Skip to main content
Top
Published in: Alzheimer's Research & Therapy 1/2018

Open Access 01-12-2018 | Research

SAR228810: an antibody for protofibrillar amyloid β peptide designed to reduce the risk of amyloid-related imaging abnormalities (ARIA)

Authors: Laurent Pradier, Véronique Blanchard-Brégeon, Andrees Bohme, Thomas Debeir, Jean Menager, Patrick Benoit, Pascal Barneoud, Véronique Taupin, Philippe Bertrand, Philippe Dugay, Béatrice Cameron, Yi Shi, Souad Naimi, Marc Duchesne, Marie Gagnaire, Tim Weeden, Tara Travaline, David Reczek, Leonard Khiroug, Mohamed Slaoui, Pascale Brunel, Hidehiro Fukuyama, Jeffrey Ravetch, Thierry Canton, Caroline Cohen

Published in: Alzheimer's Research & Therapy | Issue 1/2018

Login to get access

Abstract

Background

Anti-amyloid β (Aβ) immunotherapy represents a major area of drug development for Alzheimer’s disease (AD). However, Aβ peptide adopts multiple conformations and the pathological forms to be specifically targeted have not been identified. Aβ immunotherapy-related vasogenic edema has also been severely dose limiting for antibodies with effector functions binding vascular amyloid such as bapineuzumab. These two factors might have contributed to the limited efficacy demonstrated so far in clinical studies.

Methods

To address these limitations, we have engineered SAR228810, a humanized monoclonal antibody (mAb) with limited Fc effector functions that binds specifically to soluble protofibrillar and fibrillar forms of Aβ peptide and we tested it together with its murine precursor SAR255952 in vitro and in vivo.

Results

Unlike gantenerumab and BAN2401, SAR228810 and SAR255952 do not bind to Aβ monomers, low molecular weight Aβ oligomers or, in human brain sections, to Aβ diffuse deposits which are not specific of AD pathology. Both antibodies prevent Aβ42 oligomer neurotoxicity in primary neuronal cultures. In vivo, SAR255952, a mouse aglycosylated IgG1, dose-dependently prevented brain amyloid plaque formation and plaque-related inflammation with a minimal active dose of 3 mg/kg/week by the intraperitoneal route. No increase in plasma Aβ levels was observed with SAR255952 treatment, in line with its lack of affinity for monomeric Aβ. The effects of SAR255952 translated into synaptic functional improvement in ex-vivo hippocampal slices. Brain penetration and decoration of cerebral amyloid plaques was documented in live animals and postmortem. SAR255952 (up to 50 mg/kg/week intravenously) did not increase brain microhemorrhages and/or microscopic changes in meningeal and cerebral arteries in old APPSL mice while 3D6, the murine version of bapineuzumab, did. In immunotolerized mice, the clinical candidate SAR228810 demonstrated the same level of efficacy as the murine SAR255952.

