Skip to main content
Top
Published in: Current Neurology and Neuroscience Reports 8/2021

Open Access 01-08-2021 | Alzheimer's Disease | Dementia (K.S. Marder, Section Editor)

Critical Appraisal of Amyloid Lowering Agents in AD

Authors: Boris Decourt, Fadel Boumelhem, Evans D. Pope III, Jiong Shi, Zoltan Mari, Marwan Noel Sabbagh

Published in: Current Neurology and Neuroscience Reports | Issue 8/2021

Login to get access

Abstract

Purpose of Review

According to the amyloid cascade hypothesis, removing amyloid beta (Aβ) should cure Alzheimer’s disease (AD). In the past three decades, many agents have been tested to try to lower Aβ production, prevent Aβ aggregation, and dissolve Aβ deposits. However, the paucity in definitive preventative or curative properties of these agents in clinical trials has resulted in more avant-garde approaches to therapeutic investigations. Immunotherapy has become an area of focus for research on disease-modifying therapies for neurodegenerative diseases. In this review, we highlight the current clinical development landscape of monoclonal antibody (mAb) therapies that target Aβ plaque formation and removal in AD.

Recent Findings

Multiple potential disease-modifying therapeutics for AD are in active development. Targeting Aβ with mAbs has the potential to treat various stages of AD: prodromal, prodromal to mild, mild, and mild to moderate. Monoclonal antibodies discussed here include aducanumab, lecanemab, solanezumab, crenezumab, donanemab, and gantenerumab.

