725
Views
68
CrossRef citations to date
0
Altmetric
Drug Profile

Solanezumab for the treatment of mild-to-moderate Alzheimer’s disease

, , , , , , & show all
Pages 135-149 | Published online: 10 Jan 2014

References

  • Alzheimer’s Association, Thies W, Bleiler L. 2011 Alzheimer’s disease facts and figures. Alzheimers Dement.7(2), 208–244 (2011).
  • Frisardi V, Solfrizzi V, Imbimbo PB et al. Towards disease-modifying treatment of Alzheimer’s disease: drugs targeting β-amyloid. Curr. Alzheimer Res.7(1), 40–55 (2010).
  • Small SA, Duff K. Linking Aβ and tau in late-onset Alzheimer's disease: a dual pathway hypothesis. Neuron60(4), 534–542 (2008).
  • Walter J, Kaether C, Steiner H, Haass C. The cell biology of Alzheimer’s disease: uncovering the secrets of secretases. Curr. Opin. Neurobiol.11(5), 585–590 (2001).
  • Hardy J, Allsop D. Amyloid deposition as the central event in the etiology of Alzheimer’s disease. Trends Pharmacol. Sci.12(10), 383–388 (1991).
  • Shankar GM, Li S, Mehta TH et al. Amyloid-β protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat. Med.14(8), 837–842 (2008).
  • Klyubin I, Betts V, Welzel AT et al. Amyloid β protein dimer-containing human CSF disrupts synaptic plasticity: prevention by systemic passive immunization. J. Neurosci.28(16), 4231–4237 (2008).
  • Mc Donald JM, Savva GM, Brayne C et al. Medical Research Council Cognitive Function and Ageing Study. The presence of sodium dodecyl sulphate-stable Aβ dimers is strongly associated with Alzheimer-type dementia. Brain133(Pt 5), 1328–1341 (2010).
  • Jin M, Shepardson N, Yang T, Chen G, Walsh D, Selkoe DJ. Soluble amyloid β-protein dimers isolated from Alzheimer cortex directly induce tau hyperphosphorylation and neuritic degeneration. Proc. Natl Acad. Sci. USA108(14), 5819–5824 (2011).
  • Selkoe DJ. Soluble oligomers of the amyloid β-protein impair synaptic plasticity and behavior. Behav. Brain Res.192(1), 106–113 (2008).
  • Shankar GM, Bloodgood BL, Townsend M et al. Natural oligomers of the Alzheimer amyloid-β protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. J. Neurosci.27(11), 2866–2875 (2007).
  • Lesné S, Koh MT, Kotilinek L et al. A specific amyloid-β protein assembly in the brain impairs memory. Nature440(7082), 352–357 (2006).
  • Lee EB, Leng LZ, Zhang B et al. Targeting amyloid-β peptide (Aβ) oligomers by passive immunization with a conformation-selective monoclonal antibody improves learning and memory in Aβ precursor protein (APP) transgenic mice. J. Biol. Chem.281(7), 4292–4299 (2006).
  • Lambert MP, Velasco PT, Chang L et al. Monoclonal antibodies that target pathological assemblies of Aβ. J. Neurochem.100(1), 23–35 (2007).
  • Mangialasche F, Solomon A, Winblad B, Mecocci P, Kivipelto M. Alzheimer’s disease: clinical trials and drug development. Lancet Neurol.9(7), 702–716 (2010).
  • Panza F, Solfrizzi V, Frisardi V et al. Disease-modifying approach to the treatment of Alzheimer’s disease: from alpha-secretase activators to gamma-secretase inhibitors and modulators. Drugs Aging26(7), 537–555 (2009).
  • Imbimbo BP, Panza F, Frisardi V et al. Therapeutic intervention for Alzheimer’s disease with γ-secretase inhibitors: still a viable option? Expert Opin. Investig. Drugs20(3), 325–341 (2011).
  • Vellas B, Sol O, Snyder PJ et al. EHT0202 in Alzheimer’s disease: a 3-month, randomized, placebo-controlled, double-blind study. Curr. Alzheimer Res.8(2), 203–212 (2011).
  • Lannfelt L, Blennow K, Zetterberg H et al. PBT2–201-EURO study group. Safety, efficacy, and biomarker findings of PBT2 in targeting Abeta as a modifying therapy for Alzheimer’s disease: a Phase IIa, double-blind, randomized, placebo-controlled trial. Lancet Neurol.7(9), 779–786 (2008).
  • Faux NG, Ritchie CW, Gunn A et al. PBT2 rapidly improves cognition in Alzheimer’s disease: additional Phase II analyses. J. Alzheimers Dis.20(2), 509–516 (2010).