Conclusion

Based on the improved efficacy/safety profile in non-clinical models of SAR228810, a first-in-man single and multiple dose administration clinical study has been initiated in AD patients.
Appendix
Available only for authorised users
Literature
1.
go back to reference Holtzman DM, et al. Mapping the road forward in Alzheimer’s disease. Sci Transl Med. 2011;3(114):114ps48.CrossRef Holtzman DM, et al. Mapping the road forward in Alzheimer’s disease. Sci Transl Med. 2011;3(114):114ps48.CrossRef
2.
go back to reference Bertram L, Lill CM, Tanzi RE. The genetics of Alzheimer disease: back to the future. Neuron. 2010;68(2):270–81.CrossRef Bertram L, Lill CM, Tanzi RE. The genetics of Alzheimer disease: back to the future. Neuron. 2010;68(2):270–81.CrossRef
3.
go back to reference Jonsson T, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;488(7409):96–9.CrossRef Jonsson T, et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature. 2012;488(7409):96–9.CrossRef
4.
go back to reference Liu J, et al. Antibody-based drugs and approaches against amyloid-beta species for Alzheimer’s disease immunotherapy. Drugs Aging. 2016;33(10):685–97.CrossRef Liu J, et al. Antibody-based drugs and approaches against amyloid-beta species for Alzheimer’s disease immunotherapy. Drugs Aging. 2016;33(10):685–97.CrossRef
5.
go back to reference MerckMSD, Merck announces discontinuation of APECS study evaluating verubecestat [MK-8931] for the treatment of people with prodromal Alzheimer’s Disease. 2018. MerckMSD, Merck announces discontinuation of APECS study evaluating verubecestat [MK-8931] for the treatment of people with prodromal Alzheimer’s Disease. 2018.
6.
go back to reference Jack CR Jr, Holtzman DM. Biomarker modeling of Alzheimer’s disease. Neuron. 2013;80(6):1347–58.CrossRef Jack CR Jr, Holtzman DM. Biomarker modeling of Alzheimer’s disease. Neuron. 2013;80(6):1347–58.CrossRef
7.
go back to reference Sevigny J, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–6.CrossRef Sevigny J, et al. The antibody aducanumab reduces Abeta plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–6.CrossRef
8.
go back to reference Bohrmann B, 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 Alzheimers Dis. 2012;28(1):49–69.CrossRef Bohrmann B, 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 Alzheimers Dis. 2012;28(1):49–69.CrossRef
9.
go back to reference Voyle N, et al. The effect of low doses of gantenerumab on amyloid and tau biomarkers in cerebrospinal fluid (CSF) in the Marguerite Road Study. Alzheimer’s Assoc Int Congr. 2018:Abst ID23224. Voyle N, et al. The effect of low doses of gantenerumab on amyloid and tau biomarkers in cerebrospinal fluid (CSF) in the Marguerite Road Study. Alzheimer’s Assoc Int Congr. 2018:Abst ID23224.
10.
go back to reference Eisai, Eisai and Biogen announce detailed results of phase II clinical study of BAN2401 in early Azheimer’s disease at Alzheimer’s Association International Conference (AAIC) 2018. 2018. Eisai, Eisai and Biogen announce detailed results of phase II clinical study of BAN2401 in early Azheimer’s disease at Alzheimer’s Association International Conference (AAIC) 2018. 2018.
11.
go back to reference Ono K. Alzheimer’s disease as oligomeropathy. Neurochem Int. 2017;119:57-70CrossRef Ono K. Alzheimer’s disease as oligomeropathy. Neurochem Int. 2017;119:57-70CrossRef
12.
go back to reference McLean CA, et al. Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer’s disease. Ann Neurol. 1999;46(6):860–6.CrossRef McLean CA, et al. Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer’s disease. Ann Neurol. 1999;46(6):860–6.CrossRef
13.
