ArticlePublisher preview availableLiterature Review

Vascular Considerations for Amyloid Immunotherapy

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Purpose of Review Amyloid beta (Aβ) plaque accumulation is a hallmark pathology contributing to Alzheimer’s disease (AD) and is widely hypothesized to lead to cognitive decline. Decades of research into anti-Aβ immunotherapies provide evidence for increased Aβ clearance from the brain; however, this is frequently accompanied by complicated vascular deficits. This article reviews the history of anti-Aβ immunotherapies and clinical findings and provides recommendations moving forward. Recent Findings In 20 years of both animal and human studies, anti-Aβ immunotherapies have been a prevalent avenue of reducing hallmark Aβ plaques. In both models and with different anti-Aβ antibody designs, amyloid-related imaging abnormalities (ARIA) indicating severe cerebrovascular compromise have been common and concerning occurrence. Summary ARIA caused by anti-Aβ immunotherapy has been noted since the early 2000s, and the mechanisms driving it are still unknown. Recent approval of aducanumab comes with renewed urgency to consider vascular deficits caused by anti-Aβ immunotherapy.
This content is subject to copyright. Terms and conditions apply.
Current Neurology and Neuroscience Reports (2022) 22:709–719
Vol.:(0123456789)
1 3
https://doi.org/10.1007/s11910-022-01235-1
DEMENTIA (K.S. MARDER, SECTION EDITOR)
Vascular Considerations forAmyloid Immunotherapy
KateE.Foley1 · DonnaM.Wilcock1
Accepted: 18 September 2022
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2022
Abstract
Purpose of Review Amyloid beta (Aβ) plaque accumulation is a hallmark pathology contributing to Alzheimer’s dis-
ease (AD) and is widely hypothesized to lead to cognitive decline. Decades of research into anti-Aβ immunotherapies
provide evidence for increased Aβ clearance from the brain; however, this is frequently accompanied by complicated
vascular deficits. This article reviews the history of anti-Aβ immunotherapies and clinical findings and provides rec-
ommendations moving forward.
Recent Findings In 20years of both animal and human studies, anti-Aβ immunotherapies have been a prevalent
avenue of reducing hallmark Aβ plaques. In both models and with different anti-Aβ antibody designs, amyloid-related
imaging abnormalities (ARIA) indicating severe cerebrovascular compromise have been common and concerning
occurrence.
Summary ARIA caused by anti-Aβ immunotherapy has been noted since the early 2000s, and the mechanisms driving it are
still unknown. Recent approval of aducanumab comes with renewed urgency to consider vascular deficits caused by anti-Aβ
immunotherapy.
Keywords Vascular· Alzheimer’s disease· Aducanumab· Amyloid· Immunotherapy· Biomarkers
Introduction
The accelerated approval of aducanumab under the FDA in
June 2021 as an anti-Aβ immunotherapy for Alzheimer’s
disease (AD) has reinvigorated the field to understand if
a reduction in amyloid burden in the brain results in cog-
nitive changes in humans and, further, what causes the
detrimental side effects known as amyloid-related imag-
ing abnormalities (ARIA). Decades of anti-Aβ immuno-
therapy research have resulted in a clear dichotomy of
researchers and clinicians that support or oppose the use
of these drugs. This review will detail both the findings
that paved the way for anti-Aβ immunotherapies in trials
today and evaluate key considerations moving forward.
Preclinical History ofAβ‑Immunotherapy
Research
Nuances andCharacterization oftheAβ Peptide
Methods to remove Aβ from the brain have been investigated
for over 20years. While amyloid plaques were identified and
described by Dr. Alois Alzheimer in 1906, it was the association
of the mutation in amyloid precursor protein, APP, which relayed
an autosomal dominant mode of AD inheritance that led to the
later “amyloid cascade hypothesis” [14]. Experiments starting
in the mid-1990s began to parse out the characteristics of the Aβ
peptide that led to it being the hallmark pathology in Alzheimer’s
disease. Early studies revealed that N-terminal deletions resulted
in the formation of Aβ into β-pleated sheets and showed increased
aggregation [5]. Additionally, Iwatsubo etal. identified the Aβ
peptide’s solubility as a key determinant in pathological toxicity,
highlighting that the C-terminus of Aβ is hydrophobic, and longer
C-termini (Aβ42) results in more, and earlier Aβ deposition in
the brain versus shorter isoforms [6]. Today we understand this
to be the insoluble Aβ1-42 peptide which is the primary form of
deposited plaques, while Aβ1-40 is found around the vasculature in
cerebral amyloid angiopathy (CAA) pathology [7, 8].
