calcium signaling and neurodegenerative diseases. Trends Mol. Med. 2009, 15, 89–100. [Google Scholar] [CrossRef] [Green Version]
- Berridge, M.J. Dysregulation of neural calcium signaling in Alzheimer disease, bipolar disorder and schizophrenia. Prion 2013, 7, 2–13. [Google Scholar] [CrossRef] [Green Version]
- Berridge, M.J.; Bootman, M.D.; Roderick, H.L. Calcium signalling: Dynamics, homeostasis and remodelling. Nature Rev. Mol. Cell Biol. 2003, 4, 517–529. [Google Scholar] [CrossRef]
- Magi, S.; Castaldo, P.; Macri, M.L.; Maiolino, M.; Matteucci, A.; Bastioli, G.; Gratteri, S.; Amoroso, S.; Lariccia, V. Intracellular Calcium Dysregulation: Implications for Alzheimer’s Disease. BioMed Res. Int. 2016, 2016, 6701324. [Google Scholar] [CrossRef]
- Dreses-Werringloer, U.; Vingtdeux, V.; Zhao, H.; Chandakkar, P.; Davies, P.; Marambaud, P. CALHM1 controls the Ca(2)(+)-dependent MEK, ERK, RSK and MSK signaling cascade in neurons. J. Cell Sci. 2013, 126, 1199–1206. [Google Scholar] [CrossRef] [PubMed]
- Vingtdeux, V.; Chang, E.H.; Frattini, S.A.; Zhao, H.; Chandakkar, P.; Adrien, L.; Strohl, J.J.; Gibson, E.L.; Ohmoto, M.; Matsumoto, I.; et al. CALHM1 deficiency impairs cerebral neuron activity and memory flexibility in mice. Sci. Rep. 2016, 6, 24250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, B.; Wang, X.; Nunomura, A.; Moreira, P.I.; Lee, H.G.; Perry, G.; Smith, M.A.; Zhu, X. Oxidative stress signaling in Alzheimer’s disease. Curr. Alzheimer Res. 2008, 5, 525–532. [Google Scholar] [CrossRef] [PubMed]
- Onyango, I.G.; Khan, S.M. Oxidative stress, mitochondrial dysfunction, and stress signaling in Alzheimer’s disease. Curr. Alzheimer Res. 2006, 3, 339–349. [Google Scholar] [CrossRef] [PubMed]
- Liao, Y.; Dong, Y.; Cheng, J. The Function of the Mitochondrial Calcium Uniporter in Neurodegenerative Disorders. Int. J. Mol. Sci. 2017, 18. [Google Scholar] [CrossRef] [PubMed]
- Cheng, J.; Liao, Y.; Zhou, L.; Peng, S.; Chen, H.; Yuan, Z. Amplified RLR signaling activation through an interferon-stimulated gene-endoplasmic reticulum stress-mitochondrial calcium uniporter protein loop. Sci. Rep. 2016, 6, 20158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liao, Y.; Hao, Y.; Chen, H.; He, Q.; Yuan, Z.; Cheng, J. Mitochondrial calcium uniporter protein MCU is involved in oxidative stress-induced cell death. Protein Cell 2015, 6, 434–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lambert, J.C.; Ibrahim-Verbaas, C.A.; Harold, D.; Naj, A.C.; Sims, R.; Bellenguez, C.; DeStafano, A.L.; Bis, J.C.; Beecham, G.W.; Grenier-Boley, B.; et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 2013, 45, 1452–1458. [Google Scholar] [CrossRef] [PubMed]
- Francis, P.T.; Palmer, A.M.; Snape, M.; Wilcock, G.K. The cholinergic hypothesis of Alzheimer’s disease: A review of progress. J. Neurol. Neurosurg. Psychiatry 1999, 66, 137–147. [Google Scholar] [CrossRef] [PubMed]
- Martorana, A.; Esposito, Z.; Koch, G. Beyond the cholinergic hypothesis: Do current drugs work in Alzheimer’s disease? CNS Neurosci. Ther. 2010, 16, 235–245. [Google Scholar] [CrossRef] [PubMed]
- Barage, S.H.; Sonawane, K.D. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer’s disease. Neuropeptides 2015, 52, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Karran, E.; Mercken, M.; De Strooper, B. The amyloid cascade hypothesis for Alzheimer’s disease: An appraisal for the development of therapeutics. Nature Rev. Drug Discov. 2011, 10, 698–712. [Google Scholar] [CrossRef] [PubMed]
- Selkoe, D.J. Alzheimer’s disease: Genes, proteins, and therapy. Physiol. Rev. 2001, 81, 741–766. [Google Scholar] [CrossRef] [PubMed]
- Haass, C. Take five–BACE and the gamma-secretase quartet conduct Alzheimer’s amyloid beta-peptide generation. EMBO J. 2004, 23, 483–488. [Google Scholar] [CrossRef] [PubMed]
- De Strooper, B.; Vassar, R.; Golde, T. The secretases: Enzymes with therapeutic potential in Alzheimer disease. Nat. Rev. Neurol. 2010, 6, 99–107. [Google Scholar] [CrossRef] [PubMed]
- Hsiao, K.; Chapman, P.; Nilsen, S.; Eckman, C.; Harigaya, Y.; Younkin, S.; Yang, F.; Cole, G. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 1996, 274, 99–102. [Google Scholar] [CrossRef] [PubMed]
- Lalonde, R.; Dumont, M.; Staufenbiel, M.; Sturchler-Pierrat, C.; Strazielle, C. Spatial learning, exploration, anxiety, and motor coordination in female APP23 transgenic mice with the Swedish mutation. Brain Res. 2002, 956, 36–44. [Google Scholar] [CrossRef]
- Jiang, Q.; Lee, C.Y.; Mandrekar, S.; Wilkinson, B.; Cramer, P.; Zelcer, N.; Mann, K.; Lamb, B.; Willson, T.M.; Collins, J.L.; et al. ApoE promotes the proteolytic degradation of Abeta. Neuron 2008, 58, 681–693. [Google Scholar] [CrossRef]
- Hauser, P.S.; Ryan, R.O. Impact of apolipoprotein E on Alzheimer’s disease. Curr. Alzheimer Res. 2013, 10, 809–817. [Google Scholar] [CrossRef]
- Liu, J.; Chang, L.; Roselli, F.; Almeida, O.F.; Gao, X.; Wang, X.; Yew, D.T.; Wu, Y. Amyloid-beta induces caspase-dependent loss of PSD-95 and synaptophysin through NMDA receptors. J. Alzheimer’s Dis. JAD 2010, 22, 541–556. [Google Scholar] [CrossRef]
- Roberson, E.D.; Scearce-Levie, K.; Palop, J.J.; Yan, F.; Cheng, I.H.; Wu, T.; Gerstein, H.; Yu, G.Q.; Mucke, L. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science 2007, 316, 750–754. [Google Scholar] [CrossRef] [PubMed]
- Nussbaum, J.M.; Seward, M.E.; Bloom, G.S. Alzheimer disease: A tale of two prions. Prion 2013, 7, 14–19. [Google Scholar] [CrossRef]
- Borchelt, D.R.; Thinakaran, G.; Eckman, C.B.; Lee, M.K.; Davenport, F.; Ratovitsky, T.; Prada, C.M.; Kim, G.; Seekins, S.; Yager, D.; et al. Familial Alzheimer’s disease-linked presenilin 1 variants elevate Abeta1-42/1-40 ratio in vitro and in vivo. Neuron 1996, 17, 1005–1013. [Google Scholar] [CrossRef]
- Cavallucci, V.; D’Amelio, M.; Cecconi, F. Abeta toxicity in Alzheimer’s disease. Mol. Neurobiol. 2012, 45, 366–378. [Google Scholar] [CrossRef] [PubMed]
- Lacor, P.N.; Buniel, M.C.; Furlow, P.W.; Clemente, A.S.; Velasco, P.T.; Wood, M.; Viola, K.L.; Klein, W.L. 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. [Google Scholar] [CrossRef]
- Nikolaev, A.; McLaughlin, T.; O’Leary, D.D.; Tessier-Lavigne, M. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature 2009, 457, 981–989. [Google Scholar] [CrossRef] [Green Version]
- Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef]
- Korczyn, A.D. The amyloid cascade hypothesis. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2008, 4, 176–178. [Google Scholar] [CrossRef]
- Ricciarelli, R.; Fedele, E. The amyloid cascade hypothesis in Alzheimer’s disease: It’s time to change our mind. Curr. Neuropharmacol. 2017, 15, 926–935. [Google Scholar] [CrossRef]
- Dong, S.; Duan, Y.; Hu, Y.; Zhao, Z. Advances in the pathogenesis of Alzheimer’s disease: A re-evaluation of amyloid cascade hypothesis. Transl. Neurodegener. 2012, 1, 18. [Google Scholar] [CrossRef]
