1932

Abstract

Alzheimer's disease (AD) is the primary cause of age-related dementia. Effective strategies to prevent and treat AD remain elusive despite major efforts to understand its basic biology and clinical pathophysiology. Significant investments in therapeutic drug discovery programs over the past two decades have yielded some important insights but no blockbuster drugs to alter the course of disease. Because significant memory loss and cognitive decline are associated with neuron death and loss of gray matter, especially in the frontal cortex and hippocampus, some focus in drug development has shifted to early prevention of cellular pathology. Although clinical trial design is challenging, due in part to a lack of robust biomarkers with predictive value, some optimism has come from the identification and study of inherited forms of early-onset AD and genetic risk factors that provide insights about molecular pathophysiology and potential drug targets. In addition, better understanding of the Aβ amyloid pathway and the tau pathway—leading to amyloid plaques and neurofibrillary tangles, respectively, which are histopathological hallmarks of AD—continues to drive significant drug research and development programs. The main focus of this review is to summarize the most recent basic biology, biochemistry, and pharmacology that serve as a foundation for more than 50 active advanced-phase clinical trials for AD prevention and therapy.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-042915-103753
2017-01-14
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/med/68/1/annurev-med-042915-103753.html?itemId=/content/journals/10.1146/annurev-med-042915-103753&mimeType=html&fmt=ahah

Literature Cited

  1. Winblad B, Amouyel P, Andrieu S. 1.  et al. 2016. Defeating Alzheimer's disease and other dementias: a priority for European science and society. Lancet Neurol 15:455–532 [Google Scholar]
  2. Winblad B, Poritis N. 2.  1999. Memantine in severe dementia: results of the 9M-Best Study (benefit and efficacy in severely demented patients during treatment with memantine). Int. J. Geriatr. Psychiatry 14:135–46 [Google Scholar]
  3. Pereira AC, Lambert HK, Grossman YS. 3.  et al. 2014. Glutamatergic regulation prevents hippocampal-dependent age-related cognitive decline through dendritic spine clustering. PNAS 111:18733–38 [Google Scholar]
  4. Goate A, Chartier-Harlin MC, Mullan M. 4.  et al. 1991. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease. Nature 349:704–6 [Google Scholar]
  5. Sherrington R, Rogaev EI, Liang Y. 5.  et al. 1995. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer's disease. Nature 375:754–60 [Google Scholar]
  6. Goate A, Hardy J. 6.  2012. Twenty years of Alzheimer's disease-causing mutations. J. Neurochem. 120:Suppl. 13–8 [Google Scholar]
  7. Hardy JA, Higgins GA. 7.  1992. Alzheimer's disease: the amyloid cascade hypothesis. Science 256:184–85 [Google Scholar]
  8. Hardy J, Selkoe DJ. 8.  2002. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297:353–56 [Google Scholar]
  9. McCarthy JV, Twomey C, Wujek P. 9.  2009. Presenilin-dependent regulated intramembrane proteolysis and gamma-secretase activity. Cell. Mol. Life Sci. 66:1534–55 [Google Scholar]
  10. Kopan R, Ilagan MX. 10.  2009. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell 137:216–33 [Google Scholar]
  11. Searfoss GH, Jordan WH, Calligaro DO. 11.  et al. 2003. Adipsin, a biomarker of gastrointestinal toxicity mediated by a functional gamma-secretase inhibitor. J. Biol. Chem. 278:46107–16 [Google Scholar]
  12. Wong GT, Manfra D, Poulet FM. 12.  et al. 2004. Chronic treatment with the gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J. Biol. Chem. 279:12876–82 [Google Scholar]
  13. Doody RS, Raman R, Farlow M. 13.  et al. 2013. A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N. Engl. J. Med. 369:341–50 [Google Scholar]
  14. Coric V, van Dyck CH, Salloway S. 14.  et al. 2012. Safety and tolerability of the gamma-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Arch. Neurol. 69:1430–40 [Google Scholar]