go back to reference Lue LF, et al. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer’s disease. Am J Pathol. 1999;155(3):853–62.CrossRef Lue LF, et al. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer’s disease. Am J Pathol. 1999;155(3):853–62.CrossRef
14.
go back to reference Esparza TJ, et al. Amyloid-beta oligomerization in Alzheimer dementia versus high-pathology controls. Ann Neurol. 2013;73(1):104–19.CrossRef Esparza TJ, et al. Amyloid-beta oligomerization in Alzheimer dementia versus high-pathology controls. Ann Neurol. 2013;73(1):104–19.CrossRef
15.
go back to reference Shankar GM, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837–42.CrossRef Shankar GM, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14(8):837–42.CrossRef
16.
go back to reference Jin M, et al. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A. 2011;108(14):5819–24.CrossRef Jin M, et al. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce tau hyperphosphorylation and neuritic degeneration. Proc Natl Acad Sci U S A. 2011;108(14):5819–24.CrossRef
17.
go back to reference Sehlin D, et al. Large aggregates are the major soluble Abeta species in AD brain fractionated with density gradient ultracentrifugation. PLoS One. 2012;7(2):e32014.CrossRef Sehlin D, et al. Large aggregates are the major soluble Abeta species in AD brain fractionated with density gradient ultracentrifugation. PLoS One. 2012;7(2):e32014.CrossRef
18.
go back to reference Borlikova GG, et al. Alzheimer brain-derived amyloid beta-protein impairs synaptic remodeling and memory consolidation. Neurobiol Aging. 2013;34(5):1315–27.CrossRef Borlikova GG, et al. Alzheimer brain-derived amyloid beta-protein impairs synaptic remodeling and memory consolidation. Neurobiol Aging. 2013;34(5):1315–27.CrossRef
19.
go back to reference Esparza TJ, et al. Soluble amyloid-beta aggregates from human Alzheimer’s disease brains. Sci Rep. 2016;6:38187.CrossRef Esparza TJ, et al. Soluble amyloid-beta aggregates from human Alzheimer’s disease brains. Sci Rep. 2016;6:38187.CrossRef
20.
go back to reference Yang T, et al. Large soluble oligomers of amyloid beta-protein from Alzheimer brain are far less neuroactive than the smaller oligomers to which they dissociate. J Neurosci. 2017;37(1):152–63.CrossRef Yang T, et al. Large soluble oligomers of amyloid beta-protein from Alzheimer brain are far less neuroactive than the smaller oligomers to which they dissociate. J Neurosci. 2017;37(1):152–63.CrossRef
21.
go back to reference Brody DL, et al. Non-canonical soluble amyloid-beta aggregates and plaque buffering: controversies and future directions for target discovery in Alzheimer’s disease. Alzheimers Res Ther. 2017;9(1):62.CrossRef Brody DL, et al. Non-canonical soluble amyloid-beta aggregates and plaque buffering: controversies and future directions for target discovery in Alzheimer’s disease. Alzheimers Res Ther. 2017;9(1):62.CrossRef
22.
go back to reference Roberts BR, et al. Biochemically-defined pools of amyloid-beta in sporadic Alzheimer’s disease: correlation with amyloid PET. Brain. 2017;140(5):1486–98.CrossRef Roberts BR, et al. Biochemically-defined pools of amyloid-beta in sporadic Alzheimer’s disease: correlation with amyloid PET. Brain. 2017;140(5):1486–98.CrossRef
23.
go back to reference Savage MJ, et al. A sensitive abeta oligomer assay discriminates Alzheimer’s and aged control cerebrospinal fluid. J Neurosci. 2014;34(8):2884–97.CrossRef Savage MJ, et al. A sensitive abeta oligomer assay discriminates Alzheimer’s and aged control cerebrospinal fluid. J Neurosci. 2014;34(8):2884–97.CrossRef
24.