- The amyloid cascade hypothesis has misled the pharmaceutical industry. Biochem. Soc. Trans. 2011, 39, 920–923. [CrossRef] [PubMed] [Green Version]
- Goedert, M.; Spillantini, M.G.; Crowther, R.A. Tau proteins and neurofibrillary degeneration. Brain Pathol. 1991, 1, 279–286. [Google Scholar] [CrossRef] [PubMed]
- Maccioni, R.B.; Munoz, J.P.; Barbeito, L. The molecular bases of Alzheimer’s disease and other neurodegenerative disorders. Arch. Med. Res. 2001, 32, 367–381. [Google Scholar] [CrossRef]
- Iqbal, K.; Alonso Adel, C.; Chen, S.; Chohan, M.O.; El-Akkad, E.; Gong, C.X.; Khatoon, S.; Li, B.; Liu, F.; Rahman, A.; et al. Tau pathology in Alzheimer disease and other tauopathies. Biochim. Biophys. Acta 2005, 1739, 198–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chun, W.; Johnson, G.V. The role of tau phosphorylation and cleavage in neuronal cell death. Front. Biosci. J. Virtual Libr. 2007, 12, 733–756. [Google Scholar] [CrossRef]
- Cardona, A.E.; Huang, D.; Sasse, M.E.; Ransohoff, R.M. Isolation of murine microglial cells for RNA analysis or flow cytometry. Nat. Protoc. 2006, 1, 1947–1951. [Google Scholar] [CrossRef] [PubMed]
- Filiano, A.J.; Gadani, S.P.; Kipnis, J. Interactions of innate and adaptive immunity in brain development and function. Brain Res. 2015, 1617, 18–27. [Google Scholar] [CrossRef] [Green Version]
- Wirenfeldt, M.; Babcock, A.A.; Vinters, H.V. Microglia—Insights into immune system structure, function, and reactivity in the central nervous system. Histol. Histopathol. 2011, 26, 519–530. [Google Scholar] [CrossRef]
- Aloisi, F. Immune function of microglia. Glia 2001, 36, 165–179. [Google Scholar] [CrossRef]
- Kettenmann, H.; Hanisch, U.K.; Noda, M.; Verkhratsky, A. Physiology of microglia. Physiol. Rev. 2011, 91, 461–553. [Google Scholar] [CrossRef]
- Ji, K.; Akgul, G.; Wollmuth, L.P.; Tsirka, S.E. Microglia actively regulate the number of functional synapses. PLoS ONE 2013, 8, e56293. [Google Scholar] [CrossRef] [PubMed]
- Parkhurst, C.N.; Yang, G.; Ninan, I.; Savas, J.N.; Yates, J.R., 3rd; Lafaille, J.J.; Hempstead, B.L.; Littman, D.R.; Gan, W.B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 2013, 155, 1596–1609. [Google Scholar] [CrossRef] [PubMed]
- Keren-Shaul, H.; Spinrad, A.; Weiner, A.; Matcovitch-Natan, O.; Dvir-Szternfeld, R.; Ulland, T.K.; David, E.; Baruch, K.; Lara-Astaiso, D.; Toth, B.; et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell 2017, 169, 1276–1290. [Google Scholar] [CrossRef] [PubMed]
- Sierra, A.; Gottfried-Blackmore, A.C.; McEwen, B.S.; Bulloch, K. Microglia derived from aging mice exhibit an altered inflammatory profile. Glia 2007, 55, 412–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wong, W.T. Microglial aging in the healthy CNS: Phenotypes, drivers, and rejuvenation. Front. Cell. Neurosci. 2013, 7, 22. [Google Scholar] [CrossRef] [PubMed]
- Lopes, K.O.; Sparks, D.L.; Streit, W.J. Microglial dystrophy in the aged and Alzheimer’s disease brain is associated with ferritin immunoreactivity. Glia 2008, 56, 1048–1060. [Google Scholar] [CrossRef] [PubMed]
- Youm, Y.H.; Grant, R.W.; McCabe, L.R.; Albarado, D.C.; Nguyen, K.Y.; Ravussin, A.; Pistell, P.; Newman, S.; Carter, R.; Laque, A.; et al. Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging. Cell Metabol. 2013, 18, 519–532. [Google Scholar] [CrossRef]
- Streit, W.J. Microglial senescence: Does the brain’s immune system have an expiration date? Trends Neurosci. 2006, 29, 506–510. [Google Scholar] [CrossRef]
- Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef]
- Patel, N.S.; Paris, D.; Mathura, V.; Quadros, A.N.; Crawford, F.C.; Mullan, M.J. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer’s disease. J. Neuroinflammation 2005, 2, 9. [Google Scholar] [CrossRef]
- Xia, M.Q.; Qin, S.X.; Wu, L.J.; Mackay, C.R.; Hyman, B.T. Immunohistochemical study of the beta-chemokine receptors CCR3 and CCR5 and their ligands in normal and Alzheimer’s disease brains. Am. J. Pathol. 1998, 153, 31–37. [Google Scholar] [CrossRef]
- Ishizuka, K.; Kimura, T.; Igata-yi, R.; Katsuragi, S.; Takamatsu, J.; Miyakawa, T. Identification of monocyte chemoattractant protein-1 in senile plaques and reactive microglia of Alzheimer’s disease. Psychiatry Clin. Neurosci. 1997, 51, 135–138. [Google Scholar] [CrossRef] [PubMed]
- Stewart, C.R.; Stuart, L.M.; Wilkinson, K.; van Gils, J.M.; Deng, J.; Halle, A.; Rayner, K.J.; Boyer, L.; Zhong, R.; Frazier, W.A.; et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nature Immunol. 2010, 11, 155–161. [Google Scholar] [CrossRef] [PubMed]
- Bamberger, M.E.; Harris, M.E.; McDonald, D.R.; Husemann, J.; Landreth, G.E. A cell surface receptor complex for fibrillar beta-amyloid mediates microglial activation. J. Neurosci. 2003, 23, 2665–2674. [Google Scholar] [CrossRef] [PubMed]
- Fassbender, K.; Walter, S.; Kuhl, S.; Landmann, R.; Ishii, K.; Bertsch, T.; Stalder, A.K.; Muehlhauser, F.; Liu, Y.; Ulmer, A.J.; et al. The LPS receptor (CD14) links innate immunity with Alzheimer’s disease. FASEB J. 2004, 18, 203–205. [Google Scholar] [CrossRef] [PubMed]
- El Khoury, J.B.; Moore, K.J.; Means, T.K.; Leung, J.; Terada, K.; Toft, M.; Freeman, M.W.; Luster, A.D. CD36 mediates the innate host response to beta-amyloid. J. Exp. Med. 2003, 197, 1657–1666. [Google Scholar] [CrossRef] [PubMed]
- Kagan, J.C.; Horng, T. NLRP3 inflammasome activation: CD36 serves double duty. Nat. Immunol. 2013, 14, 772–774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sheedy, F.J.; Grebe, A.; Rayner, K.J.; Kalantari, P.; Ramkhelawon, B.; Carpenter, S.B.; Becker, C.E.; Ediriweera, H.N.; Mullick, A.E.; Golenbock, D.T.; et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013, 14, 812–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saresella, M.; La Rosa, F.; Piancone, F.; Zoppis, M.; Marventano, I.; Calabrese, E.; Rainone, V.; Nemni, R.; Mancuso, R.; Clerici, M. The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer’s disease. Mol. Neurodegener. 2016, 11, 23. [Google Scholar] [CrossRef] [PubMed]
- Heneka, M.T.; Kummer, M.P.; Stutz, A.; Delekate, A.; Schwartz, S.; Vieira-Saecker, A.; Griep, A.; Axt, D.; Remus, A.; Tzeng, T.C.; et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 2013, 493, 674–678. [Google Scholar] [CrossRef] [PubMed]
- Bradshaw, E.M.; Chibnik, L.B.; Keenan, B.T.; Ottoboni, L.; Raj, T.; Tang, A.; Rosenkrantz, L.L.; Imboywa, S.; Lee, M.; Von Korff, A.; et al. CD33 Alzheimer’s disease locus: Altered monocyte function and amyloid biology. Nat. Neurosci. 2013, 16, 848–850. [Google Scholar] [CrossRef] [PubMed]
- Liu, G.; Jiang, Q. Alzheimer’s disease CD33 rs3865444 variant does not contribute to cognitive performance. Proc. Natl. Acad. Sci. USA 2016, 113, E1589–E1590. [Google Scholar] [CrossRef] [PubMed]