Summary

The final decision by the FDA regarding the approval of aducanumab will offer valuable insight into the trajectory of drug development for mAbs in AD and other neurodegenerative diseases. Future directions for improving the treatment of AD will include more inquiry into the efficacy of mAbs as disease-modifying agents that specifically target Aβ peptides and/or multimers. In addition, a more robust trial design for AD immunotherapy agents should improve outcomes such that objective measures of clinical efficacy will eventually lead to higher chances of drug approval.
Literature
20.
go back to reference Ikeda S, Allsop D, Glenner GG. Morphology and distribution of plaque and related deposits in the brains of Alzheimer’s disease and control cases. An immunohistochemical study using amyloid β-protein antibody. Lab Investig. 1989;60:113–22.PubMed Ikeda S, Allsop D, Glenner GG. Morphology and distribution of plaque and related deposits in the brains of Alzheimer’s disease and control cases. An immunohistochemical study using amyloid β-protein antibody. Lab Investig. 1989;60:113–22.PubMed
34.
37.
go back to reference Green RC, Schneider LS, Amato DA, Beelen AP, Wilcock G, Swabb EA, et al. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA - J Am Med Assoc. American Medical Association. 2009;302:2557–64. https://doi.org/10.1001/jama.2009.1866.CrossRef Green RC, Schneider LS, Amato DA, Beelen AP, Wilcock G, Swabb EA, et al. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA - J Am Med Assoc. American Medical Association. 2009;302:2557–64. https://​doi.​org/​10.​1001/​jama.​2009.​1866.CrossRef
38.
go back to reference Meyer P-F, Tremblay-Mercier J, Leoutsakos J, Madjar C, Lafaille-Maignan M-É, Savard M, Rosa-Neto P, Poirier J, Etienne P, Breitner J, for the PREVENT-AD research group INTREPAD: a randomized trial of naproxen to slow progress of presymptomatic Alzheimer disease. Neurology. Lippincott Williams and Wilkins; 2019;92:e2070–2080; https://doi.org/10.1212/WNL.0000000000007232 Meyer P-F, Tremblay-Mercier J, Leoutsakos J, Madjar C, Lafaille-Maignan M-É, Savard M, Rosa-Neto P, Poirier J, Etienne P, Breitner J, for the PREVENT-AD research group INTREPAD: a randomized trial of naproxen to slow progress of presymptomatic Alzheimer disease. Neurology. Lippincott Williams and Wilkins; 2019;92:e2070–2080; https://​doi.​org/​10.​1212/​WNL.​0000000000007232​
42.
go back to reference Adlard PA, Bica L, White AR, Nurjono M, Filiz G, Crouch PJ, Donnelly PS, Cappai R, Finkelstein DI, Bush AI Metal ionophore treatment restores dendritic spine density and synaptic protein levels in a mouse model of Alzheimer’s disease. PLoS One PLoS One; 2011;6:e17669; https://doi.org/10.1371/journal.pone.0017669 Adlard PA, Bica L, White AR, Nurjono M, Filiz G, Crouch PJ, Donnelly PS, Cappai R, Finkelstein DI, Bush AI Metal ionophore treatment restores dendritic spine density and synaptic protein levels in a mouse model of Alzheimer’s disease. PLoS One PLoS One; 2011;6:e17669; https://​doi.​org/​10.​1371/​journal.​pone.​0017669
43.
go back to reference Lannfelt L, Blennow K, Zetterberg H, Batsman S, Ames D, Harrison J, et al. Safety, efficacy, and biomarker findings of PBT2 in targeting Aβ as a modifying therapy for Alzheimer’s disease: a phase IIa, double-blind, randomised, placebo-controlled trial. Lancet Neurol Lancet Publishing Group. 2008;7:779–86. https://doi.org/10.1016/S1474-4422(08)70167-4.CrossRefPubMed Lannfelt L, Blennow K, Zetterberg H, Batsman S, Ames D, Harrison J, et al. Safety, efficacy, and biomarker findings of PBT2 in targeting Aβ as a modifying therapy for Alzheimer’s disease: a phase IIa, double-blind, randomised, placebo-controlled trial. Lancet Neurol Lancet Publishing Group. 2008;7:779–86. https://​doi.​org/​10.​1016/​S1474-4422(08)70167-4.CrossRefPubMed
56.
go back to reference Ferrero J, Williams L, Stella H, Leitermann K, Mikulskis A, O’Gorman J, et al. First-in-human, double-blind, placebo-controlled, single-dose escalation study of aducanumab (BIIB037) in mild-to-moderate Alzheimer’s disease. Alzheimer’s Dement Transl Res Clin Interv Elsevier Inc. 2016;2:169–76. https://doi.org/10.1016/j.trci.2016.06.002.CrossRef Ferrero J, Williams L, Stella H, Leitermann K, Mikulskis A, O’Gorman J, et al. First-in-human, double-blind, placebo-controlled, single-dose escalation study of aducanumab (BIIB037) in mild-to-moderate Alzheimer’s disease. Alzheimer’s Dement Transl Res Clin Interv Elsevier Inc. 2016;2:169–76. https://​doi.​org/​10.​1016/​j.​trci.​2016.​06.​002.CrossRef
57.•
go back to reference Haeberlein SB, von Hehn C, Tian Y, Chalkias S, Muralidharan KK, Chen T, et al. Emerge and engage topline results: phase 3 studies of aducanumab in early Alzheimer’s disease. Alzheimers Dement. Wiley; 2020;16; https://doi.org/10.1002/alz.047259.Results from EMERGE and ENGAGE offered conflicting data that has led some researchers to question and oppose the potential FDA approval aducanumab for treatment of AD. Haeberlein SB, von Hehn C, Tian Y, Chalkias S, Muralidharan KK, Chen T, et al. Emerge and engage topline results: phase 3 studies of aducanumab in early Alzheimer’s disease. Alzheimers Dement. Wiley; 2020;16; https://​doi.​org/​10.​1002/​alz.​047259.Results from EMERGE and ENGAGE offered conflicting data that has led some researchers to question and oppose the potential FDA approval aducanumab for treatment of AD.
60.
go back to reference Logovinsky V, Satlin A, Lai R, Swanson C, Kaplow J, Osswald G, et al. Safety and tolerability of BAN2401 - a clinical study in Alzheimer’s disease with a protofibril selective Aβ antibody. Alzheimers Res Ther BioMed Central Ltd.; 2016;8; https://doi.org/10.1186/s13195-016-0181-2 Logovinsky V, Satlin A, Lai R, Swanson C, Kaplow J, Osswald G, et al. Safety and tolerability of BAN2401 - a clinical study in Alzheimer’s disease with a protofibril selective Aβ antibody. Alzheimers Res Ther BioMed Central Ltd.; 2016;8; https://​doi.​org/​10.​1186/​s13195-016-0181-2
