Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Korean J Radiol ; 25(8): 726-741, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39109501

ABSTRACT

Recent advancements in Alzheimer's disease treatment have focused on the elimination of amyloid-beta (Aß) plaque, a hallmark of the disease. Monoclonal antibodies such as lecanemab and donanemab can alter disease progression by binding to different forms of Aß aggregates. However, these treatments raise concerns about adverse effects, particularly amyloid-related imaging abnormalities (ARIA). Careful assessment of safety, especially regarding ARIA, is crucial. ARIA results from treatment-related disruption of vascular integrity and increased vascular permeability, leading to the leakage of proteinaceous fluid (ARIA-E) and heme products (ARIA-H). ARIA-E indicates treatment-induced edema or sulcal effusion, while ARIA-H indicates treatment-induced microhemorrhage or superficial siderosis. The minimum recommended magnetic resonance imaging sequences for ARIA assessment are T2-FLAIR, T2* gradient echo (GRE), and diffusion-weighted imaging (DWI). T2-FLAIR and T2* GRE are necessary to detect ARIA-E and ARIA-H, respectively. DWI plays a role in differentiating ARIA-E from acute to subacute infarcts. Physicians, including radiologists, must be familiar with the imaging features of ARIA, the appropriate imaging protocol for the ARIA workup, and the reporting of findings in clinical practice. This review aims to describe the clinical and imaging features of ARIA and suggest points for the timely detection and monitoring of ARIA in clinical practice.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Antibodies, Monoclonal , Magnetic Resonance Imaging , Humans , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Magnetic Resonance Imaging/methods
2.
Biomedicines ; 12(8)2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39200334

ABSTRACT

(1) Background: The 2023 approval of lecanemab for early-stage Alzheimer's disease (AD) highlighted the need for routine 1.5T or 3.0T MRI scans to monitor amyloid-related imaging abnormalities (ARIAs). Regional disparities in MRI scan frequency, MRI scanner availability, and scanner magnetic field strengths could affect readiness for anti-amyloid therapy and lead to inconsistencies in ARIA detection nationwide. (2) Methods: We assessed regional variance in MRI scan frequency and field strength across Japan using the National Database (NDB) Open Data website, which summarizes Japanese public health insurance claims from the fiscal years (FYs) 2015 to 2021. We employed a mixed-effects model with prefecture-level random intercepts and slopes over time, subsequently categorizing prefectures into clusters based on MRI usage. (3) Results: 1.5T MRI was the most common magnetic field strength, remaining stable from FY2015 to FY2021. 3.0T MRI usage slightly increased, although the COVID-19 pandemic in FY2020 led to a maximum reduction of 5%. Prefecture-level variance was higher for 3.0T MRIs, with more frequent usage in western Japan. (4) Conclusions: This study highlights prefecture-level variance in MRI usage across Japan. The insights gained could be instrumental in improving healthcare preparedness for anti-amyloid treatment and patient management.

3.
J Neuroimmunol ; 394: 578406, 2024 09 15.
Article in English | MEDLINE | ID: mdl-39094435

ABSTRACT

Cerebral amyloid angiopathy-related inflammation (CAA-ri) is a rapid but reversible autoimmune encephalopathy where spontaneous autoantibody reaction against amyloid beta deposited in cerebral blood vessels produces characteristic neuroinflammatory changes such as vasogenic edema and microhemorrhages on MRI. The term amyloid-related imaging abnormalities (ARIA) is sometimes used to describe these changes but are more often reserved for similar MRI signal abnormalities seen after administration of anti-amyloid immunotherapy, using treatment exposure as an antecedent. It is unclear if there is any biological basis for this dichotomized distinction. We report a case of severe CAA-ri after exposure to SARS-CoV-2 vaccine and performed a literature review of CAA-ri related to vaccination. CAA-ri precipitated by immunogenic triggers other than anti-amyloid therapy would lend support to the hypothesis that ARIA seen on MRI may represent the same disease underpinned by a shared anti-Aß autoantibody response irrespective of etiology. A thorough history should be taken before labelling CAA-ri as spontaneous.


Subject(s)
Cerebral Amyloid Angiopathy , Aged , Humans , Cerebral Amyloid Angiopathy/immunology , Cerebral Amyloid Angiopathy/diagnostic imaging , COVID-19/immunology , COVID-19/complications , COVID-19 Vaccines/adverse effects , Inflammation/immunology , Inflammation/etiology , Inflammation/chemically induced , Magnetic Resonance Imaging , Vaccination/adverse effects
4.
Alzheimers Dement ; 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39115941