  • Lemere CA, Masliah E. Can Alzheimer disease be prevented by amyloid-beta immunotherapy? Nat. Rev. Neurol.6(2), 108–119 (2010).
  • Gilman S, Koller M, Black RS et al. AN1792(QS-21)-201 Study Team. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology64(9), 1553–1562 (2005).
  • Jicha GA. Is passive immunization for Alzheimer’s disease ‘alive and well’ or ‘dead and buried’? Expert Opin. Biol. Ther.9(4), 481–491 (2009).
  • Bard F, Cannon C, Barbour R et al. Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat. Med.6(8), 916–919 (2000).
  • Cattepoel S, Hanenberg M, Kulic L, Nitsch RM. Chronic intranasal treatment with an anti-Aβ(30–42) scFv antibody ameliorates amyloid pathology in a transgenic mouse model of Alzheimer’s disease. PLoS One6(4), e18296 (2011).
  • Lichtlen P, Mohajeri MH. Antibody-based approaches in Alzheimer’s research: safety, pharmacokinetics, metabolism, and analytical tools. J. Neurochem.104, 859–874 (2008).
  • Panza F, Frisardi V, Imbimbo BP, Seripa D, Solfrizzi V, Pilotto A. Monoclonal antibodies against β-amyloid (Aβ) for the treatment of Alzheimer’s disease: the Aβ target at a crossroads. Expert Opin. Biol. Ther.11(6), 679–686 (2011).
  • Kerchner GA, Boxer AL. Bapineuzumab. Expert Opin. Biol. Ther.10(7), 1121–1130 (2010).
  • Panza F, Frisardi V, Imbimbo BP et al. Bapineuzumab: anti-β-amyloid monoclonal antibodies for the treatment of Alzheimer’s disease. Immunotherapy2(2), 767–782 (2010).
  • Black RS, Sperling RA, Safirstein B et al. A single ascending dose study of bapineuzumab in patients with Alzheimer disease. Alzheimer Dis. Assoc. Disord.24(2), 198–203 (2010).
  • Rinne JO, Brooks DJ, Rossor MN et al. 11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients with Alzheimer’s disease treated with bapineuzumab: a Phase II, double-blind, placebo-controlled, ascending-dose study. Lancet Neurol.9(4), 363–372 (2010).
  • Salloway S, Sperling R, Gilman S et al. A Phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology73(24), 2061–2070 (2009).
  • Panza F, Frisardi V, Imbimbo BP et al. Anti-β-amyloid immunotherapy for Alzheimer’s disease: focus on bapineuzumab. Curr. Alzheimer Res. doi:10.2174/1567211215920552050 (2011) (Epub ahead of print).
  • Samadi H, Sultzer D. Solanezumab for Alzheimer’s disease. Expert Opin. Biol. Ther.11(6), 787–798 (2011).
  • DeMattos RB, Racke MM, Gelfanova V et al. Identification, characterization, and comparison of amino-terminally truncated Aβ42 peptides in Alzheimer’s disease brain tissue and in plasma from Alzheimer’s patients receiving solanezumab immunotherapy treatment. Alzheimers Dement.5(4 Suppl.), P156–P157 (2009).
  • Watts RJ, Chen M, Atwal J et al. Selection of an anti-Aβ antibody that binds various forms of Aβ and blocks toxicity both in vitro and in vivo. Alzheimers Dement.5(4 Suppl.), P426 (2009).
  • Pul R, Dodel R, Stangel M. Antibody-based therapy in Alzheimer’s disease. Expert Opin. Biol. Ther.11(3), 343–357 (2011).
  • Relkin NR. Natural human antibodies targeting amyloid aggregates in intravenous immunoglobulin. Alzheimers Dement.4, T101 (2008).
  • Du Y, Wei X, Dodel R et al. Human anti-β-amyloid antibodies block β-amyloid fibril formation and prevent β-amyloid-induced neurotoxicity. Brain126, 1935–1939 (2003).
  • Ma QL, Lim GP, Harris-White ME et al. Antibodies against β-amyloid reduce Aβ oligomers, glycogen synthase kinase-3β activation and τ phosphorylation in vivo and in vitro. J. Neurosci. Res.83, 374–384 (2006).
  • Istrin G, Bosis E, Solomon B. Intravenous immunoglobulin enhances the clearance of fibrillar amyloid-β peptide. J. Neurosci. Res.84, 434–443 (2006).
  • Dodel RC, Du Y, Depboylu C et al. Intravenous immunoglobulins containing antibodies against β-amyloid for the treatment of Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry75, 1472–1474 (2004).
  • Relkin NR, Szabo P, Adamiak B et al. 18-month study of intravenous immunoglobulin for treatment of mild Alzheimer disease. Neurobiol. Aging30, 1728–1736 (2009).