61.
62.
go back to reference Söllvander S, Nikitidou E, Gallasch L, Zyśk M, Söderberg L, Sehlin D, et al. The Aβ protofibril selective antibody mAb158 prevents accumulation of Aβ in astrocytes and rescues neurons from Aβ-induced cell death. J Neuroinflammation. BioMed Central Ltd.; 2018;15; https://doi.org/10.1186/s12974-018-1134-4 Söllvander S, Nikitidou E, Gallasch L, Zyśk M, Söderberg L, Sehlin D, et al. The Aβ protofibril selective antibody mAb158 prevents accumulation of Aβ in astrocytes and rescues neurons from Aβ-induced cell death. J Neuroinflammation. BioMed Central Ltd.; 2018;15; https://​doi.​org/​10.​1186/​s12974-018-1134-4
68.••
go back to reference Klein G, Delmar P, Kerchner GA, Hofmann C, Abi-Saab D, Davis A, et al. Thirty-six-month amyloid positron emission tomography results show continued reduction in amyloid burden with subcutaneous gantenerumab. J Prev Alzheimer’s Dis. 2021;8:3–6. https://doi.org/10.14283/jpad.2020.68This study demonstrates potential long-term efficacy of gantenerumab as an amyloid lowering mAb in AD.CrossRef Klein G, Delmar P, Kerchner GA, Hofmann C, Abi-Saab D, Davis A, et al. Thirty-six-month amyloid positron emission tomography results show continued reduction in amyloid burden with subcutaneous gantenerumab. J Prev Alzheimer’s Dis. 2021;8:3–6. https://​doi.​org/​10.​14283/​jpad.​2020.​68This study demonstrates potential long-term efficacy of gantenerumab as an amyloid lowering mAb in AD.CrossRef
69.•
go back to reference Klein G, Delmar P, Voyle N, Rehal S, Hofmann C, Abi-Saab D, et al. Gantenerumab reduces amyloid-β plaques in patients with prodromal to moderate Alzheimer’s disease: a PET substudy interim analysis. Alzheimers Res Ther. 2019;11:101; https://doi.org/10.1186/s13195-019-0559-z. Based on these results, gantenerumab was established as a safe and effective potential AD treatment. Klein G, Delmar P, Voyle N, Rehal S, Hofmann C, Abi-Saab D, et al. Gantenerumab reduces amyloid-β plaques in patients with prodromal to moderate Alzheimer’s disease: a PET substudy interim analysis. Alzheimers Res Ther. 2019;11:101; https://​doi.​org/​10.​1186/​s13195-019-0559-z. Based on these results, gantenerumab was established as a safe and effective potential AD treatment.
75.
go back to reference Salloway S, Honigberg LA, Cho W, Ward M, Friesenhahn M, Brunstein F, 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). Alzheimers Res Ther Alzheimer’s Research & Therapy;. 2018;10:1–13. https://doi.org/10.1186/s13195-018-0424-5.CrossRef Salloway S, Honigberg LA, Cho W, Ward M, Friesenhahn M, Brunstein F, 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). Alzheimers Res Ther Alzheimer’s Research & Therapy;. 2018;10:1–13. https://​doi.​org/​10.​1186/​s13195-018-0424-5.CrossRef
76.
go back to reference Yoshida K, Moein A, Bittner T, Ostrowitzki S, Lin H, Honigberg L, et al. Pharmacokinetics and pharmacodynamic effect of crenezumab on plasma and cerebrospinal fluid beta-amyloid in patients with mild-to-moderate Alzheimer’s disease. Alzheimers Res Ther. 2020;12:1–12. https://doi.org/10.1186/s13195-020-0580-2.CrossRef Yoshida K, Moein A, Bittner T, Ostrowitzki S, Lin H, Honigberg L, et al. Pharmacokinetics and pharmacodynamic effect of crenezumab on plasma and cerebrospinal fluid beta-amyloid in patients with mild-to-moderate Alzheimer’s disease. Alzheimers Res Ther. 2020;12:1–12. https://​doi.​org/​10.​1186/​s13195-020-0580-2.CrossRef
78.
go back to reference Irizarry MC, Sims JR, Lowe SL, Nakano M, Hawdon A, Willis BA, et al. O4-08-06: Safety, pharmacokinetics (PK), and florbetapir F-18 positron emission tomography (PET) after multiple dose administration of LY3002813, A β-amyloid plaque-specific antibody, IN Alzheimer’s disease (AD). Alzheimers Dement Wiley. 2016;12:P352–3. https://doi.org/10.1016/j.jalz.2016.06.665.CrossRef Irizarry MC, Sims JR, Lowe SL, Nakano M, Hawdon A, Willis BA, et al. O4-08-06: Safety, pharmacokinetics (PK), and florbetapir F-18 positron emission tomography (PET) after multiple dose administration of LY3002813, A β-amyloid plaque-specific antibody, IN Alzheimer’s disease (AD). Alzheimers Dement Wiley. 2016;12:P352–3. https://​doi.​org/​10.​1016/​j.​jalz.​2016.​06.​665.CrossRef
79.••
go back to reference Lowe SL, Willis BA, Hawdon A, Natanegara F, Chua L, Foster J, et al. Donanemab (LY3002813) dose-escalation study in Alzheimer’s disease. Alzheimer’s Dement (New York, N Y). 2021\;7; https://doi.org/10.1002/trc2.12112. Donanemab improved iADRS score and reduced both amyloid plaque level and global tau load in participants with early symptomatic AD. Lowe SL, Willis BA, Hawdon A, Natanegara F, Chua L, Foster J, et al. Donanemab (LY3002813) dose-escalation study in Alzheimer’s disease. Alzheimer’s Dement (New York, N Y). 2021\;7; https://​doi.​org/​10.​1002/​trc2.​12112. Donanemab improved iADRS score and reduced both amyloid plaque level and global tau load in participants with early symptomatic AD.
Metadata
Title
Critical Appraisal of Amyloid Lowering Agents in AD
Authors
Boris Decourt
Fadel Boumelhem
Evans D. Pope III
Jiong Shi
Zoltan Mari
Marwan Noel Sabbagh
Publication date
01-08-2021
Publisher
Springer US
Published in
Current Neurology and Neuroscience Reports / Issue 8/2021
Print ISSN: 1528-4042
Electronic ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-021-01125-y

Other articles of this Issue 8/2021

Current Neurology and Neuroscience Reports 8/2021 Go to the issue

Headache (R.B. Halker Singh and J. VanderPluym, Section Editors)

Debunking Myths: Sinus Headache

Headache (R.B. Halker Singh and J. VanderPluym, Section Editors)

Approach to Pediatric Intractable Migraine

Behavior (H.S. Kirshner, Section Editor)

The History of Amnesia—a Review

Neurology of Systemic Diseases (J. Biller, Section Editor)

Neurologic Manifestations of the Antiphospholipid Syndrome — an Update