  15. Wolfe MS. 15.  2007. gamma-Secretase modulators. Curr. Alzheimer Res. 4:571–73 [Google Scholar]
  16. Teranishi Y, Inoue M, Yamamoto NG. 16.  et al. 2015. Proton myo-inositol cotransporter is a novel gamma-secretase associated protein that regulates Aβ production without affecting Notch cleavage. FEBS J 282:3438–51 [Google Scholar]
  17. Frykman S, Teranishi Y, Hur J-YY. 17.  et al. 2012. Identification of two novel synaptic γ-secretase associated proteins that affect amyloid β-peptide levels without altering Notch processing. Neurochem. Int. 61:108–18 [Google Scholar]
  18. Teranishi Y, Hur J-YY, Gu GJ. 18.  et al. 2012. Erlin-2 is associated with active γ-secretase in brain and affects amyloid β-peptide production. Biochem. Biophys. Res. Commun. 424:476–81 [Google Scholar]
  19. Kandalepas PC, Sadleir KR, Eimer WA. 19.  et al. 2013. The Alzheimer's beta-secretase BACE1 localizes to normal presynaptic terminals and to dystrophic presynaptic terminals surrounding amyloid plaques. Acta Neuropathol 126:329–52 [Google Scholar]
  20. Ghosh AK, Gemma S, Tang J. 20.  2008. beta-Secretase as a therapeutic target for Alzheimer's disease. Neurotherapeutics 5:399–408 [Google Scholar]
  21. Yan R, Vassar R. 21.  2014. Targeting the beta secretase BACE1 for Alzheimer's disease therapy. Lancet Neurol 13:319–29 [Google Scholar]
  22. Atwal JK, Chen Y, Chiu C. 22.  et al. 2011. A therapeutic antibody targeting BACE1 inhibits amyloid-beta production in vivo. Sci. Transl. Med. 3:84ra43 [Google Scholar]
  23. Arbel M, Yacoby I, Solomon B. 23.  2005. Inhibition of amyloid precursor protein processing by beta-secretase through site-directed antibodies. PNAS 102:7718–23 [Google Scholar]
  24. Mawuenyega KG, Sigurdson W, Ovod V. 24.  et al. 2010. Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science 330:1774 [Google Scholar]
  25. Selkoe DJ. 25.  2001. Clearing the brain's amyloid cobwebs. Neuron 32:177–80 [Google Scholar]
  26. Saido T, Leissring MA. 26.  2012. Proteolytic degradation of amyloid beta-protein. Cold Spring Harb. Perspect. Med. 2:a006379 [Google Scholar]
  27. Saito T, Iwata N, Tsubuki S. 27.  et al. 2005. Somatostatin regulates brain amyloid beta peptide Abeta42 through modulation of proteolytic degradation. Nat. Med. 11:434–39 [Google Scholar]
  28. Jacobsen JS, Comery TA, Martone RL. 28.  et al. 2008. Enhanced clearance of Abeta in brain by sustaining the plasmin proteolysis cascade. PNAS 105:8754–59 [Google Scholar]
  29. Mueller-Steiner S, Zhou Y, Arai H. 29.  et al. 2006. Antiamyloidogenic and neuroprotective functions of cathepsin B: implications for Alzheimer's disease. Neuron 51:703–14 [Google Scholar]
  30. Sun B, Zhou Y, Halabisky B. 30.  et al. 2008. Cystatin C-cathepsin B axis regulates amyloid beta levels and associated neuronal deficits in an animal model of Alzheimer's disease. Neuron 60:247–57 [Google Scholar]
  31. Schenk D, Barbour R, Dunn W. 31.  et al. 1999. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400:173–77 [Google Scholar]
  32. Morgan D, Diamond DM, Gottschall PE. 32.  et al. 2000. A beta peptide vaccination prevents memory loss in an animal model of Alzheimer's disease. Nature 408:982–85 [Google Scholar]
  33. Holmes C, Boche D, Wilkinson D. 33.  et al. 2008. Long-term effects of Abeta42 immunisation in Alzheimer's disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet 372:216–23 [Google Scholar]
  34. Solomon B, Koppel R, Hanan E. 34.  et al. 1996. Monoclonal antibodies inhibit in vitro fibrillar aggregation of the Alzheimer beta-amyloid peptide. PNAS 93:452–55 [Google Scholar]
  35. Legleiter J, Czilli DL, Gitter B. 35.  et al. 2004. Effect of different anti-Abeta antibodies on Abeta fibrillogenesis as assessed by atomic force microscopy. J. Mol. Biol. 335:997–1006 [Google Scholar]
  36. Solomon B, Koppel R, Frankel D. 36.  et al. 1997. Disaggregation of Alzheimer beta-amyloid by site-directed mAb. PNAS 94:4109–12 [Google Scholar]