  • Seubert P, Vigo-Pelfrey C, Esch F et al. Isolation and quantification of soluble Alzheimer’s beta-peptide from biological fluids. Nature359(6393), 325–327 (1992).
  • Legleiter J, Czilli DL, Gitter B, DeMattos RB, Holtzman DM, Kowalewski T. Effect of different anti-Aβ antibodies on Aβ fibrillogenesis as assessed by atomic force microscopy. J. Mol. Biol.35(4), 997–1006 (2004).
  • DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM. Holtzman DM. Peripheral anti-Aβ antibody alters CNS and plasma Aβ clearance and decreases brain Aβ burden in a mouse model of Alzheimer's disease. Proc. Natl Acad. Sci. USA98(15), 8850–8855 (2001).
  • DeMattos RB, Bales K, Cummins DJ, Paul SM, Holtzman DM. Brain to plasma amyloid-beta efflux: a measure of brain amyloid burden in a mouse model of Alzheimer’s disease. Science295(5563), 2264 –2267 (2002).
  • Dodart JC, Bales KR, Gannon KS et al. Immunization reverses memory deficits without reducing brain Aβ burden in Alzheimer's disease model. Nat. Neurosci.5(5), 452–457 (2002).
  • Racke MM, Boone LI, Hepburn DL et al. Exacerbation of cerebral amyloid angiopathy-associated microhemorrhage in amyloid precursor protein transgenic mice by immunotherapy is dependent on antibody recognition of deposited forms of amyloid β. J. Neurosci.25(2), 629–636 (2005).
  • DeMattos RB, Racke MM, Gelfanova V et al. Characterization of amino-terminally truncated Abeta-42 peptides in plasma from Alzheimer’s patients receiving solanezumab immunotherapy treatment. Alzheimers Dement.6(Suppl. 1), S542 (2010).
  • Seubert P, Barbour R, Khan K et al. Antibody capture of soluble Abeta does not reduce cortical Abeta amyloidosis in the PDAPP mouse. Neurodegener. Dis.5(2), 65–71 (2008).
  • Morgan D. Immunotherapy for Alzheimer’s disease. J. Intern. Med.269(1), 54–63 (2011).
  • Yamada K, Yabuki C, Seubert P et al. Ab immunotherapy: intracerebral sequestration of Abeta by an anti-Abeta monoclonal antibody 266 with high affinity to soluble Abeta. J. Neurosci.29(36), 11393–11398 (2009).
  • Golde TE, Das P, Levites Y. Quantitative and mechanistic studies of Abeta immunotherapy. CNS Neurol. Disord. Drug Targets8(1), 31–49 (2009).
  • Siemers ER, Friedrich S, Dean RA et al. Safety and changes in plasma and cerebrospinal fluid amyloid β after a single administration of an amyloid β monoclonal antibody in subjects with Alzheimer disease. Clin. Neuropharmacol.33(2), 67–73 (2010).
  • Walsh DM, Selkoe DJ. Aβ oligomers – a decade of discovery. J. Neurochem.101(5), 1172–1184 (3007).
  • Paul SM, DeMattos RB, Wu X et al. Plasma Abeta levels in young APP V717F transgenic mice following anti – Abeta antibody administration predict brain amyloid burden at an older age. Presented at: 33rd Annual Meeting of the Society for Neuroscience. New Orleans, LA, USA, 8–12 November 2003 (Abstract 842.19).
  • Gitter BD, Hepburn DL, Cummins DJ et al. Passive immunization of APPV717F transgenic mice with mid-domain- or amino-terminal-reactive anti-Abeta antibodies produce differential effects on immunoreactive Abeta burden and fibrillar (thioflavin-S positive) plaque deposits. Presented at: 33rd Annual Meeting of the Society for Neuroscience. New Orleans, USA, 8–12 November 2003 (Abstract 201.9).
  • Gitter BD, Gannon KS, Cummins DJ et al. Reduction in brain amyloid beta burden and reversal of memory impairment in APP V717F transgenic mice following chronic administration of the anti-amyloid beta antibody m266. Neurobiol. Aging23(1), S105 (2002).
  • Coyle JT, Price Dl, DeLong MR. Alzheimers disease – a disorder of cortical cholinergic innervation. Science219(4589), 1184–1190 (1983).
  • Whitehouse PJ, Price DL, Clark AW, Coyle JT, DeLong MR. Alzheimer-disease–evidence for selective loss of cholinergic neurons in the nucleus basalis. Ann. Neurol.10(2), 122–126 (1981).