This article is part of the Topical Collection on Dementia
* Donna M. Wilcock
donna.wilcock@uky.edu
1 Sanders-Brown Center On Aging, Department
ofPhysiology, University ofKentucky, Lexington, KY, USA
/ Published online: 21 October 2022
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... However, some patients who experienced more severe symptoms have been withdrawn from the clinical trials. There is a higher risk of developing this side effect for carriers of APOE-ε4 [53] and having two copies of this gene virtually guarantees the development of Alzheimer's at an earlier age [54,55]. ...
Article
Full-text available
Antibodies that can selectively remove rogue proteins in the brain are an obvious choice to treat neurodegenerative disorders (NDs), but after decades of efforts, only two antibodies to treat Alzheimer’s disease are approved, dozens are in the testing phase, and one was withdrawn, and the other halted, likely due to efficacy issues. However, these outcomes should have been evident since these antibodies cannot enter the brain sufficiently due to the blood–brain barrier (BBB) protectant. However, all products can be rejuvenated by binding them with transferrin, preferably as smaller fragments. This model can be tested quickly and at a low cost and should be applied to bapineuzumab, solanezumab, crenezumab, gantenerumab, aducanumab, lecanemab, donanemab, cinpanemab, and gantenerumab, and their fragments. This paper demonstrates that conjugating with transferrin does not alter the binding to brain proteins such as amyloid-β (Aβ) and α-synuclein. We also present a selection of conjugate designs that will allow cleavage upon entering the brain to prevent their exocytosis while keeping the fragments connected to enable optimal binding to proteins. The identified products can be readily tested and returned to patients with the lowest regulatory cost and delays. These engineered antibodies can be manufactured by recombinant engineering, preferably by mRNA technology, as a more affordable solution to meet the dire need to treat neurodegenerative disorders effectively.
... This comparative approach using different types of Aβ species is required, given that modern therapies against AD are directed toward targeting Aβ peptides employing specific antibodies [58], as demonstrated by the recent US Food and Drug Administration approval of aducanumab as the first AD-specific treatment [59,60]. Despite the controversy regarding its efficacy, clinical trials reveal that the main undesired effects of this approach are related to enhanced vascular damage [61][62][63]. Particularly, evidence accumulated from murine models and clinical trials suggests that passive immunization with antibodies targeting fibrillar Aβ42 aggravates cerebral amyloid angiopathy in some patients, exacerbating vascular damage and brain edema [64][65][66]. This suggests that amyloid angiopathy should not be considered an event occurring independently of vascular alterations and that the interaction between both should be elucidated by basic studies to support future clinical studies. ...
Article
Full-text available
Alzheimer’s disease (AD) is characterized by the accumulation of aggregated amyloid peptides in the brain parenchyma and within the walls of cerebral vessels. The hippocampus—a complex brain structure with a pivotal role in learning and memory—is implicated in this disease. However, there is limited data on vascular changes during AD pathological degeneration in this susceptible structure, which has distinctive vascular traits. Our aim was to evaluate vascular alterations in the hippocampus of AD patients and PDAPP-J20 mice—a model of AD—and to determine the impact of Aβ40 and Aβ42 on endothelial cell activation. We found a loss of physical astrocyte-endothelium interaction in the hippocampus of individuals with AD as compared to non-AD donors, along with reduced vascular density. Astrocyte-endothelial interactions and levels of the tight junction protein occludin were altered early in PDAPP-J20 mice, preceding any signs of morphological changes or disruption of the blood–brain barrier in these mice. At later stages, PDAPP-J20 mice exhibited decreased vascular density in the hippocampus and leakage of fluorescent tracers, indicating dysfunction of the vasculature and the BBB. In vitro studies showed that soluble Aβ40 exposure in human brain microvascular endothelial cells (HBMEC) was sufficient to induce NFκB translocation to the nucleus, which may be linked with an observed reduction in occludin levels. The inhibition of the membrane receptor for advanced glycation end products (RAGE) prevented these changes in HBMEC. Additional results suggest that Aβ42 indirectly affects the endothelium by inducing astrocytic factors. Furthermore, our results from human and mouse brain samples provide evidence for the crucial involvement of the hippocampal vasculature in Alzheimer’s disease.