- Griciuc, A.; Serrano-Pozo, A.; Parrado, A.R.; Lesinski, A.N.; Asselin, C.N.; Mullin, K.; Hooli, B.; Choi, S.H.; Hyman, B.T.; Tanzi, R.E. Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron 2013, 78, 631–643. [Google Scholar] [CrossRef] [PubMed]
- Hickman, S.E.; Allison, E.K.; El Khoury, J. Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer’s disease mice. J. Neurosci. 2008, 28, 8354–8360. [Google Scholar] [CrossRef] [PubMed]
- Mawuenyega, K.G.; Sigurdson, W.; Ovod, V.; Munsell, L.; Kasten, T.; Morris, J.C.; Yarasheski, K.E.; Bateman, R.J. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science 2010, 330, 1774. [Google Scholar] [CrossRef]
- Xue, J.; Schmidt, S.V.; Sander, J.; Draffehn, A.; Krebs, W.; Quester, I.; De Nardo, D.; Gohel, T.D.; Emde, M.; Schmidleithner, L.; et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 2014, 40, 274–288. [Google Scholar] [CrossRef]
- Ransohoff, R.M. A polarizing question: Do M1 and M2 microglia exist? Nat. Neurosci. 2016, 19, 987–991. [Google Scholar] [CrossRef]
- Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 polarization and metabolic states. Br. J. Pharmacol. 2016, 173, 649–665. [Google Scholar] [CrossRef]
- Weldon, D.T.; Rogers, S.D.; Ghilardi, J.R.; Finke, M.P.; Cleary, J.P.; O’Hare, E.; Esler, W.P.; Maggio, J.E.; Mantyh, P.W. Fibrillar beta-amyloid induces microglial phagocytosis, expression of inducible nitric oxide synthase, and loss of a select population of neurons in the rat CNS in vivo. J. Neurosci. 1998, 18, 2161–2173. [Google Scholar] [CrossRef]
- Koenigsknecht-Talboo, J.; Landreth, G.E. Microglial phagocytosis induced by fibrillar beta-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J. Neurosci. 2005, 25, 8240–8249. [Google Scholar] [CrossRef]
- Yamamoto, M.; Kiyota, T.; Walsh, S.M.; Liu, J.; Kipnis, J.; Ikezu, T. Cytokine-mediated inhibition of fibrillar amyloid-beta peptide degradation by human mononuclear phagocytes. J. Immunol. 2008, 181, 3877–3886. [Google Scholar] [CrossRef] [PubMed]
- Michelucci, A.; Heurtaux, T.; Grandbarbe, L.; Morga, E.; Heuschling, P. Characterization of the microglial phenotype under specific pro-inflammatory and anti-inflammatory conditions: Effects of oligomeric and fibrillar amyloid-beta. J. Neuroimmunol. 2009, 210, 3–12. [Google Scholar] [CrossRef] [PubMed]
- Dempsey, C.; Rubio Araiz, A.; Bryson, K.J.; Finucane, O.; Larkin, C.; Mills, E.L.; Robertson, A.A.; Cooper, M.A.; O’Neill, L.A.; Lynch, M.A. Inhibiting the NLRP3 inflammasome with MCC950 promotes non-phlogistic clearance of amyloid-beta and cognitive function in APP/PS1 mice. Brain Behav. Immun. 2017, 61, 306–316. [Google Scholar] [CrossRef] [PubMed]
- Flores, J.; Noel, A.; Foveau, B.; Lynham, J.; Lecrux, C.; LeBlanc, A.C. Caspase-1 inhibition alleviates cognitive impairment and neuropathology in an Alzheimer’s disease mouse model. Nat. Commun. 2018, 9, 3916. [Google Scholar] [CrossRef] [PubMed]
- Tan, M.S.; Yu, J.T.; Jiang, T.; Zhu, X.C.; Wang, H.F.; Zhang, W.; Wang, Y.L.; Jiang, W.; Tan, L. NLRP3 polymorphisms are associated with late-onset Alzheimer’s disease in Han Chinese. J. Neuroimmunol. 2013, 265, 91–95. [Google Scholar] [CrossRef] [PubMed]
- Tan, M.S.; Yu, J.T.; Jiang, T.; Zhu, X.C.; Tan, L. The NLRP3 inflammasome in Alzheimer’s disease. Mol. Neurobiol. 2013, 48, 875–882. [Google Scholar] [CrossRef]
- Crismon, M.L. Tacrine: First drug approved for Alzheimer’s disease. Ann. Pharmacother. 1994, 28, 744–751. [Google Scholar] [CrossRef]
- Pan, S.Y.; Guo, B.F.; Zhang, Y.; Yu, Q.; Yu, Z.L.; Dong, H.; Ye, Y.; Han, Y.F.; Ko, K.M. Tacrine treatment at high dose suppresses the recognition memory in juvenile and adult mice with attention to hepatotoxicity. Basic Clin. Pharmacol. Toxicol. 2011, 108, 421–427. [Google Scholar] [CrossRef]
- Galisteo, M.; Rissel, M.; Sergent, O.; Chevanne, M.; Cillard, J.; Guillouzo, A.; Lagadic-Gossmann, D. Hepatotoxicity of tacrine: Occurrence of membrane fluidity alterations without involvement of lipid peroxidation. J. Pharmacol. Exp. Ther. 2000, 294, 160–167. [Google Scholar]
- Gutzmann, H.; Kuhl, K.P.; Hadler, D.; Rapp, M.A. Safety and efficacy of idebenone versus tacrine in patients with Alzheimer’s disease: Results of a randomized, double-blind, parallel-group multicenter study. Pharmacopsychiatry 2002, 35, 12–18. [Google Scholar] [CrossRef]
- Gracon, S.I.; Knapp, M.J.; Berghoff, W.G.; Pierce, M.; DeJong, R.; Lobbestael, S.J.; Symons, J.; Dombey, S.L.; Luscombe, F.A.; Kraemer, D. Safety of tacrine: Clinical trials, treatment IND, and postmarketing experience. Alzheimer Dis. Assoc. Disord. 1998, 12, 93–101. [Google Scholar] [CrossRef] [PubMed]
- Grossberg, G.T. Effect of rivastigmine in the treatment of behavioral disturbances associated with dementia: Review of neuropsychiatric impairment in Alzheimer’s disease. Curr. Med. Res. Opin. 2005, 21, 1631–1639. [Google Scholar] [CrossRef] [PubMed]
- Rosler, M.; Anand, R.; Cicin-Sain, A.; Gauthier, S.; Agid, Y.; Dal-Bianco, P.; Stahelin, H.B.; Hartman, R.; Gharabawi, M. Efficacy and safety of rivastigmine in patients with Alzheimer’s disease: International randomised controlled trial. BMJ 1999, 318, 633–638. [Google Scholar] [CrossRef] [PubMed]
- Olin, J.; Schneider, L. Galantamine for Alzheimer’s disease. Cochrane Database Syst. Rev. 2001, 3, CD001747. [Google Scholar] [CrossRef]
- Pirttila, T.; Wilcock, G.; Truyen, L.; Damaraju, C.V. Long-term efficacy and safety of galantamine in patients with mild-to-moderate Alzheimer’s disease: Multicenter trial. Eur. J. Neurol. 2004, 11, 734–741. [Google Scholar] [CrossRef] [PubMed]
- Benjamin, B.; Burns, A. Donepezil for Alzheimer’s disease. Expert Rev. Neurother. 2007, 7, 1243–1249. [Google Scholar] [CrossRef] [PubMed]
- Winblad, B.; Kilander, L.; Eriksson, S.; Minthon, L.; Batsman, S.; Wetterholm, A.L.; Jansson-Blixt, C.; Haglund, A.; Severe Alzheimer’s Disease Study Group. Donepezil in patients with severe Alzheimer’s disease: Double-blind, parallel-group, placebo-controlled study. Lancet 2006, 367, 1057–1065. [Google Scholar] [CrossRef]
- Birks, J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane Database Syst. Rev. 2006, 1, CD005593. [Google Scholar] [CrossRef]
- Ali, T.B.; Schleret, T.R.; Reilly, B.M.; Chen, W.Y.; Abagyan, R. Adverse Effects of Cholinesterase Inhibitors in Dementia, According to the Pharmacovigilance Databases of the United-States and Canada. PLoS ONE 2015, 10, e0144337. [Google Scholar] [CrossRef] [PubMed]
- Inglis, F. The tolerability and safety of cholinesterase inhibitors in the treatment of dementia. Int. J. Clin. Pract. Suppl. 2002, 45–63. [Google Scholar]
- Bleich, S.; Wiltfang, J.; Kornhuber, J. Memantine in moderate-to-severe Alzheimer’s disease. New Engl. J. Med. 2003, 349, 609–610. [Google Scholar] [CrossRef] [PubMed]
- Schneider, L.S.; Dagerman, K.S.; Higgins, J.P.; McShane, R. Lack of evidence for the efficacy of memantine in mild Alzheimer disease. Arch. Neurol. 2011, 68, 991–998. [Google Scholar] [CrossRef] [PubMed]