go back to reference Yang T, et al. A highly sensitive novel immunoassay specifically detects low levels of soluble Abeta oligomers in human cerebrospinal fluid. Alzheimers Res Ther. 2015;7(1):14.CrossRef Yang T, et al. A highly sensitive novel immunoassay specifically detects low levels of soluble Abeta oligomers in human cerebrospinal fluid. Alzheimers Res Ther. 2015;7(1):14.CrossRef
25.
go back to reference Schuster J, Funke SA. Methods for the specific detection and quantitation of amyloid-beta oligomers in cerebrospinal fluid. J Alzheimers Dis. 2016;53(1):53–67.CrossRef Schuster J, Funke SA. Methods for the specific detection and quantitation of amyloid-beta oligomers in cerebrospinal fluid. J Alzheimers Dis. 2016;53(1):53–67.CrossRef
26.
go back to reference Mo JJ, et al. Efficacy and safety of anti-amyloid-beta immunotherapy for Alzheimer’s disease: a systematic review and network meta-analysis. Ann Clin Transl Neurol. 2017;4(12):931–42.CrossRef Mo JJ, et al. Efficacy and safety of anti-amyloid-beta immunotherapy for Alzheimer’s disease: a systematic review and network meta-analysis. Ann Clin Transl Neurol. 2017;4(12):931–42.CrossRef
27.
go back to reference Cummings JL, et al. ABBY: A phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology. 2018;90(21):e1889–97.CrossRef Cummings JL, et al. ABBY: A phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology. 2018;90(21):e1889–97.CrossRef
28.
go back to reference Golde TE, Das P, Levites Y. Quantitative and mechanistic studies of Abeta immunotherapy. CNS Neurol Disord Drug Targets. 2009;8(1):31–49.CrossRef Golde TE, Das P, Levites Y. Quantitative and mechanistic studies of Abeta immunotherapy. CNS Neurol Disord Drug Targets. 2009;8(1):31–49.CrossRef
29.
go back to reference Schupf N, et al. Peripheral Abeta subspecies as risk biomarkers of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2008;105(37):14052–7.CrossRef Schupf N, et al. Peripheral Abeta subspecies as risk biomarkers of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2008;105(37):14052–7.CrossRef
30.
go back to reference Reddy MP, et al. Elimination of Fc receptor-dependent effector functions of a modified IgG4 monoclonal antibody to human CD4. J Immunol. 2000;164(4):1925–33.CrossRef Reddy MP, et al. Elimination of Fc receptor-dependent effector functions of a modified IgG4 monoclonal antibody to human CD4. J Immunol. 2000;164(4):1925–33.CrossRef
31.
go back to reference Bard F, 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. 2000;6(8):916–9.CrossRef Bard F, 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. 2000;6(8):916–9.CrossRef
32.
go back to reference Johansson AS, et al. Physiochemical characterization of the Alzheimer’s disease-related peptides A beta 1-42Arctic and A beta 1-42wt. FEBS J. 2006;273(12):2618–30.CrossRef Johansson AS, et al. Physiochemical characterization of the Alzheimer’s disease-related peptides A beta 1-42Arctic and A beta 1-42wt. FEBS J. 2006;273(12):2618–30.CrossRef
33.
go back to reference Stine WB Jr, et al. In vitro characterization of conditions for amyloid-beta peptide oligomerization and fibrillogenesis. J Biol Chem. 2003;278(13):11612–22.CrossRef Stine WB Jr, et al. In vitro characterization of conditions for amyloid-beta peptide oligomerization and fibrillogenesis. J Biol Chem. 2003;278(13):11612–22.CrossRef
34.
go back to reference Blanchard V, et al. Time sequence of maturation of dystrophic neurites associated with Abeta deposits in APP/PS1 transgenic mice. Exp Neurol. 2003;184(1):247–63.CrossRef Blanchard V, et al. Time sequence of maturation of dystrophic neurites associated with Abeta deposits in APP/PS1 transgenic mice. Exp Neurol. 2003;184(1):247–63.CrossRef
35.