Article
Full-text available
INTRODUCTION In September 2022, The Jackson Laboratory Center for Alzheimer's and Dementia Research (JAX CADR) hosted a workshop with leading researchers in the Alzheimer's disease and related dementias (ADRD) field. METHODS During the workshop, the participants brainstormed new directions to overcome current barriers to providing patients with effective ADRD therapeutics. The participants outlined specific areas of focus. Following the workshop, each group used standard literature search methods to provide background for each topic. RESULTS The team of invited experts identified four key areas that can be collectively addressed to make a significant impact in the field: (1) Prioritize the diversification of disease targets, (2) enhance factors promoting resilience, (3) de‐risk clinical pipeline, and (4) centralize data management. DISCUSSION In this report, we review these four objectives and propose innovations to expedite ADRD therapeutic pipelines.
Preprint
Full-text available
Alzheimer's disease (AD) is characterized by the accumulation of aggregated amyloid peptides in the brain parenchyma and also around vasculature. The hippocampus -a complex brain structure with a crucial role in learning and memory- is considered a target in the pathology. However, there is scarce information regarding vascular changes during the AD neurodegenerative process in this vulnerable structure, that is a unique in terms of vasculature features. Our aim was to evaluate hippocampal vascular alterations in AD patients and PDAPP-J20 mice -model of AD- and define the impact of Aβ40 and Aβ42 on endothelial activation. We found loss of physical astrocyte-endothelium interaction in the hippocampus of AD subjects as compared to non-AD donors together with decreased vascular density. Astrocyte-endothelial interactions and levels of the tight junction protein occludin were early altered in PDAPP-J20 mice, before vascular morphological changes or blood-brain barrier disruption were evident. At later stages, PDAPP-J20 mice showed decreased hippocampal vascular density and extravasation of fluorescent tracers, indicating vascular and BBB dysfunction. In vitro studies showed that exposure of human brain microvascular endothelial cells (HBMEC) to soluble Aβ40 was sufficient to promote NFκB translocation to the nucleus, leading to a reduction in occludin levels. These changes were prevented by treatment of HBMEC with an inhibitor of the membrane receptor for advanced glycation endproducts (RAGE). Additional results suggest that Aβ42 acts indirectly on the endothelium by inducing astrocytic factors. Furthermore, our results from human and mouse brain samples provide evidence for the crucial involvement of the hippocampal vasculature during Alzheimer's disease.
Article
Full-text available
Aducanumab (Aduhelm) is approved in the United States for the treatment of patients with mild cognitive impairment due to Alzheimer's disease or mild AD dementia. Aducanumab Appropriate Use Recommendations (AURs) have been published and have helped guide best practices for use of aducanumab. As real-world use has occurred and more information has accrued, the AURs require refinement. We update the AURs to better inform appropriate patient selection and improve shared decision-making, safety monitoring, and risk mitigation in treated patients. Based on evolving experience we emphasize the importance of detecting past medical conditions that may predispose to amyloid related imaging abnormalities (ARIA) or may increase the likelihood of ARIA complications including autoimmune or inflammatory conditions, seizures, or disorders associated with extensive white matter pathology. The apolipoprotein E ε4 (APOE4) genotype is strongly associated with ARIA and exhibits a gene dose effect. We recommend that clinicians perform APOE genotyping to better inform patient care decisions, discussions regarding risk, and clinician vigilance concerning ARIA. As most ARIA occurs during the titration period of aducanumab, we suggest performing MRI before the 5th, 7th, 9th, and 12th infusions to improve detection. Uncommonly, ARIA may be recurrent or serious; we suggest additional parameters for treatment discontinuation taking these observations into account. It is important to continue to learn from the real-world use of aducanumab and the AURs will continue to evolve as new information becomes available. This AUR update does not address efficacy, price, or insurance coverage and is provided to assist clinicians to establish best practices for use of aducanumab in the treatment of patients with mild cognitive impairment and mild Alzheimer's dementia.
Article
Full-text available
Alzheimer’s disease (AD) is the most common cause for dementia worldwide. Until recently, all approved treatments for AD were symptomatic and not disease modifying. On 7 June 2021, the US FDA approved aducanumab, a human IgG1 anti-Aβ monoclonal antibody selective for Aβ aggregates, as the first disease-modifying treatment for AD. Aducanumab is approved in the United States for the treatment of mild cognitive impairment or mild-dementia stage of AD. In this Editorial, we review the trial data for aducanumab in the treatment of AD and the controversies that its approval has generated.