  37. Frenkel D, Katz O, Solomon B. 37.  2000. Immunization against Alzheimer's beta-amyloid plaques via EFRH phage administration. PNAS 97:11455–59 [Google Scholar]
  38. Moreth J, Mavoungou C, Schindowski K. 38.  2013. Passive anti-amyloid immunotherapy in Alzheimer's disease: What are the most promising targets?. Immun. Ageing 10:18 [Google Scholar]
  39. Bohrmann B, Baumann K, Benz J. 39.  et al. 2012. Gantenerumab: a novel human anti-Abeta antibody demonstrates sustained cerebral amyloid-beta binding and elicits cell-mediated removal of human amyloid-beta. J. Alzheimers Dis. 28:49–69 [Google Scholar]
  40. Haass C, Selkoe DJ. 40.  2007. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta-peptide. Nat. Rev. Mol. Cell. Biol. 8:101–12 [Google Scholar]
  41. Englund H, Sehlin D, Johansson AS. 41.  et al. 2007. Sensitive ELISA detection of amyloid-beta protofibrils in biological samples. J. Neurochem. 103:334–45 [Google Scholar]
  42. Sevigny J, Chiao P, Bussière T. 42  et al. 2016. The antibody aducanumab reduces Aβ plaques in Alzheimer's disease. Nature 537:50–56 [Google Scholar]
  43. Hartl FU, Hayer-Hartl M. 43.  2002. Molecular chaperones in the cytosol: from nascent chain to folded protein. Science 295:1852–58 [Google Scholar]
  44. Arosio P, Michaels TC, Linse S. 44.  et al. 2016. Kinetic analysis reveals the diversity of microscopic mechanisms through which molecular chaperones suppress amyloid formation. Nat. Commun. 7:10948 [Google Scholar]
  45. Gupta R, Kapoor N, Raleigh DP. 45.  et al. 2012. Nucleobindin 1 caps human islet amyloid polypeptide protofibrils to prevent amyloid fibril formation. J. Mol. Biol. 421:378–89 [Google Scholar]
  46. Xu LQ, Wu S, Buell AK. 46.  et al. 2013. Influence of specific HSP70 domains on fibril formation of the yeast prion protein Ure2. Philos. Trans. R. Soc. Lond. B Biol. Sci. 368:20110410 [Google Scholar]
  47. Muchowski PJ. 47.  2002. Protein misfolding, amyloid formation, and neurodegeneration: a critical role for molecular chaperones?. Neuron 35:9–12 [Google Scholar]
  48. Knowles TP, Vendruscolo M, Dobson CM. 48.  2014. The amyloid state and its association with protein misfolding diseases. Nat. Rev. Mol. Cell. Biol. 15:384–96 [Google Scholar]
  49. Cohen SI, Vendruscolo M, Dobson CM. 49.  et al. 2012. From macroscopic measurements to microscopic mechanisms of protein aggregation. J. Mol. Biol. 421:160–71 [Google Scholar]
  50. Miura K, Titani K, Kurosawa Y. 50.  et al. 1992. Molecular cloning of nucleobindin, a novel DNA-binding protein that contains both a signal peptide and a leucine zipper structure. Biochem. Biophys. Res. Commun. 187:375–80 [Google Scholar]
  51. Lin P, Li F, Zhang YW. 51.  et al. 2007. Calnuc binds to Alzheimer's beta-amyloid precursor protein and affects its biogenesis. J. Neurochem. 100:1505–14 [Google Scholar]
  52. Wang Y, Mandelkow E. 52.  2016. Tau in physiology and pathology. Nat. Rev. Neurosci. 17:5–21 [Google Scholar]
  53. Caillet-Boudin M-LL, Buée L, Sergeant N. 53.  et al. 2015. Regulation of human MAPT gene expression. Mol. Neurodegeneration 10:28 [Google Scholar]
  54. Ke YD, Suchowerska AK, van der Hoven J. 54.  et al. 2012. Lessons from tau-deficient mice. Int. J. Alzheimer's Dis 2012:87327 [Google Scholar]
  55. Roberson ED, Scearce-Levie K, Palop JJ. 55.  et al. 2007. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model. Science 316:750–54 [Google Scholar]
  56. Leroy K, Ando K, Laporte V. 56.  et al. 2012. Lack of tau proteins rescues neuronal cell death and decreases amyloidogenic processing of APP in APP/PS1 mice. Am. J. Pathol. 181:1928–40 [Google Scholar]
  57. DeVos SL, Goncharoff DK, Chen G. 57.  et al. 2013. Antisense reduction of tau in adult mice protects against seizures. J. Neurosci. 33:12887–97 [Google Scholar]
  58. Santa-Maria I, Alaniz ME, Renwick N. 58.  et al. 2015. Dysregulation of microRNA-219 promotes neurodegeneration through post-transcriptional regulation of tau. J. Clin. Investig. 125:681–86 [Google Scholar]