- Tan, M.G.; Shear, N.H.; Walsh, S.; Alhusayen, R. Acitretin. J. Cutan. Med. Surg. 2017, 21, 48–53. [Google Scholar] [CrossRef] [PubMed]
- Guenther, L.C.; Kunynetz, R.; Lynde, C.W.; Sibbald, R.G.; Toole, J.; Vender, R.; Zip, C. Acitretin Use in Dermatology. J. Cutan. Med. Surg. 2017, 21, 2S–S12. [Google Scholar] [CrossRef] [PubMed]
- Heath, M.S.; Sahni, D.R.; Curry, Z.A.; Feldman, S.R. Pharmacokinetics of tazarotene and acitretin in psoriasis. Expert Opin. Drug Metabol. Toxicol. 2018, 14, 919–927. [Google Scholar] [CrossRef] [PubMed]
- Endres, K.; Fahrenholz, F.; Lotz, J.; Hiemke, C.; Teipel, S.; Lieb, K.; Tuscher, O.; Fellgiebel, A. Increased CSF APPs-alpha levels in patients with Alzheimer disease treated with acitretin. Neurology 2014, 83, 1930–1935. [Google Scholar] [CrossRef] [PubMed]
- Xicota, L.; Rodriguez-Morato, J.; Dierssen, M.; de la Torre, R. Potential Role of (−)-Epigallocatechin-3-Gallate (EGCG) in the Secondary Prevention of Alzheimer Disease. Curr. Drug Targets 2017, 18, 174–195. [Google Scholar] [CrossRef] [PubMed]
- Singh, N.A.; Mandal, A.K.; Khan, Z.A. Potential neuroprotective properties of epigallocatechin-3-gallate (EGCG). Nutr. J. 2016, 15, 60. [Google Scholar] [CrossRef]
- Siopi, E.; Llufriu-Daben, G.; Cho, A.H.; Vidal-Lletjos, S.; Plotkine, M.; Marchand-Leroux, C.; Jafarian-Tehrani, M. Etazolate, an alpha-secretase activator, reduces neuroinflammation and offers persistent neuroprotection following traumatic brain injury in mice. Neuropharmacology 2013, 67, 183–192. [Google Scholar] [CrossRef] [PubMed]
- Marcade, M.; Bourdin, J.; Loiseau, N.; Peillon, H.; Rayer, A.; Drouin, D.; Schweighoffer, F.; Desire, L. Etazolate, a neuroprotective drug linking GABA(A) receptor pharmacology to amyloid precursor protein processing. J. Neurochem. 2008, 106, 392–404. [Google Scholar] [CrossRef] [PubMed]
- Cebers, G.; Alexander, R.C.; Haeberlein, S.B.; Han, D.; Goldwater, R.; Ereshefsky, L.; Olsson, T.; Ye, N.; Rosen, L.; Russell, M.; et al. AZD3293: Pharmacokinetic and Pharmacodynamic Effects in Healthy Subjects and Patients with Alzheimer’s Disease. J. Alzheimer’s Dis. JAD 2017, 55, 1039–1053. [Google Scholar] [CrossRef] [PubMed]
- Eketjall, S.; Janson, J.; Kaspersson, K.; Bogstedt, A.; Jeppsson, F.; Falting, J.; Haeberlein, S.B.; Kugler, A.R.; Alexander, R.C.; Cebers, G. AZD3293: A Novel, Orally Active BACE1 Inhibitor with High Potency and Permeability and Markedly Slow Off-Rate Kinetics. J. Alzheimer’s Dis. JAD 2016, 50, 1109–1123. [Google Scholar] [CrossRef] [PubMed]
- Burki, T. Alzheimer’s disease research: The future of BACE inhibitors. Lancet 2018, 391, 2486. [Google Scholar] [CrossRef]
- Blume, T.; Filser, S.; Jaworska, A.; Blain, J.F.; Koenig, G.; Moschke, K.; Lichtenthaler, S.F.; Herms, J. BACE1 Inhibitor MK-8931 Alters Formation but Not Stability of Dendritic Spines. Front. Aging Neurosci. 2018, 10, 229. [Google Scholar] [CrossRef] [PubMed]
- Thaisrivongs, D.A.; Morris, W.J.; Tan, L.; Song, Z.J.; Lyons, T.W.; Waldman, J.H.; Naber, J.R.; Chen, W.; Chen, L.; Zhang, B.; et al. A Next Generation Synthesis of BACE1 Inhibitor Verubecestat (MK-8931). Org. Lett. 2018, 20, 1568–1571. [Google Scholar] [CrossRef]
- Scott, J.D.; Li, S.W.; Brunskill, A.P.; Chen, X.; Cox, K.; Cumming, J.N.; Forman, M.; Gilbert, E.J.; Hodgson, R.A.; Hyde, L.A.; et al. Discovery of the 3-Imino-1,2,4-thiadiazinane 1,1-Dioxide Derivative Verubecestat (MK-8931)-A beta-Site Amyloid Precursor Protein Cleaving Enzyme 1 Inhibitor for the Treatment of Alzheimer’s Disease. J. Med. Chem. 2016, 59, 10435–10450. [Google Scholar] [CrossRef]
- Kennedy, M.E.; Stamford, A.W.; Chen, X.; Cox, K.; Cumming, J.N.; Dockendorf, M.F.; Egan, M.; Ereshefsky, L.; Hodgson, R.A.; Hyde, L.A.; et al. The BACE1 inhibitor verubecestat (MK-8931) reduces CNS beta-amyloid in animal models and in Alzheimer’s disease patients. Sci. Transl. Med. 2016, 8, 363ra150. [Google Scholar] [CrossRef]
- Timmers, M.; Streffer, J.R.; Russu, A.; Tominaga, Y.; Shimizu, H.; Shiraishi, A.; Tatikola, K.; Smekens, P.; Borjesson-Hanson, A.; Andreasen, N.; et al. Pharmacodynamics of atabecestat (JNJ-54861911), an oral BACE1 inhibitor in patients with early Alzheimer’s disease: Randomized, double-blind, placebo-controlled study. Alzheimer’s Res. Ther. 2018, 10, 85. [Google Scholar] [CrossRef]
- Timmers, M.; Sinha, V.; Darpo, B.; Smith, B.; Brown, R.; Xue, H.; Ferber, G.; Streffer, J.; Russu, A.; Tritsmans, L.; et al. Evaluating Potential QT Effects of JNJ-54861911, a BACE Inhibitor in Single- and Multiple-Ascending Dose Studies, and a Thorough QT Trial With Additional Retrospective Confirmation, Using Concentration-QTc Analysis. J. Clin. Pharmacol. 2018, 58, 952–964. [Google Scholar] [CrossRef]
- Timmers, M.; Van Broeck, B.; Ramael, S.; Slemmon, J.; De Waepenaert, K.; Russu, A.; Bogert, J.; Stieltjes, H.; Shaw, L.M.; Engelborghs, S.; et al. Profiling the dynamics of CSF and plasma Abeta reduction after treatment with JNJ-54861911, a potent oral BACE inhibitor. Alzheimers Dement. (N. Y.) 2016, 2, 202–212. [Google Scholar] [CrossRef]
- Neumann, U.; Ufer, M.; Jacobson, L.H.; Rouzade-Dominguez, M.L.; Huledal, G.; Kolly, C.; Luond, R.M.; Machauer, R.; Veenstra, S.J.; Hurth, K.; et al. The BACE-1 inhibitor CNP520 for prevention trials in Alzheimer’s disease. EMBO Mol. Med. 2018, 10, e9316. [Google Scholar] [CrossRef] [PubMed]
- Lopez Lopez, C.; Caputo, A.; Liu, F.; Riviere, M.E.; Rouzade-Dominguez, M.L.; Thomas, R.G.; Langbaum, J.B.; Lenz, R.; Reiman, E.M.; Graf, A.; et al. The Alzheimer’s Prevention Initiative Generation Program: Evaluating CNP520 Efficacy in the Prevention of Alzheimer’s Disease. J. Prev. Alzheimer’s Dis. 2017, 4, 242–246. [Google Scholar] [CrossRef]
- Folch, J.; Petrov, D.; Ettcheto, M.; Abad, S.; Sanchez-Lopez, E.; Garcia, M.L.; Olloquequi, J.; Beas-Zarate, C.; Auladell, C.; Camins, A. Current Research Therapeutic Strategies for Alzheimer’s Disease Treatment. Neural Plast. 2016, 2016, 8501693. [Google Scholar] [CrossRef] [PubMed]
- Dockens, R.; Wang, J.S.; Castaneda, L.; Sverdlov, O.; Huang, S.P.; Slemmon, R.; Gu, H.; Wong, O.; Li, H.; Berman, R.M.; et al. A placebo-controlled, multiple ascending dose study to evaluate the safety, pharmacokinetics and pharmacodynamics of avagacestat (BMS-708163) in healthy young and elderly subjects. Clin. Pharmacokinet. 2012, 51, 681–693. [Google Scholar] [CrossRef] [PubMed]
- Tong, G.; Castaneda, L.; Wang, J.S.; Sverdlov, O.; Huang, S.P.; Slemmon, R.; Gu, H.; Wong, O.; Li, H.; Berman, R.M.; et al. Effects of single doses of avagacestat (BMS-708163) on cerebrospinal fluid Abeta levels in healthy young men. Clin. Drug Investig. 2012, 32, 761–769. [Google Scholar] [CrossRef] [PubMed]
- Coric, V.; van Dyck, C.H.; Salloway, S.; Andreasen, N.; Brody, M.; Richter, R.W.; Soininen, H.; Thein, S.; Shiovitz, T.; Pilcher, G.; et al. Safety and tolerability of the gamma-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Arch. Neurol. 2012, 69, 1430–1440. [Google Scholar] [CrossRef]