  • Bales KR, Tzavara ET, Paul SM, Bymaster FP, Nomikos GG. Cholinergic dysfunction in APP V717F transgenic mice is normalized following anti-Abeta antibody administration. Presented at: 33rd Annual Meeting of the Society for Neuroscience. New Orleans, USA, 8–12 November 2003 (Abstract 133.1).
  • Bales KR, Tzavara ET, Wu S et al. Cholinergic dysfunction in a mouse model of Alzheimer disease is reversed by an anti-A beta antibody. J. Clin. Invest.116(3), 825–832 (2006).
  • Wu S, Nomikos GG, Bales KR. Modulation of high-affinity choline uptake by the abeta peptide. Presented at: 35th Annual Meeting of the Society for Neuroscience. Washington, DC, USA, 12–16 November 2005 (Abstract 422.2).
  • Brody DL, MacDonald C, Fujimoto S et al. Anti-Abeta antibody attenuates cognitive impairment in a model of experimental traumatic brain injury. Presented at: 35th Annual Meeting of the Society for Neuroscience. Washington, DC, USA, 12–16 November 2005 (Abstract 335.4).
  • Schroeter S, Khan K, Barbour R et al. Immunotherapy reduces vascular amyloid-beta in PDAPP mice. J. Neurosci.28(27), 6787–6793 (2008).
  • Pfeifer M, Boncristiano S, Bondolfi L et al. Cerebral hemorrhage after passive anti-Aβ immunotherapy. Science298(5597), 1379 (2002).
  • Siemers ER, Friedrich S, Dean RA et al. Safety, tolerability and biomarker effects of an Aβ monoclonal antibody administered to patients with Alzheimer’s disease. Alzheimers Dement.4(Suppl. 2), T774 (2008).
  • Goto T, Fukjikoshi S, Uenaka K et al. Solanezumab was safe and well-tolerated for Asian patients with mild-to-moderate Alzheimer’s disease in a multicenter, randomized, open-label, multi-dose study. Alzheimers Dement.6(Suppl. 4), S308 (2010).
  • Carlson C, Estergard W, Oh J et al. Prevalence of asymptomatic vasogenic edema in pretreatment Alzheimer’s disease study cohorts from Phase III trials of semagacestat and solanezumab. Alzheimers Dement.7(4), 396–401 (2011).
  • Britschgi M, Olin CE, Johns HT et al. Neuroprotective natural antibodies to assemblies of amyloidogenic peptides decrease with normal aging and advancing Alzheimer’s disease. Proc. Natl Acad. Sci. USA106(29), 12145–12150 (2009).
  • Weksler ME, Relkin N, Turkenich R et al. Patients with Alzheimer’s disease have lower levels of serum anti-amyloid peptide antibodies than healthy elderly individuals. Exp. Gerontol.37(7), 943–948 (2002).
  • Chen WH, Kozlovsky BF, Effros RB, Grubeck-Loebenstein B, Edelman R, Sztein MB. Vaccination in the elderly: an immunological perspective. Trends Immunol.30(7), 351–359 (2009).
  • Holmes C, Boche D, Wilkinson D et al. Long-term effects of Aβ42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled Phase I trial. Lancet372(9634), 216–223 (2008).
  • Siemers E. Evaluating semagacestat, a gamma-secretase inhibitor, in a Phase III trial. Presented at: Alzheimer’s Association International Conference 2011. Paris, France, 16–21 July 2011.
  • Bateman RJ, Siemers ER, Mawuenyega KG et al. A gamma-secretase inhibitor decreases amyloid-beta production in the central nervous system. Ann. Neurol.66(1), 48–54 (2009).
  • Castellani RJ, Lee HG, Siedlak SL et al. Reexamining Alzheimer’s disease: evidence for a protective role for amyloid-beta protein precursor and amyloid-beta. J. Alzheimers Dis.18(2), 447–452 (2009).
  • McKhann GM, Knopman DS, Chertkow H et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement.7(3), 263–269 (2011).
  • Sperling RA, Aisen PS, Beckett LA et al. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement.7(3), 280–292 (2011).
  • Dubois B, Feldman HH, Jacova C et al. Revising the definition of Alzheimer’s disease: a new lexicon. Lancet Neurol.9(11), 1118–1127 (2010).

Patents

  • Eli Lilly and Company; Jia AY, Tsurushita N, Vasquez M. Anti-amyloid beta peptide antibodies. WO/2003/016466 (2003).
  • Raso V. Immunological control of beta-amyloid levels in vivo. WO 01/62801 (2005).

Websites

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.