  59. Barten DM, Fanara P, Andorfer C. 59.  et al. 2012. Hyperdynamic microtubules, cognitive deficits, and pathology are improved in tau transgenic mice with low doses of the microtubule-stabilizing agent BMS-241027. J. Neurosci. 32:7137–45 [Google Scholar]
  60. Zhang B, Carroll J, Trojanowski JQ. 60.  et al. 2012. The microtubule-stabilizing agent, epothilone D, reduces axonal dysfunction, neurotoxicity, cognitive deficits, and Alzheimer-like pathology in an interventional study with aged tau transgenic mice. J. Neurosci. 32:3601–11 [Google Scholar]
  61. Jeganathan S, von Bergen M, Brutlach H. 61.  et al. 2006. Global hairpin folding of tau in solution. Biochemistry 45:2283–93 [Google Scholar]
  62. Wischik CM, Novak M, Thøgersen HC. 62.  et al. 1988. Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimer disease. PNAS 85:4506–10 [Google Scholar]
  63. Wegmann S, Medalsy ID, Mandelkow E. 63.  et al. 2013. The fuzzy coat of pathological human Tau fibrils is a two-layered polyelectrolyte brush. PNAS 110: 21:E313–E321 [Google Scholar]
  64. von Bergen M, Friedhoff P, Biernat J. 64.  et al. 2000. Assembly of tau protein into Alzheimer paired helical filaments depends on a local sequence motif ((306)VQIVYK(311)) forming beta structure. PNAS 97:5129–34 [Google Scholar]
  65. Peeraer E, Bottelbergs A, Van Kolen K. 65.  et al. 2015. Intracerebral injection of preformed synthetic tau fibrils initiates widespread tauopathy and neuronal loss in the brains of tau transgenic mice. Neurobiol. Dis. 73:83–95 [Google Scholar]
  66. Clavaguera F, Hench J, Lavenir I. 66.  et al. 2014. Peripheral administration of tau aggregates triggers intracerebral tauopathy in transgenic mice. Acta Neuropathol 127:299–301 [Google Scholar]
  67. Sanders DW, Kaufman SK, DeVos SL. 67.  et al. 2014. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 82:1271–88 [Google Scholar]
  68. Hochgräfe K, Sydow A, Matenia D. 68.  et al. 2015. Preventive methylene blue treatment preserves cognition in mice expressing full-length pro-aggregant human Tau. Acta Neuropathol. Commun. 3:25 [Google Scholar]
  69. Baddeley TC, McCaffrey J, Storey JM. 69.  et al. 2015. Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimer's disease. J. Pharmacol. Exp. Ther. 352:110–18 [Google Scholar]
  70. Theunis C, Crespo-Biel N, Gafner V. 70.  et al. 2013. Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau.P301L mice that model tauopathy. PLOS ONE 8:e72301 [Google Scholar]
  71. Kontsekova E, Zilka N, Kovacech B. 71.  et al. 2014. 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. Alzheimers Res. Ther. 6:44 [Google Scholar]
  72. Pedersen JT, Sigurdsson EM. 72.  2015. Tau immunotherapy for Alzheimer's disease. Trends Mol. Med. 21:394–402 [Google Scholar]
  73. Hanisch UK, Kettenmann H. 73.  2007. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 10:1387–94 [Google Scholar]
  74. Franceschi C, Bonafe M, Valensin S. 74.  et al. 2000. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 908:244–54 [Google Scholar]
  75. Streit WJ, Sammons NW, Kuhns AJ. 75.  et al. 2004. Dystrophic microglia in the aging human brain. Glia 45:208–12 [Google Scholar]
  76. Wes PD, Holtman IR, Boddeke EW. 76.  et al. 2016. Next generation transcriptomics and genomics elucidate biological complexity of microglia in health and disease. Glia 64:197–213 [Google Scholar]
  77. Orre M, Kamphuis W, Osborn LM. 77.  et al. 2014. Acute isolation and transcriptome characterization of cortical astrocytes and microglia from young and aged mice. Neurobiol. Aging 35:1–14 [Google Scholar]
  78. Ulland TK, Wang Y, Colonna M. 78.  2015. Regulation of microglial survival and proliferation in health and diseases. Semin. Immunol. 27:410–15 [Google Scholar]
  79. Ransohoff RM. 79.  2009. Chemokines and chemokine receptors: standing at the crossroads of immunobiology and neurobiology. Immunity 31:711–21 [Google Scholar]
  80. Aisen PS, Gauthier S, Vellas B. 80.  et al. 2007. Alzhemed: a potential treatment for Alzheimer's disease. Curr. Alzheimer Res. 4:473–78 [Google Scholar]