ABSTRACT

Phase four of the Alzheimer's Disease Neuroimaging Initiative (ADNI4) magnetic resonance imaging (MRI) protocols aim to maintain longitudinal consistency across two decades of data acquisition, while adopting new technologies. Here we describe and justify the study's design and targeted biomarkers. The ADNI4 MRI protocol includes nine MRI sequences. Some sequences require the latest hardware and software system upgrades and are continuously rolled out as they become available at each site. The main sequence additions/changes in ADNI4 are: (1) compressed sensing (CS) T1-weighting, (2) pseudo-continuous arterial spin labeling (ASL) on all three vendors (GE, Siemens, Philips), (3) multiple-post-labeling-delay ASL, (4) 1 mm3 isotropic 3D fluid-attenuated inversion recovery, and (5) CS 3D T2-weighted. ADNI4 aims to help the neuroimaging community extract valuable imaging biomarkers and provide a database to test the impact of advanced imaging strategies on diagnostic accuracy and disease sensitivity among individuals lying on the cognitively normal to impaired spectrum. HIGHLIGHTS: A summary of MRI protocols for phase four of the Alzheimer's Disease Neuroimaging Initiative (ADNI 4). The design and justification for the ADNI 4 MRI protocols. Compressed sensing and multi-band advances have been applied to improve scan time. ADNI4 protocols aim to streamline safety screening and therapy monitoring. The ADNI4 database will be a valuable test bed for academic research.

5.
Geriatr Gerontol Int ; 2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39034660

ABSTRACT

The introduction of lecanemab has dramatically changed the field of dementia medicine. Lecanemab, defined as an anti-amyloid-ß (Aß) drug, comprises an antibody against Aß, a protein structure believed to cause Alzheimer's disease. This drug represents a new direction in dementia treatment. In a phase III study, lecanemab was found to significantly slow cognitive decline, while showing manageable levels of amyloid-related imaging abnormalities, which are side-effects of lecanemab. Furthermore, lecanemab has been shown to effectively reduce Aß accumulation in patients with early Alzheimer's disease, which might contribute not only to delaying the progression of cognitive decline, but also to improving the quality of life of patients and their families. However, there are conditions for the use of lecanemab, for which the Ministry of Health, Labor and Welfare has issued the Guidelines for Promotion of Optimal Use. These guidelines specify requirements for appropriate patient selection, prescribing physicians and administering medical institutions to ensure safe and effective use. Particular emphasis is placed on the confirmation of amyloid-ß accumulation, amyloid-related imaging abnormalities risk management and appropriate handling of side-effects. The clinical use of lecanemab represents an important advancement in the treatment of dementia; however, the understanding and cooperation of healthcare professionals, patients and families are essential to maximize its efficacy and safety. Future issues to be addressed include the sustainability and long-term efficacy of treatment, improvement of clinical symptoms after removal of Aß and motivation to administer the drug. Although lecanemab offers hope for the treatment of dementia, its use requires careful management. Geriatr Gerontol Int 2024; ••: ••-••.

6.
J Prev Alzheimers Dis ; 11(4): 857-868, 2024.
Article in English | MEDLINE | ID: mdl-39044494

ABSTRACT

BACKGROUND: Preclinical Alzheimer's disease is increasingly studied in clinical trials. Although safety signals are routinely monitored in clinical trial populations with Alzheimer's disease, it can be challenging to identify new safety signals against background rates of age-related medical comorbidities. OBJECTIVES: To report detailed safety data from a cognitively unimpaired older population with evidence of elevated cerebral amyloid levels on amyloid positron emission tomography in the placebo arm of a Phase 3 clinical trial. DESIGN: Phase 3, 4.5-year, multicenter, placebo-controlled trial. SETTING: Placebo data from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease (A4) study. PARTICIPANTS: Enrolled participants were aged 65-85 years with a global Clinical Dementia Rating score of 0, a Mini-Mental State Examination score of 25-30, a Wechsler Memory Scale Logical Memory IIa (delayed recall) score of 6-18, and elevated brain amyloid levels on 18F-florbetapir positron emission tomography. MEASUREMENTS: Study participants who received placebo were followed up with post-baseline safety measures. Assessments included review of concomitant medication and adverse events, the Columbia Suicide Severity Rating Scale, electrocardiograms, and neuroimaging (brain magnetic resonance imaging). RESULTS: In total, 591 study participants (mean age [standard deviation] 71.9 [5.0] years) were assigned to and received placebo in the A4 study, and were followed up to 240 weeks. Participants were primarily White (93.9%) and from the United States (86.8%); 60.4% were women. The most common serious adverse events (incidence rate per 100 person-years) were pneumonia (incidence rate=0.4; 95% confidence interval=0.2-0.7) and atrial fibrillation (incidence rate=0.4; 95% confidence interval=0.2-0.7). The most common treatment-emergent adverse events were upper respiratory tract infection (incidence rate=10.9; 95% confidence interval=9.4-12.5), fall (incidence rate=7.7; 95% confidence interval=6.6-9.0), and nasopharyngitis (incidence rate=5.8; 95% confidence interval=4.8-6.9). The most common ischemia-related findings on magnetic resonance imaging were subcortical infarction (incidence rate=1.4; 95% confidence interval=1.0-2.0) and acute ischemia (incidence rate=0.6; 95% confidence interval=0.3-1.0). Emergent amyloid-related imaging abnormalities with hemosiderin deposition occurred in 32.8% of participants who received placebo; the primary factor associated with these events during the post-baseline period was the number of microhemorrhages at baseline (odds ratio=349.9; 95% confidence interval=247.6-494.4; adjusted p<0.001). CONCLUSION: Safety findings in the placebo-treated group from the A4 study provide a robust characterization of expected safety in a clinical trial population with preclinical Alzheimer's disease. These results may provide context in planning future studies and safety evaluations during ongoing blinded studies in preclinical Alzheimer's disease.