Article
Full-text available
Aducanumab has been approved by the US Food and Drug Administration for treatment of Alzheimer's disease (AD). Clinicians require guidance on the appropriate use of this new therapy. An Expert Panel was assembled to construct Appropriate Use Recommendations based on the participant populations, conduct of the pivotal trials of aducanumab, updated Prescribing Information, and expert consensus. Aducanumab is an amyloid-targeting monoclonal antibody delivered by monthly intravenous infusions. The pivotal trials included patients with early AD (mild cognitive impairment due to AD and mild AD dementia) who had confirmed brain amyloid using amyloid positron tomography. The Expert Panel recommends that use of aducanumab be restricted to this population in which efficacy and safety have been studied. Aducanumab is titrated to a dose of 10 mg/kg over a 6-month period. The Expert Panel recommends that the aducanumab be titrated to the highest dose to maximize the opportunity for efficacy. Aducanumab can substantially increase the incidence of amyloid-related imaging abnormalities (ARIA) with brain effusion or hemorrhage. Dose interruption or treatment discontinuation is recommended for symptomatic ARIA and for moderate-severe ARIA. The Expert Panel recommends MRIs prior to initiating therapy, during the titration of the drug, and at any time the patient has symptoms suggestive of ARIA. Recommendations are made for measures less cumbersome than those used in trials for the assessment of effectiveness in the practice setting. The Expert Panel emphasized the critical importance of engaging in a process of patient-centered informed decision-making that includes comprehensive discussions and clear communication with the patient and care partner regarding the requirements for therapy, the expected outcome of therapy, potential risks and side effects, and the required safety monitoring, as well as uncertainties regarding individual responses and benefits.
Article
Full-text available
Aducanumab recently underwent two large phase III clinical trials that were stopped prematurely by the sponsor Biogen. One trial was trending positive while the other showed no benefits from aducanumab. Post hoc analyses led the sponsor to assert that there was a sufficient efficacy signal to justify a new drug application as a treatment for Alzheimer's disease. The sponsor claimed that subsets of participants receiving sufficiently high doses of aducanumab demonstrated benefits in both trials. In contrast, we identified alternative accounts for the apparent drug benefits in post hoc subgroups that are unrelated to dose effects. Biomarker data were consistent with target engagement, but no evidence was presented to correlate biomarker changes to cognitive benefits. Our analysis supports the conduct of a third, phase III trial with high‐dose aducanumab. Aducanumab's efficacy as a treatment for the cognitive dysfunction in Alzheimer's disease cannot be proven by clinical trials with divergent outcomes.
Article
Full-text available
Activation of the humoral immune system is initiated when antibodies recognize an antigen and trigger effector functions through the interaction with Fc engaging molecules. The most abundant immunoglobulin isotype in serum is Immunoglobulin G (IgG), which is involved in many humoral immune responses, strongly interacting with effector molecules. The IgG subclass, allotype, and glycosylation pattern, among other factors, determine the interaction strength of the IgG-Fc domain with these Fc engaging molecules, and thereby the potential strength of their effector potential. The molecules responsible for the effector phase include the classical IgG-Fc receptors (FcγR), the neonatal Fc-receptor (FcRn), the Tripartite motif-containing protein 21 (TRIM21), the first component of the classical complement cascade (C1), and possibly, the Fc-receptor-like receptors (FcRL4/5). Here we provide an overview of the interactions of IgG with effector molecules and discuss how natural variation on the antibody and effector molecule side shapes the biological activities of antibodies. The increasing knowledge on the Fc-mediated effector functions of antibodies drives the development of better therapeutic antibodies for cancer immunotherapy or treatment of autoimmune diseases.
Article
Full-text available
Background Our objective was to evaluate the efficacy (clinical and biomarker) and safety of intravenous bapineuzumab in patients with mild to moderate Alzheimer’s disease (AD). Methods Two of four phase 3, multicenter, randomized, double-blind, placebo-controlled, 18-month trials were conducted globally: one in apolipoprotein E ε4 carriers and another in noncarriers. Patients received bapineuzumab 0.5 mg/kg (both trials) or 1.0 mg/kg (noncarrier trial) or placebo every 13 weeks. Coprimary endpoints were change from baseline to week 78 on the 11-item Alzheimer’s Disease Assessment Scale–Cognitive subscale and the Disability Assessment for Dementia. ResultsA total of 683 and 329 patients completed the current carrier and noncarrier trials, respectively, which were terminated prematurely owing to lack of efficacy in the two other phase 3 trials of bapineuzumab in AD. The current trials showed no significant difference between bapineuzumab and placebo for the coprimary endpoints and no effect of bapineuzumab on amyloid load or cerebrospinal fluid phosphorylated tau. (Both measures were stable over time in the placebo group.) Amyloid-related imaging abnormalities with edema or effusion were confirmed as the most notable adverse event. Conclusions These phase 3 global trials confirmed lack of efficacy of bapineuzumab at tested doses on clinical endpoints in patients with mild to moderate AD. Some differences in the biomarker results were seen compared with the other phase 3 bapineuzumab trials. No unexpected adverse events were observed. Trial registrationNoncarriers (3000) ClinicalTrials.gov identifier NCT00667810; registered 24 Apr 2008.Carriers (3001) ClinicalTrials.gov identifier NCT00676143; registered 2 May 2008.