go back to reference Dewachter I, et al. Aging increased amyloid peptide and caused amyloid plaques in brain of old APP/V717I transgenic mice by a different mechanism than mutant presenilin1. J Neurosci. 2000;20(17):6452–8.CrossRef Dewachter I, et al. Aging increased amyloid peptide and caused amyloid plaques in brain of old APP/V717I transgenic mice by a different mechanism than mutant presenilin1. J Neurosci. 2000;20(17):6452–8.CrossRef
36.
go back to reference Holtmaat A, et al. Long-term, high-resolution imaging in the mouse neocortex through a chronic cranial window. Nat Protoc. 2009;4(8):1128–44.CrossRef Holtmaat A, et al. Long-term, high-resolution imaging in the mouse neocortex through a chronic cranial window. Nat Protoc. 2009;4(8):1128–44.CrossRef
37.
go back to reference Burgold S, et al. In vivo multiphoton imaging reveals gradual growth of newborn amyloid plaques over weeks. Acta Neuropathol. 2011;121(3):327–35.CrossRef Burgold S, et al. In vivo multiphoton imaging reveals gradual growth of newborn amyloid plaques over weeks. Acta Neuropathol. 2011;121(3):327–35.CrossRef
38.
go back to reference Wilde DB, et al. Evidence implicating L3T4 in class II MHC antigen reactivity; monoclonal antibody GK1.5 (anti-L3T4a) blocks class II MHC antigen-specific proliferation, release of lymphokines, and binding by cloned murine helper T lymphocyte lines. J Immunol. 1983;131(5):2178–83.PubMed Wilde DB, et al. Evidence implicating L3T4 in class II MHC antigen reactivity; monoclonal antibody GK1.5 (anti-L3T4a) blocks class II MHC antigen-specific proliferation, release of lymphokines, and binding by cloned murine helper T lymphocyte lines. J Immunol. 1983;131(5):2178–83.PubMed
39.
go back to reference Alters SE, et al. Mechanisms of anti-CD4-mediated depletion and immunotherapy. A study using a set of chimeric anti-CD4 antibodies. J Immunol. 1990;144(12):4587–92.PubMed Alters SE, et al. Mechanisms of anti-CD4-mediated depletion and immunotherapy. A study using a set of chimeric anti-CD4 antibodies. J Immunol. 1990;144(12):4587–92.PubMed
40.
go back to reference Racke MM, 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 beta. J Neurosci. 2005;25(3):629–36.CrossRef Racke MM, 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 beta. J Neurosci. 2005;25(3):629–36.CrossRef
41.
go back to reference Ghiso J, et al. Systemic catabolism of Alzheimer’s Abeta40 and Abeta42. J Biol Chem. 2004;279(44):45897–908.CrossRef Ghiso J, et al. Systemic catabolism of Alzheimer’s Abeta40 and Abeta42. J Biol Chem. 2004;279(44):45897–908.CrossRef
42.
go back to reference Kastanenka KV, et al. Immunotherapy with aducanumab restores calcium homeostasis in Tg2576 mice. J Neurosci. 2016;36(50):12549–58.CrossRef Kastanenka KV, et al. Immunotherapy with aducanumab restores calcium homeostasis in Tg2576 mice. J Neurosci. 2016;36(50):12549–58.CrossRef
43.
go back to reference Beckmann N, et al. Noninvasive magnetic resonance imaging detection of cerebral amyloid angiopathy-related microvascular alterations using superparamagnetic iron oxide particles in APP transgenic mouse models of Alzheimer’s disease: application to passive Abeta immunotherapy. J Neurosci. 2011;31(3):1023–31.CrossRef Beckmann N, et al. Noninvasive magnetic resonance imaging detection of cerebral amyloid angiopathy-related microvascular alterations using superparamagnetic iron oxide particles in APP transgenic mouse models of Alzheimer’s disease: application to passive Abeta immunotherapy. J Neurosci. 2011;31(3):1023–31.CrossRef
44.
go back to reference Fuller JP, et al. Comparing the efficacy and neuroinflammatory potential of three anti-abeta antibodies. Acta Neuropathol. 2015;130(5):699–711.CrossRef Fuller JP, et al. Comparing the efficacy and neuroinflammatory potential of three anti-abeta antibodies. Acta Neuropathol. 2015;130(5):699–711.CrossRef
45.