- Matlack, K.E.; Tardiff, D.F.; Narayan, P.; Hamamichi, S.; Caldwell, K.A.; Caldwell, G.A.; Lindquist, S. Clioquinol promotes the degradation of metal-dependent amyloid-beta (Abeta) oligomers to restore endocytosis and ameliorate Abeta toxicity. Proc. Natl. Acad. Sci. USA 2014, 111, 4013–4018. [Google Scholar] [CrossRef] [PubMed]
- Liang, E.; Garzone, P.; Cedarbaum, J.M.; Koller, M.; Tran, T.; Xu, V.; Ross, B.; Jhee, S.S.; Ereshefsky, L.; Pastrak, A.; et al. Pharmacokinetic Profile of Orally Administered Scyllo-Inositol (Elnd005) in Plasma, Cerebrospinal Fluid and Brain, and Corresponding Effect on Amyloid-Beta in Healthy Subjects. Clin. Pharmacol. Drug Dev. 2013, 2, 186–194. [Google Scholar] [CrossRef] [PubMed]
- Salloway, S.; Sperling, R.; Keren, R.; Porsteinsson, A.P.; van Dyck, C.H.; Tariot, P.N.; Gilman, S.; Arnold, D.; Abushakra, S.; Hernandez, C.; et al. A phase 2 randomized trial of ELND005, scyllo-inositol, in mild to moderate Alzheimer disease. Neurology 2011, 77, 1253–1262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caltagirone, C.; Ferrannini, L.; Marchionni, N.; Nappi, G.; Scapagnini, G.; Trabucchi, M. The potential protective effect of tramiprosate (homotaurine) against Alzheimer’s disease: A review. Aging Clin. Exp. Res. 2012, 24, 580–587. [Google Scholar] [CrossRef] [PubMed]
- Aisen, P.S.; Gauthier, S.; Ferris, S.H.; Saumier, D.; Haine, D.; Garceau, D.; Duong, A.; Suhy, J.; Oh, J.; Lau, W.C.; et al. Tramiprosate in mild-to-moderate Alzheimer’s disease—A randomized, double-blind, placebo-controlled, multi-centre study (the Alphase Study). Arch. Med. Sci. AMS 2011, 7, 102–111. [Google Scholar] [CrossRef] [PubMed]
- Gervais, F.; Paquette, J.; Morissette, C.; Krzywkowski, P.; Yu, M.; Azzi, M.; Lacombe, D.; Kong, X.; Aman, A.; Laurin, J.; et al. Targeting soluble Abeta peptide with Tramiprosate for the treatment of brain amyloidosis. Neurobiol. Aging 2007, 28, 537–547. [Google Scholar] [CrossRef] [PubMed]
- Gauthier, S.; Aisen, P.S.; Ferris, S.H.; Saumier, D.; Duong, A.; Haine, D.; Garceau, D.; Suhy, J.; Oh, J.; Lau, W.; 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. [Google Scholar] [CrossRef]
- Sabbagh, M.N. Clinical Effects of Oral Tramiprosate in APOE4/4 Homozygous Patients with Mild Alzheimer’s Disease Suggest Disease Modification. J. Prev. Alzheimer’s Dis. 2017, 4, 136–137. [Google Scholar] [CrossRef]
- Kocis, P.; Tolar, M.; Yu, J.; Sinko, W.; Ray, S.; Blennow, K.; Fillit, H.; Hey, J.A. Elucidating the Abeta42 Anti-Aggregation Mechanism of Action of Tramiprosate in Alzheimer’s Disease: Integrating Molecular Analytical Methods, Pharmacokinetic and Clinical Data. CNS Drugs 2017, 31, 495–509. [Google Scholar] [CrossRef] [PubMed]
- Abushakra, S.; Porsteinsson, A.; Scheltens, P.; Sadowsky, C.; Vellas, B.; Cummings, J.; Gauthier, S.; Hey, J.A.; Power, A.; Wang, P.; et al. Clinical Effects of Tramiprosate in APOE4/4 Homozygous Patients with Mild Alzheimer’s Disease Suggest Disease Modification Potential. J. Prev. Alzheimer’s Dis. 2017, 4, 149–156. [Google Scholar] [CrossRef]
- Abushakra, S.; Porsteinsson, A.; Vellas, B.; Cummings, J.; Gauthier, S.; Hey, J.A.; Power, A.; Hendrix, S.; Wang, P.; Shen, L.; et al. Clinical Benefits of Tramiprosate in Alzheimer’s Disease Are Associated with Higher Number of APOE4 Alleles: The “APOE4 Gene-Dose Effect”. J. Prev. Alzheimer’s Dis. 2016, 3, 219–228. [Google Scholar] [CrossRef]
- Liu, Y.; Xu, L.P.; Wang, Q.; Yang, B.; Zhang, X. Synergistic Inhibitory Effect of GQDs-Tramiprosate Covalent Binding on Amyloid Aggregation. ACS Chem. Neurosci. 2018, 9, 817–823. [Google Scholar] [CrossRef]
- Hey, J.A.; Yu, J.Y.; Versavel, M.; Abushakra, S.; Kocis, P.; Power, A.; Kaplan, P.L.; Amedio, J.; Tolar, M. Clinical Pharmacokinetics and Safety of ALZ-801, a Novel Prodrug of Tramiprosate in Development for the Treatment of Alzheimer’s Disease. Clin. Pharmacokinet. 2018, 57, 315–333. [Google Scholar] [CrossRef]
- Hey, J.A.; Kocis, P.; Hort, J.; Abushakra, S.; Power, A.; Vyhnalek, M.; Yu, J.Y.; Tolar, M. Discovery and Identification of an Endogenous Metabolite of Tramiprosate and Its Prodrug ALZ-801 that Inhibits Beta Amyloid Oligomer Formation in the Human Brain. CNS Drugs 2018, 32, 849–861. [Google Scholar] [CrossRef]
- Cummings, J.; Lee, G.; Ritter, A.; Zhong, K. Alzheimer’s disease drug development pipeline: 2018. Alzheimers Dement. (N. Y.) 2018, 4, 195–214. [Google Scholar] [CrossRef] [PubMed]
- Gilman, S.; Koller, M.; Black, R.S.; Jenkins, L.; Griffith, S.G.; Fox, N.C.; Eisner, L.; Kirby, L.; Rovira, M.B.; Forette, F.; et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology 2005, 64, 1553–1562. [Google Scholar] [CrossRef] [PubMed]
- Holmes, C.; Boche, D.; Wilkinson, D.; Yadegarfar, G.; Hopkins, V.; Bayer, A.; Jones, R.W.; Bullock, R.; Love, S.; Neal, J.W.; et al. Long-term effects of Abeta42 immunisation in Alzheimer’s disease: Follow-up of a randomised, placebo-controlled phase I trial. Lancet 2008, 372, 216–223. [Google Scholar] [CrossRef]
- Hull, M.; Sadowsky, C.; Arai, H.; Le Prince Leterme, G.; Holstein, A.; Booth, K.; Peng, Y.; Yoshiyama, T.; Suzuki, H.; Ketter, N.; et al. Long-Term Extensions of Randomized Vaccination Trials of ACC-001 and QS-21 in Mild to Moderate Alzheimer’s Disease. Curr. Alzheimer Res. 2017, 14, 696–708. [Google Scholar] [CrossRef] [PubMed]
- Pasquier, F.; Sadowsky, C.; Holstein, A.; Leterme Gle, P.; Peng, Y.; Jackson, N.; Fox, N.C.; Ketter, N.; Liu, E.; Ryan, J.M.; et al. Two Phase 2 Multiple Ascending-Dose Studies of Vanutide Cridificar (ACC-001) and QS-21 Adjuvant in Mild-to-Moderate Alzheimer’s Disease. J. Alzheimer’s Dis. JAD 2016, 51, 1131–1143. [Google Scholar] [CrossRef]
- van Dyck, C.H.; Sadowsky, C.; Le Prince Leterme, G.; Booth, K.; Peng, Y.; Marek, K.; Ketter, N.; Liu, E.; Wyman, B.T.; Jackson, N.; et al. Vanutide Cridificar (ACC-001) and QS-21 Adjuvant in Individuals with Early Alzheimer’s Disease: Amyloid Imaging Positron Emission Tomography and Safety Results from a Phase 2 Study. J. Prev. Alzheimer’s Dis. 2016, 3, 75–84. [Google Scholar] [CrossRef]
- Tayeb, H.O.; Murray, E.D.; Price, B.H.; Tarazi, F.I. Bapineuzumab and solanezumab for Alzheimer’s disease: Is the ‘amyloid cascade hypothesis’ still alive? Expert Opin. Biol. Ther. 2013, 13, 1075–1084. [Google Scholar] [CrossRef]
- Doody, R.S.; Farlow, M.; Aisen, P.S.; Alzheimer’s Disease Cooperative Study Data, A.; Publication, C. Phase 3 trials of solanezumab and bapineuzumab for Alzheimer’s disease. N. Engl. J. Med. 2014, 370, 1460. [Google Scholar] [CrossRef]
- La Porte, S.L.; Bollini, S.S.; Lanz, T.A.; Abdiche, Y.N.; Rusnak, A.S.; Ho, W.H.; Kobayashi, D.; Harrabi, O.; Pappas, D.; Mina, E.W.; et al. Structural basis of C-terminal beta-amyloid peptide binding by the antibody ponezumab for the treatment of Alzheimer’s disease. J. Mol. Biol. 2012, 421, 525–536. [Google Scholar] [CrossRef]