Article
Full-text available
Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive decline that afflicts as many as 45 % of individuals who survive past the age of 85. AD has been associated with neurovascular dysfunction and brain accumulation of amyloid-β peptide, as well as tau phosphorylation and neurodegeneration, but the pathogenesis of the disease is still somewhat unclear. According to the amyloid cascade hypothesis of AD, accumulation of amyloid-β peptide (Aβ) aggregates initiates a sequence of events leading to neuronal injury and loss, and dementia. Alternatively, the vascular hypothesis of AD incorporates the vascular contribution to the disease, stating that a primary insult to brain microcirculation (e.g., stroke) not only contributes to amyloidopathy but initiates a non-amyloidogenic pathway of vascular-mediated neuronal dysfunction and injury, which involves blood-brain barrier compromise, with increased permeability of blood vessels, leakage of blood-borne components into the brain, and, consequently, neurotoxicity. Vascular dysfunction also includes a diminished brain capillary flow, causing multiple focal ischemic or hypoxic microinjuries, diminished amyloid-β clearance, and formation of neurotoxic oligomers, which lead to neuronal dysfunction. Here we present and discuss relevant findings on the contribution of vascular alterations during aging to AD, with the hope that a better understanding of the players in the "orchestra" of neurodegeneration will be useful in developing therapies to modulate the "symphony".
Article
Background Aducanumab is a human monoclonal antibody that selectively targets aggregated forms of Aβ, including soluble oligomers and insoluble fibrils. EMERGE and ENGAGE are two 18‐month, randomized, double‐blind, placebo‐controlled, global Phase 3 studies with identical design that evaluated the efficacy and safety of aducanumab in patients aged 50–85 years with early Alzheimer’s disease (MCI due to AD or mild AD dementia). Method Key inclusion criteria included positive amyloid PET, MMSE score of 24–30, CDR Global score of 0.5, and an RBANS‐DMI score ≤85. During the 18‐month placebo‐controlled period, patients were randomized 1:1:1 to low‐dose aducanumab, high‐dose aducanumab, or placebo, administered via IV infusion every 4 weeks. The primary endpoint for EMERGE and ENGAGE was change from baseline at Week 78 on the CDR‐SB. Secondary endpoints included change from baseline on MMSE, ADAS‐Cog13, and ADCS‐ADL‐MCI. Result Following pre‐planned futility analysis, analysis of the data from the final database lock showed that EMERGE met its primary endpoint, based on the pre‐specified statistical analysis plan. Patients treated with high dose aducanumab showed a significant reduction of clinical decline from baseline in CDR‐SB scores at 78 weeks (22% versus placebo, P = 0.01). ENGAGE did not meet its primary endpoint. However, data from patients in ENGAGE who achieved sufficient exposure to high dose aducanumab supported the findings of EMERGE. Conclusion EMERGE met its primary endpoint, based on the pre‐specified statistical analysis plan. Data from a subset of patients in ENGAGE support the results of EMERGE. The safety and tolerability profile of aducanumab in EMERGE and ENGAGE was consistent with previous studies of aducanumab.
Article
Alzheimer's disease (AD) is characterized by deposition of amyloid-β (Aβ) plaques and neurofibrillary tangles in the brain, accompanied by synaptic dysfunction and neurodegeneration. Antibody-based immunotherapy against Aβ to trigger its clearance or mitigate its neurotoxicity has so far been unsuccessful. Here we report the generation of aducanumab, a human monoclonal antibody that selectively targets aggregated Aβ. In a transgenic mouse model of AD, aducanumab is shown to enter the brain, bind parenchymal Aβ, and reduce soluble and insoluble Aβ in a dose-dependent manner. In patients with prodromal or mild AD, one year of monthly intravenous infusions of aducanumab reduces brain Aβ in a dose- and time-dependent manner. This is accompanied by a slowing of clinical decline measured by Clinical Dementia Rating - Sum of Boxes and Mini Mental State Examination scores. The main safety and tolerability findings are amyloid-related imaging abnormalities. These results justify further development of aducanumab for the treatment of AD. Should the slowing of clinical decline be confirmed in ongoing phase 3 clinical trials, it would provide compelling support for the amyloid hypothesis.