go back to reference Ryman JT, Meibohm B. Pharmacokinetics of monoclonal antibodies. CPT Pharmacometrics Syst Pharmacol. 2017;6(9):576–88.CrossRef Ryman JT, Meibohm B. Pharmacokinetics of monoclonal antibodies. CPT Pharmacometrics Syst Pharmacol. 2017;6(9):576–88.CrossRef
46.
go back to reference Blennow K, et al. Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Arch Neurol. 2012;69(8):1002–10.CrossRef Blennow K, et al. Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Arch Neurol. 2012;69(8):1002–10.CrossRef
47.
go back to reference Gertz MA, Landau HJ, Weiss BM. Organ response in patients with AL amyloidosis treated with NEOD001, an amyloid-directed monoclonal antibody. Am J Hematol. 2016;91(12):E506–8.CrossRef Gertz MA, Landau HJ, Weiss BM. Organ response in patients with AL amyloidosis treated with NEOD001, an amyloid-directed monoclonal antibody. Am J Hematol. 2016;91(12):E506–8.CrossRef
48.
go back to reference AlzForumNews, High-dose gantenerumab lowers plaque load. 2017. AlzForumNews, High-dose gantenerumab lowers plaque load. 2017.
49.
go back to reference Sperling RA, et al. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimers Dement. 2011;7(4):367–85.CrossRef Sperling RA, et al. Amyloid-related imaging abnormalities in amyloid-modifying therapeutic trials: recommendations from the Alzheimer’s Association Research Roundtable Workgroup. Alzheimers Dement. 2011;7(4):367–85.CrossRef
50.
go back to reference Bacskai BJ, et al. Non-Fc-mediated mechanisms are involved in clearance of amyloid-beta in vivo by immunotherapy. J Neurosci. 2002;22(18):7873–8.CrossRef Bacskai BJ, et al. Non-Fc-mediated mechanisms are involved in clearance of amyloid-beta in vivo by immunotherapy. J Neurosci. 2002;22(18):7873–8.CrossRef
51.
go back to reference Tamura Y, et al. The F(ab)’2 fragment of an Abeta-specific monoclonal antibody reduces Abeta deposits in the brain. Neurobiol Dis. 2005;20(2):541–9.CrossRef Tamura Y, et al. The F(ab)’2 fragment of an Abeta-specific monoclonal antibody reduces Abeta deposits in the brain. Neurobiol Dis. 2005;20(2):541–9.CrossRef
52.
go back to reference Levites Y, et al. Intracranial adeno-associated virus-mediated delivery of anti-pan amyloid beta, amyloid beta40, and amyloid beta42 single-chain variable fragments attenuates plaque pathology in amyloid precursor protein mice. J Neurosci. 2006;26(46):11923–8.CrossRef Levites Y, et al. Intracranial adeno-associated virus-mediated delivery of anti-pan amyloid beta, amyloid beta40, and amyloid beta42 single-chain variable fragments attenuates plaque pathology in amyloid precursor protein mice. J Neurosci. 2006;26(46):11923–8.CrossRef
53.
go back to reference Das P, et al. Amyloid-beta immunization effectively reduces amyloid deposition in FcRgamma−/− knock-out mice. J Neurosci. 2003;23(24):8532–8.CrossRef Das P, et al. Amyloid-beta immunization effectively reduces amyloid deposition in FcRgamma−/− knock-out mice. J Neurosci. 2003;23(24):8532–8.CrossRef
54.
go back to reference Wilcock DM, et al. Deglycosylated anti-amyloid-beta antibodies eliminate cognitive deficits and reduce parenchymal amyloid with minimal vascular consequences in aged amyloid precursor protein transgenic mice. J Neurosci. 2006;26(20):5340–6.CrossRef Wilcock DM, et al. Deglycosylated anti-amyloid-beta antibodies eliminate cognitive deficits and reduce parenchymal amyloid with minimal vascular consequences in aged amyloid precursor protein transgenic mice. J Neurosci. 2006;26(20):5340–6.CrossRef
55.
go back to reference Adolfsson O, et al. An effector-reduced anti-beta-amyloid (Abeta) antibody with unique abeta binding properties promotes neuroprotection and glial engulfment of Abeta. J Neurosci. 2012;32(28):9677–89.CrossRef Adolfsson O, et al. An effector-reduced anti-beta-amyloid (Abeta) antibody with unique abeta binding properties promotes neuroprotection and glial engulfment of Abeta. J Neurosci. 2012;32(28):9677–89.CrossRef