- Landen, J.W.; Zhao, Q.; Cohen, S.; Borrie, M.; Woodward, M.; Billing, C.B., Jr.; Bales, K.; Alvey, C.; McCush, F.; Yang, J.; et al. Safety and pharmacology of a single intravenous dose of ponezumab in subjects with mild-to-moderate Alzheimer disease: A phase I, randomized, placebo-controlled, double-blind, dose-escalation study. Clin. Neuropharmacol. 2013, 36, 14–23. [Google Scholar] [CrossRef]
- Landen, J.W.; Cohen, S.; Billing, C.B., Jr.; Cronenberger, C.; Styren, S.; Burstein, A.H.; Sattler, C.; Lee, J.H.; Jack, C.R., Jr.; Kantarci, K.; et al. Multiple-dose ponezumab for mild-to-moderate Alzheimer’s disease: Safety and efficacy. Alzheimers Dement. (N. Y.) 2017, 3, 339–347. [Google Scholar] [CrossRef] [PubMed]
- Landen, J.W.; Andreasen, N.; Cronenberger, C.L.; Schwartz, P.F.; Borjesson-Hanson, A.; Ostlund, H.; Sattler, C.A.; Binneman, B.; Bednar, M.M. Ponezumab in mild-to-moderate Alzheimer’s disease: Randomized phase II PET-PIB study. Alzheimers Dement. (N. Y.) 2017, 3, 393–401. [Google Scholar] [CrossRef] [PubMed]
- Andreasen, N.; Simeoni, M.; Ostlund, H.; Lisjo, P.I.; Fladby, T.; Loercher, A.E.; Byrne, G.J.; Murray, F.; Scott-Stevens, P.T.; Wallin, A.; et al. First administration of the Fc-attenuated anti-beta amyloid antibody GSK933776 to patients with mild Alzheimer’s disease: A randomized, placebo-controlled study. PLoS ONE 2015, 10, e0098153. [Google Scholar] [CrossRef] [PubMed]
- Leyhe, T.; Andreasen, N.; Simeoni, M.; Reich, A.; von Arnim, C.A.; Tong, X.; Yeo, A.; Khan, S.; Loercher, A.; Chalker, M.; et al. Modulation of beta-amyloid by a single dose of GSK933776 in patients with mild Alzheimer’s disease: A phase I study. Alzheimer’s Res. Ther. 2014, 6, 19. [Google Scholar] [CrossRef] [PubMed]
- Ferrero, J.; Williams, L.; Stella, H.; Leitermann, K.; Mikulskis, A.; O’Gorman, J.; Sevigny, J. First-in-human, double-blind, placebo-controlled, single-dose escalation study of aducanumab (BIIB037) in mild-to-moderate Alzheimer’s disease. Alzheimers Dement. (N. Y.) 2016, 2, 169–176. [Google Scholar] [CrossRef] [PubMed]
- Arndt, J.W.; Qian, F.; Smith, B.A.; Quan, C.; Kilambi, K.P.; Bush, M.W.; Walz, T.; Pepinsky, R.B.; Bussiere, T.; Hamann, S.; et al. Structural and kinetic basis for the selectivity of aducanumab for aggregated forms of amyloid-beta. Sci. Rep. 2018, 8, 6412. [Google Scholar] [CrossRef]
- Gamage, K.K.; Kumar, S. Aducanumab Therapy Ameliorates Calcium Overload in a Mouse Model of Alzheimer’s Disease. J. Neurosci. 2017, 37, 4430–4432. [Google Scholar] [CrossRef]
- Kastanenka, K.V.; Bussiere, T.; Shakerdge, N.; Qian, F.; Weinreb, P.H.; Rhodes, K.; Bacskai, B.J. Immunotherapy with Aducanumab Restores Calcium Homeostasis in Tg2576 Mice. J. Neurosci. 2016, 36, 12549–12558. [Google Scholar] [CrossRef]
- Budd Haeberlein, S.; O’Gorman, J.; Chiao, P.; Bussiere, T.; von Rosenstiel, P.; Tian, Y.; Zhu, Y.; von Hehn, C.; Gheuens, S.; Skordos, L.; et al. Clinical Development of Aducanumab, an Anti-Abeta Human Monoclonal Antibody Being Investigated for the Treatment of Early Alzheimer’s Disease. J. Prev. Alzheimer’s Dis. 2017, 4, 255–263. [Google Scholar] [CrossRef]
- Sevigny, J.; Chiao, P.; Bussiere, T.; Weinreb, P.H.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y.; et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature 2016, 537, 50–56. [Google Scholar] [CrossRef]
- Chiao, P.; Bedell, B.J.; Avants, B.; Zijdenbos, A.P.; Grand’Maison, M.; O’Neill, P.; O’Gorman, J.; Chen, T.; Koeppe, R. Impact of Reference/Target Region Selection on Amyloid PET Standard Uptake Value Ratios in the Phase 1b PRIME Study of Aducanumab. J. Nucl. Med. 2018. [Google Scholar] [CrossRef]
- Ultsch, M.; Li, B.; Maurer, T.; Mathieu, M.; Adolfsson, O.; Muhs, A.; Pfeifer, A.; Pihlgren, M.; Bainbridge, T.W.; Reichelt, M.; et al. Structure of Crenezumab Complex with Abeta Shows Loss of beta-Hairpin. Sci. Rep. 2016, 6, 39374. [Google Scholar] [CrossRef]
- Salloway, S.; Honigberg, L.A.; Cho, W.; Ward, M.; Friesenhahn, M.; Brunstein, F.; Quartino, A.; Clayton, D.; Mortensen, D.; Bittner, T.; et al. Amyloid positron emission tomography and cerebrospinal fluid results from a crenezumab anti-amyloid-beta antibody double-blind, placebo-controlled, randomized phase II study in mild-to-moderate Alzheimer’s disease (BLAZE). Alzheimer’s Res. Ther. 2018, 10, 96. [Google Scholar] [CrossRef]
- Cummings, J.L.; Cohen, S.; van Dyck, C.H.; Brody, M.; Curtis, C.; Cho, W.; Ward, M.; Friesenhahn, M.; Rabe, C.; Brunstein, F.; et al. ABBY: A phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology 2018, 90, e1889–e1897. [Google Scholar] [CrossRef] [PubMed]
- Bohrmann, B.; Baumann, K.; Benz, J.; Gerber, F.; Huber, W.; Knoflach, F.; Messer, J.; Oroszlan, K.; Rauchenberger, R.; Richter, W.F.; et al. Gantenerumab: A novel human anti-Abeta antibody demonstrates sustained cerebral amyloid-beta binding and elicits cell-mediated removal of human amyloid-beta. J. Alzheimer’s Dis. JAD 2012, 28, 49–69. [Google Scholar] [CrossRef]
- Ostrowitzki, S.; Deptula, D.; Thurfjell, L.; Barkhof, F.; Bohrmann, B.; Brooks, D.J.; Klunk, W.E.; Ashford, E.; Yoo, K.; Xu, Z.X.; et al. Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Arch. Neurol. 2012, 69, 198–207. [Google Scholar] [CrossRef]
- Delrieu, J.; Ousset, P.J.; Vellas, B. Gantenerumab for the treatment of Alzheimer’s disease. Expert Opin. Biol. Ther. 2012, 12, 1077–1086. [Google Scholar] [CrossRef]
- Novakovic, D.; Feligioni, M.; Scaccianoce, S.; Caruso, A.; Piccinin, S.; Schepisi, C.; Errico, F.; Mercuri, N.B.; Nicoletti, F.; Nistico, R. Profile of gantenerumab and its potential in the treatment of Alzheimer’s disease. Drug Des. Dev. Ther. 2013, 7, 1359–1364. [Google Scholar] [CrossRef]
- Ostrowitzki, S.; Lasser, R.A.; Dorflinger, E.; Scheltens, P.; Barkhof, F.; Nikolcheva, T.; Ashford, E.; Retout, S.; Hofmann, C.; Delmar, P.; et al. A phase III randomized trial of gantenerumab in prodromal Alzheimer’s disease. Alzheimer’s Res. Ther. 2017, 9, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baddeley, T.C.; McCaffrey, J.; Storey, J.M.; Cheung, J.K.; Melis, V.; Horsley, D.; Harrington, C.R.; Wischik, C.M. 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. [Google Scholar] [CrossRef]
- Gauthier, S.; Feldman, H.H.; Schneider, L.S.; Wilcock, G.K.; Frisoni, G.B.; Hardlund, J.H.; Moebius, H.J.; Bentham, P.; Kook, K.A.; Wischik, D.J.; 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. [Google Scholar] [CrossRef]
- Wilcock, G.K.; Gauthier, S.; Frisoni, G.B.; Jia, J.; Hardlund, J.H.; Moebius, H.J.; Bentham, P.; Kook, K.A.; Schelter, B.O.; Wischik, D.J.; et al. Potential of Low Dose Leuco-Methylthioninium Bis(Hydromethanesulphonate) (LMTM) Monotherapy for Treatment of Mild Alzheimer’s Disease: Cohort Analysis as Modified Primary Outcome in a Phase III Clinical Trial. J. Alzheimer’s Dis. JAD 2018, 61, 435–457. [Google Scholar] [CrossRef] [PubMed]