  81. Yan SD, Chen X, Fu J. 81.  et al. 1996. RAGE and amyloid-beta peptide neurotoxicity in Alzheimer's disease. Nature 382:685–91 [Google Scholar]
  82. Lue LFF, Yan SD, Stern DM. 82.  et al. 2005. Preventing activation of receptor for advanced glycation endproducts in Alzheimer's disease. Curr. Drug Targets CNS Neurol. Disord. 4:249–66 [Google Scholar]
  83. Gómez-Nicola D, Fransen NL, Suzzi S. 83.  et al. 2013. Regulation of microglial proliferation during chronic neurodegeneration. J. Neurosci. 33:2481–93 [Google Scholar]
  84. Gomez-Nicola D, Perry VH. 84.  2015. Microglial dynamics and role in the healthy and diseased brain: a paradigm of functional plasticity. Neuroscientist 21:169–84 [Google Scholar]
  85. Li T, Pang S, Yu Y. 85.  et al. 2013. Proliferation of parenchymal microglia is the main source of microgliosis after ischaemic stroke. Brain J. Neurol. 136:3578–88 [Google Scholar]
  86. Miguel-Alvarez M, Santos-Lozano A, Sanchis-Gomar F. 86.  et al. 2015. Non-steroidal anti-inflammatory drugs as a treatment for Alzheimer's disease: a systematic review and meta-analysis of treatment effect. Drugs Aging 32:139–47 [Google Scholar]
  87. Torrent M, Pulido D, Nogués MV. 88.  et al. 2012. Exploring new biological functions of amyloids: bacteria cell agglutination mediated by host protein aggregation. PLOS Pathog 8:11e1003005 [Google Scholar]
  88. Kagan BL. 89.  2011. Antimicrobial amyloids?. Biophys. J. 100:1597–98 [Google Scholar]
  89. Kagan BL, Jang H, Capone R. 90.  et al. 2012. Antimicrobial properties of amyloid peptide. Mol. Pharm. 9:708–17 [Google Scholar]
  90. Kumar DKV, Choi SH, Washicosky KJ. 101.  et al. 2016. Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer's disease. Sci. Transl. Med. 8:340ra72 [Google Scholar]
  91. Peila R, Rodriguez BL, Launer LJ. 91.  et al. 2002. Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: the Honolulu-Asia Aging Study. Diabetes 51:1256–62 [Google Scholar]
  92. Hughes TF, Borenstein AR, Schofield E. 92.  et al. 2009. Association between late-life body mass index and dementia: the Kame Project. Neurology 72:1741–46 [Google Scholar]
  93. Kivipelto M, Ngandu T, Fratiglioni L. 93.  et al. 2005. Obesity and vascular risk factors at midlife and the risk of dementia and Alzheimer disease. Arch. Neurol. 62:1556–60 [Google Scholar]
  94. Whitmer RA, Gunderson EP, Quesenberry CP. 94.  et al. 2007. Body mass index in midlife and risk of Alzheimer disease and vascular dementia. Curr. Alzheimer Res. 4:103–9 [Google Scholar]
  95. Martin B, Mattson MP, Maudsley S. 95.  2006. Caloric restriction and intermittent fasting: two potential diets for successful brain aging. Ageing Res. Rev. 5:332–53 [Google Scholar]
  96. Scarmeas N, Luchsinger JA, Schupf N. 96.  et al. 2009. Physical activity, diet, and risk of Alzheimer disease. JAMA 302:627–37 [Google Scholar]
  97. Frölich L, Blum-Degen D, Bernstein HG. 97.  et al. 1998. Brain insulin and insulin receptors in aging and sporadic Alzheimer's disease. J. Neural Trans. 105:423–38 [Google Scholar]
  98. Qiu WQ, Folstein MF. 98.  2006. Insulin, insulin-degrading enzyme and amyloid-beta peptide in Alzheimer's disease: review and hypothesis. Neurobiol. Aging 27:190–98 [Google Scholar]
  99. Heneka MT, Sastre M, Dumitrescu-Ozimek L. 99.  et al. 2005. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain J. Neurol. 128:1442–53 [Google Scholar]
  100. Pedersen WA, McMillan PJ, Kulstad JJ. 100.  et al. 2006. Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Exp. Neurol. 199:265–73 [Google Scholar]
  101. Risner ME, Saunders AM, Altman JF. 87.  et al. 2006. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer's disease. Pharmacogenom. J. 6:246–54 [Google Scholar]
  102. Jack CR Jr., Bennett DA, Blennow K. 102.  et al. 2016. A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 87:539–47 [Google Scholar]
/content/journals/10.1146/annurev-med-042915-103753
Loading
/content/journals/10.1146/annurev-med-042915-103753
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error