56.
go back to reference Delnomdedieu M, et al. First-In-Human safety and long-term exposure data for AAB-003 (PF-05236812) and biomarkers after intravenous infusions of escalating doses in patients with mild to moderate Alzheimer’s disease. Alzheimers Res Ther. 2016;8(1):12.CrossRef Delnomdedieu M, et al. First-In-Human safety and long-term exposure data for AAB-003 (PF-05236812) and biomarkers after intravenous infusions of escalating doses in patients with mild to moderate Alzheimer’s disease. Alzheimers Res Ther. 2016;8(1):12.CrossRef
57.
go back to reference Yirmiya R, Goshen I. Immune modulation of learning, memory, neural plasticity and neurogenesis. Brain Behav Immun. 2011;25(2):181–213.CrossRef Yirmiya R, Goshen I. Immune modulation of learning, memory, neural plasticity and neurogenesis. Brain Behav Immun. 2011;25(2):181–213.CrossRef
58.
go back to reference Lee SH, et al. Antibody-mediated targeting of tau in vivo does not require effector function and microglial engagement. Cell Rep. 2016;16(6):1690–700.CrossRef Lee SH, et al. Antibody-mediated targeting of tau in vivo does not require effector function and microglial engagement. Cell Rep. 2016;16(6):1690–700.CrossRef
59.
go back to reference Cohen SI, et al. Proliferation of amyloid-beta42 aggregates occurs through a secondary nucleation mechanism. Proc Natl Acad Sci U S A. 2013;110(24):9758–63.CrossRef Cohen SI, et al. Proliferation of amyloid-beta42 aggregates occurs through a secondary nucleation mechanism. Proc Natl Acad Sci U S A. 2013;110(24):9758–63.CrossRef
60.
go back to reference Condello C, et al. Microglia constitute a barrier that prevents neurotoxic protofibrillar Abeta42 hotspots around plaques. Nat Commun. 2015;6:6176.CrossRef Condello C, et al. Microglia constitute a barrier that prevents neurotoxic protofibrillar Abeta42 hotspots around plaques. Nat Commun. 2015;6:6176.CrossRef
61.
go back to reference Wang A, et al. Robust amyloid clearance in a mouse model of Alzheimer’s disease provides novel insights into the mechanism of amyloid-beta immunotherapy. J Neurosci. 2011;31(11):4124–36.CrossRef Wang A, et al. Robust amyloid clearance in a mouse model of Alzheimer’s disease provides novel insights into the mechanism of amyloid-beta immunotherapy. J Neurosci. 2011;31(11):4124–36.CrossRef
62.
go back to reference Jacobsen H, et al. Combined treatment with a BACE inhibitor and anti-Abeta antibody gantenerumab enhances amyloid reduction in APPLondon mice. J Neurosci. 2014;34(35):11621–30.CrossRef Jacobsen H, et al. Combined treatment with a BACE inhibitor and anti-Abeta antibody gantenerumab enhances amyloid reduction in APPLondon mice. J Neurosci. 2014;34(35):11621–30.CrossRef
Metadata
Title
SAR228810: an antibody for protofibrillar amyloid β peptide designed to reduce the risk of amyloid-related imaging abnormalities (ARIA)
Authors
Laurent Pradier
Véronique Blanchard-Brégeon
Andrees Bohme
Thomas Debeir
Jean Menager
Patrick Benoit
Pascal Barneoud
Véronique Taupin
Philippe Bertrand
Philippe Dugay
Béatrice Cameron
Yi Shi
Souad Naimi
Marc Duchesne
Marie Gagnaire
Tim Weeden
Tara Travaline
David Reczek
Leonard Khiroug
Mohamed Slaoui
Pascale Brunel
Hidehiro Fukuyama
Jeffrey Ravetch
Thierry Canton
Caroline Cohen
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Alzheimer's Research & Therapy / Issue 1/2018
Electronic ISSN: 1758-9193
DOI
https://doi.org/10.1186/s13195-018-0447-y

Other articles of this Issue 1/2018

Alzheimer's Research & Therapy 1/2018 Go to the issue