- Lovestone, S.; Boada, M.; Dubois, B.; Hull, M.; Rinne, J.O.; Huppertz, H.J.; Calero, M.; Andres, M.V.; Gomez-Carrillo, B.; Leon, T.; et al. A phase II trial of tideglusib in Alzheimer’s disease. J. Alzheimer’s Dis. JAD 2015, 45, 75–88. [Google Scholar] [CrossRef]
- West, T.; Hu, Y.; Verghese, P.B.; Bateman, R.J.; Braunstein, J.B.; Fogelman, I.; Budur, K.; Florian, H.; Mendonca, N.; Holtzman, D.M. Preclinical and Clinical Development of ABBV-8E12, a Humanized Anti-Tau Antibody, for Treatment of Alzheimer’s Disease and Other Tauopathies. J. Prev. Alzheimer’s Dis. 2017, 4, 236–241. [Google Scholar] [CrossRef]
- Lee, S.H.; Le Pichon, C.E.; Adolfsson, O.; Gafner, V.; Pihlgren, M.; Lin, H.; Solanoy, H.; Brendza, R.; Ngu, H.; Foreman, O.; et al. Antibody-Mediated Targeting of Tau In Vivo Does Not Require Effector Function and Microglial Engagement. Cell Rep. 2016, 16, 1690–1700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kontsekova, E.; Zilka, N.; Kovacech, B.; Novak, P.; Novak, M. First-in-man tau vaccine targeting structural determinants essential for pathological tau-tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimer’s disease model. Alzheimer’s Res. Ther. 2014, 6, 44. [Google Scholar] [CrossRef]
- Novak, P.; Schmidt, R.; Kontsekova, E.; Kovacech, B.; Smolek, T.; Katina, S.; Fialova, L.; Prcina, M.; Parrak, V.; Dal-Bianco, P.; et al. FUNDAMANT: An interventional 72-week phase 1 follow-up study of AADvac1, an active immunotherapy against tau protein pathology in Alzheimer’s disease. Alzheimer’s Res. Ther. 2018, 10, 108. [Google Scholar] [CrossRef]
- Novak, P.; Schmidt, R.; Kontsekova, E.; Zilka, N.; Kovacech, B.; Skrabana, R.; Vince-Kazmerova, Z.; Katina, S.; Fialova, L.; Prcina, M.; et al. Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer’s disease: A randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol. 2017, 16, 123–134. [Google Scholar] [CrossRef]
- Fitzgerald, D.P.; Emerson, D.L.; Qian, Y.; Anwar, T.; Liewehr, D.J.; Steinberg, S.M.; Silberman, S.; Palmieri, D.; Steeg, P.S. TPI-287, a new taxane family member, reduces the brain metastatic colonization of breast cancer cells. Mol. Cancer Ther. 2012, 11, 1959–1967. [Google Scholar] [CrossRef]
- Etminan, M.; Gill, S.; Samii, A. Effect of non-steroidal anti-inflammatory drugs on risk of Alzheimer’s disease: Systematic review and meta-analysis of observational studies. BMJ 2003, 327, 128. [Google Scholar] [CrossRef]
- Breitner, J.C.; Haneuse, S.J.; Walker, R.; Dublin, S.; Crane, P.K.; Gray, S.L.; Larson, E.B. Risk of dementia and AD with prior exposure to NSAIDs in an elderly community-based cohort. Neurology 2009, 72, 1899–1905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vlad, S.C.; Miller, D.R.; Kowall, N.W.; Felson, D.T. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology 2008, 70, 1672–1677. [Google Scholar] [CrossRef] [Green Version]
- in t’ Veld, B.A.; Ruitenberg, A.; Hofman, A.; Launer, L.J.; van Duijn, C.M.; Stijnen, T.; Breteler, M.M.; Stricker, B.H. Nonsteroidal antiinflammatory drugs and the risk of Alzheimer’s disease. N. Engl. J. Med. 2001, 345, 1515–1521. [Google Scholar] [CrossRef] [PubMed]
- Daniels, M.J.; Rivers-Auty, J.; Schilling, T.; Spencer, N.G.; Watremez, W.; Fasolino, V.; Booth, S.J.; White, C.S.; Baldwin, A.G.; Freeman, S.; et al. Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer’s disease in rodent models. Nat. Commun. 2016, 7, 12504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; Tan, L.; Wang, H.F.; Tan, C.C.; Meng, X.F.; Wang, C.; Tang, S.W.; Yu, J.T. Anti-inflammatory drugs and risk of Alzheimer’s disease: An updated systematic review and meta-analysis. J. Alzheimer’s Dis. JAD 2015, 44, 385–396. [Google Scholar] [CrossRef] [PubMed]
- Miguel-Alvarez, M.; Santos-Lozano, A.; Sanchis-Gomar, F.; Fiuza-Luces, C.; Pareja-Galeano, H.; Garatachea, N.; Lucia, A. Non-steroidal anti-inflammatory drugs as a treatment for Alzheimer’s disease: A systematic review and meta-analysis of treatment effect. Drugs Aging 2015, 32, 139–147. [Google Scholar] [CrossRef] [PubMed]
- Jaturapatporn, D.; Isaac, M.G.; McCleery, J.; Tabet, N. Aspirin, steroidal and non-steroidal anti-inflammatory drugs for the treatment of Alzheimer’s disease. Cochrane Database Syst. Rev. 2012, 2, CD006378. [Google Scholar] [CrossRef]
- Pasqualetti, P.; Bonomini, C.; Dal Forno, G.; Paulon, L.; Sinforiani, E.; Marra, C.; Zanetti, O.; Rossini, P.M. A randomized controlled study on effects of ibuprofen on cognitive progression of Alzheimer’s disease. Aging Clin. Exp. Res. 2009, 21, 102–110. [Google Scholar] [CrossRef]
- Muntimadugu, E.; Dhommati, R.; Jain, A.; Challa, V.G.; Shaheen, M.; Khan, W. Intranasal delivery of nanoparticle encapsulated tarenflurbil: A potential brain targeting strategy for Alzheimer’s disease. Eur. J. Pharm. Sci. 2016, 92, 224–234. [Google Scholar] [CrossRef]
- Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S.; et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 2015, 85, 1383–1391. [Google Scholar] [CrossRef] [Green Version]
- Butchart, J.; Brook, L.; Hopkins, V.; Teeling, J.; Puntener, U.; Culliford, D.; Sharples, R.; Sharif, S.; McFarlane, B.; Raybould, R.; et al. Etanercept in Alzheimer disease: A randomized, placebo-controlled, double-blind, phase 2 trial. Neurology 2015, 84, 2161–2168. [Google Scholar] [CrossRef] [Green Version]
- Sano, M.; Bell, K.L.; Galasko, D.; Galvin, J.E.; Thomas, R.G.; van Dyck, C.H.; Aisen, P.S. A randomized, double-blind, placebo-controlled trial of simvastatin to treat Alzheimer disease. Neurology 2011, 77, 556–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duffy, J.P.; Harrington, E.M.; Salituro, F.G.; Cochran, J.E.; Green, J.; Gao, H.; Bemis, G.W.; Evindar, G.; Galullo, V.P.; Ford, P.J.; et al. The Discovery of VX-745: A Novel and Selective p38alpha Kinase Inhibitor. ACS Med. Chem. Lett. 2011, 2, 758–763. [Google Scholar] [CrossRef] [PubMed]
- Alam, J.; Blackburn, K.; Patrick, D. Neflamapimod: Clinical Phase 2b-Ready Oral Small Molecule Inhibitor of p38alpha to Reverse Synaptic Dysfunction in Early Alzheimer’s Disease. J. Prev. Alzheimer’s Dis. 2017, 4, 273–278. [Google Scholar] [CrossRef]
- Burstein, A.H.; Sabbagh, M.; Andrews, R.; Valcarce, C.; Dunn, I.; Altstiel, L. Development of Azeliragon, an Oral Small Molecule Antagonist of the Receptor for Advanced Glycation Endproducts, for the Potential Slowing of Loss of Cognition in Mild Alzheimer’s Disease. J. Prev. Alzheimer’s Dis. 2018, 5, 149–154. [Google Scholar] [CrossRef]
- Burstein, A.H.; Grimes, I.; Galasko, D.R.; Aisen, P.S.; Sabbagh, M.; Mjalli, A.M. Effect of TTP488 in patients with mild to moderate Alzheimer’s disease. BMC Neurol. 2014, 14, 12. [Google Scholar] [CrossRef] [PubMed]
- Galimberti, D.; Scarpini, E. Pioglitazone for the treatment of Alzheimer’s disease. Expert Opin. Investig. Drugs 2017, 26, 97–101. [Google Scholar] [CrossRef] [PubMed]
- Geldmacher, D.S.; Fritsch, T.; McClendon, M.J.; Landreth, G. A randomized pilot clinical trial of the safety of pioglitazone in treatment of patients with Alzheimer disease. Arch. Neurol. 2011, 68, 45–50. [Google Scholar] [CrossRef] [PubMed]
- Sato, T.; Hanyu, H.; Hirao, K.; Kanetaka, H.; Sakurai, H.; Iwamoto, T. Efficacy of PPAR-gamma agonist pioglitazone in mild Alzheimer disease. Neurobiol. Aging 2011, 32, 1626–1633. [Google Scholar] [CrossRef] [PubMed]
Drug | Description | Phase | CT Identifier | Status |
---|---|---|---|---|
Drugs Target Amyloid β Production | ||||
Acitretin | α-secretase enhancer | II | NCT01078168 | Completed |
Epigallocatechin-Gallate (EGCG) | α-secretase enhancer, prevent amyloid-β (Aβ) aggregation | II/III | NCT00951834 | Completed |
Etazolate (EHT-0202) | γ-aminobutyric acid GABA)A receptor modulator, α-secretase enhancer | II | NCT00880412 | Completed |
Lanabecestat (AZD3293, LY3314814) | β-secretase inhibitor | II/III | NCT02245737 NCT02972658 NCT02783573 | Terminated |
LY3202626 | β-secretase inhibitor | II | NCT02791191 | Terminated |
LY2286721 | β-secretase inhibitor | I/II | NCT01561430 | Terminated |
Verubecestat (MK-8931) | β-secretase inhibitor | II/III | NCT01739348 NCT01953601 | Terminated |
Atabecestat (JNJ-54861911) | β-secretase inhibitor | II/III | NCT02569398 NCT01760005 | Active, not recruiting |
II/III | NCT02406027 | Terminated | ||
Elenbecestat (E2609) | β-secretase inhibitor | III | NCT03036280 NCT02956486 | Recruiting |
II | NCT02322021 | Active, not recruiting | ||
CNP520 | β-secretase inhibitor | II | NCT02565511 NCT03131453 | Recruiting |
Semagacestat | γ-secretase inhibitor | III | NCT01035138 NCT00762411 NCT00594568 | Completed |
Avagacestat (BMS-708163) | γ-secretase inhibitor | II | NCT00890890 etc. | Terminated |
Drugs prevent Amyloid β Aggregation | ||||
PBT2 | metal protein-attenuating compound (MPAC), Aβ aggregation inhibitor | II/III | NCT00471211 | Terminated |
Scyllo-inositol (ELND005, AZD-103) | inositol stereoisomer, Aβ aggregation inhibitor | II | NCT00934050, NCT00568776, NCT01735630 | Completed |
Tramiprosate (3APS) | Prevent β-sheet formation, Aβ aggregation inhibitor | III | NCT00314912, NCT00088673, NCT00217763 | Unknown |
GV-971 | Aβ aggregation inhibitor | III | NCT02293915 | Completed |
Immunotherapy | ||||
AN-1792 (AIP-001) | Anti-Aβ vaccine | II | NCT00021723 | Terminated |
CAD106 | Anti-Aβ vaccine, induce Anti-Aβ antibody | II | NCT02565511 | Recruiting |
Vanutide cridificar (ACC-001) | Anti-Aβ vaccine | II | NCT00960531, etc. | Terminated |
Bapineuzumab (AAB-001) | Anti-Aβ monoclonal antibody | III | NCT00676143, etc. | Terminated |
Solanezumab (LY2062430) | Anti-Aβ IgG1 monoclonal antibody | III | NCT01127633 NCT01900665 NCT02760602 | Terminated |
II/III | NCT02008357 NCT01760005 | Active, not recruiting | ||
Ponezumab (PF-04360365) | Anti-Aβ IgG2 antibody | II | NCT00722046, NCT00945672 | Completed |
GSK933776 | Anti-Aβ antibody | I | NCT00459550, NCT01424436 | Completed, |
LY2599666 | Aβ antibody | I | NCT02614131 | Terminated |
Octagam® 10% | Immune globulin intravenous, 10% solution | III | NCT01736579 NCT01524887 | Terminated |
II/III | NCT01561053 NCT01300728 | Active, not recruiting | ||
II | NCT03319810 | Enrolling by invitation | ||
Aducanumab (BIIB037) | Anti-Aβ IgG1 monoclonal antibody | III | NCT02484547 NCT02477800 | Active, not recruiting |
II | NCT03639987 | Recruiting | ||
I | NCT01677572 | Active, not recruiting | ||
Crenezumab (MABT5102A, RG7412) | Anti-Aβ IgG4 antibody | I/II/III | NCT02670083 NCT01998841 NCT02353598 | Active, not recruiting |
III | NCT03491150 NCT03114657 | Recruiting | ||
Gantenerumab (R1450) | Anti-Aβ IgG1 antibody | III | NCT01224106 NCT02051608 | Active, not recruiting |
III | NCT03444870 NCT03443973 | Recruiting | ||
II/III | NCT01760005 | Active, not recruiting | ||
SAR228810 | Anti-Aβ monoclonal antibody | I | NCT01485302 | Completed |
Drugs Target Tau Production | ||||
TRx0014 | Methylene blue, tau aggregation inhibitor | II | NCT00684944 NCT00515333 | Completed |
LMTM (TRx0237) | Methylene blue, tau aggregation inhibitor | II/III | NCT03446001 | Recruiting |
NCT03539380 | Available | |||
III | NCT01689246 NCT01689233 | Completed | ||
Tideglusib (NP031112) | GSK3-β inhibitor, prevent tau hyperphosphorylation | I/II | NCT00948259 NCT01350362 | Completed |
ABBV-8E12 | Anti-tau antibody | II | NCT02880956 | Recruiting |
II | NCT03712787 | Not yet recruiting | ||
RO 7105705 | Anti-tau antibody | I | NCT02820896 | Completed |
II | NCT03289143 | Recruiting | ||
AADvac1 | Tau vaccine | I | NCT02031198 NCT01850238 | Completed |
II | NCT02579252 | Active, not recruiting | ||
TPI 287 | abeo-taxane, bind on tubulin, and stabilize microtubule | I | NCT01966666 | Active, not recruiting |
Drugs Target Inflammation | ||||
Ibuprofen | non-steroidal anti-inflammatory drugs (NSAIDs) | III | NCT02547818 | Recruiting |
Tarenflurbil | NSAIDs | III | NCT00380276 NCT00322036 | Terminated |
Salsalate | NSAIDs | I | NCT03277573 | Recruiting |
Celecoxib | NSAIDs | III | NCT00007189 | Completed |
Resveratrol | Phenol, antioxidant | II | NCT01504854 NCT01716637 NCT00678431 | Completed |
Etanercept | Tumor necrosis factor-alpha (TNF-α) inhibitor | I/II | NCT01068353 NCT01716637 | Completed |
Simvastatin | 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitor, cholesterol targeting | II | NCT00939822 | Active, not recruiting |
Neflamapimod (VX-745) | p38 mitogen-activated serine/threonine protein kinase p38 MAPK (p38 MAPKα) selective inhibitor | II | NCT03435861 NCT03402659 | Recruiting |
Azeliragon (TTP488) | Receptor for advanced glycation endproducts (RAGE) inhibitor | III | NCT02080364 NCT02916056 | Terminated |
Pioglitazone | Peroxisome-proliferator activated receptor γ (PPARγ) agonists | III | NCT01931566 NCT02284906 | Terminated |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Dong, Y.; Li, X.; Cheng, J.; Hou, L. Drug Development for Alzheimer’s Disease: Microglia Induced Neuroinflammation as a Target? Int. J. Mol. Sci. 2019, 20, 558. https://doi.org/10.3390/ijms20030558
Dong Y, Li X, Cheng J, Hou L. Drug Development for Alzheimer’s Disease: Microglia Induced Neuroinflammation as a Target? International Journal of Molecular Sciences. 2019; 20(3):558. https://doi.org/10.3390/ijms20030558
Chicago/Turabian StyleDong, Yuan, Xiaoheng Li, Jinbo Cheng, and Lin Hou. 2019. "Drug Development for Alzheimer’s Disease: Microglia Induced Neuroinflammation as a Target?" International Journal of Molecular Sciences 20, no. 3: 558. https://doi.org/10.3390/ijms20030558
APA StyleDong, Y., Li, X., Cheng, J., & Hou, L. (2019). Drug Development for Alzheimer’s Disease: Microglia Induced Neuroinflammation as a Target? International Journal of Molecular Sciences, 20(3), 558. https://doi.org/10.3390/ijms20030558