Subject(s)
Alzheimer Disease , Aniline Compounds , Positron-Emission Tomography , Humans , Aged , Female , Male , Aged, 80 and over , Aniline Compounds/therapeutic use , Aniline Compounds/adverse effects , Alzheimer Disease/drug therapy , Alzheimer Disease/diagnostic imaging , Brain/diagnostic imaging , Brain/metabolism , Ethylene Glycols , Double-Blind Method , Amyloid beta-Peptides/metabolism
7.
J Prev Alzheimers Dis ; 11(4): 869-873, 2024.
Article in English | MEDLINE | ID: mdl-39044495

ABSTRACT

BACKGROUND: Increased white matter hyperintensity (WMH) volume visible on MRI is a common finding in Alzheimer's disease (AD). We hypothesized that WMH in preclinical AD is associated with the presence of advanced vessel amyloidosis manifested as microhemorrhages (MCH). OBJECTIVES: 1) To assess the relationship between baseline WMH volume and baseline MCH. 2) To assess the relationship between longitudinal WMH accumulation and last MRI MCH during the double-blind phase of the A4 trial. DESIGN: A multicenter, randomized, double-blind, placebo-controlled, Phase 3 study comparing solanezumab with placebo given as infusions once every 4 weeks over 4.5 years in subjects with preclinical AD, defined as having evidence of elevated brain amyloid before the stage of clinically evident cognitive impairment, with an optional open-label extension period. SETTING: Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease (A4) study. PARTICIPANTS: A sample of 1157 cognitively unimpaired older adults (mean age = 71.9 years [SD = 4.8 years], 59% women, 59% APOE ε4 carriers). MEASUREMENTS: A linear regression model was used to assess the impact of baseline MCH amount (0, 1, 2+) on WMH volume. A linear mixed-effects model was used to assess the impact of last MRI MCH on longitudinal WMH. All models were corrected for age, sex, grey matter volume, cortical amyloid PET, APOE ε4 status, and treatment group. RESULTS: Baseline WMH volume was greater in individuals with more than one MCH compared to those with no MCH (t=4.8, p<0.001). The longitudinal increase in WMH amongst individuals with one (t=2.3, p=0.025) and more than one MCH (t=6.7, p<0.001) at the last MRI was greater than those with no MCH. CONCLUSION: These results indicate a strong association between WMH and MCH, a common manifestation of cerebral amyloid angiopathy and ARIA-H. These results suggest that increased WMH volume may represent an early sign of vessel amyloidosis, likely prior to the emergence of MCH.


Subject(s)
Alzheimer Disease , Antibodies, Monoclonal, Humanized , Magnetic Resonance Imaging , White Matter , Humans , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Female , Male , Aged , White Matter/pathology , White Matter/diagnostic imaging , White Matter/drug effects , Double-Blind Method , Antibodies, Monoclonal, Humanized/therapeutic use , Prodromal Symptoms
8.
Int J Mol Sci ; 25(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38891941

ABSTRACT

Alzheimer's disease (AD) is a devastating disorder with a global prevalence estimated at 55 million people. In clinical studies administering certain anti-beta-amyloid (Aß) antibodies, amyloid-related imaging abnormalities (ARIAs) have emerged as major adverse events. The frequency of these events is higher among apolipoprotein ε4 allele carriers (APOE4) compared to non-carriers. To reflect patients most at risk for vascular complications of anti-Aß immunotherapy, we selected an APPswe/PS1dE9 transgenic mouse model bearing the human APOE4 gene (APPPS1:E4) and compared it with the same APP/PS1 mouse model bearing the human APOE3 gene (APOE ε3 allele; APPPS1:E3). Using histological and biochemical analyses, we characterized mice at three ages: 8, 12, and 16 months. Female and male mice were assayed for general cerebral fibrillar and pyroglutamate (pGlu-3) Aß deposition, cerebral amyloid angiopathy (CAA), microhemorrhages, apoE and cholesterol composition, astrocytes, microglia, inflammation, lysosomal dysfunction, and neuritic dystrophy. Amyloidosis, lipid deposition, and astrogliosis increased with age in APPPS1:E4 mice, while inflammation did not reveal significant changes with age. In general, APOE4 carriers showed elevated Aß, apoE, reactive astrocytes, pro-inflammatory cytokines, microglial response, and neuritic dystrophy compared to APOE3 carriers at different ages. These results highlight the potential of the APPPS1:E4 mouse model as a valuable tool in investigating the vascular side effects associated with anti-amyloid immunotherapy.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Mice, Transgenic , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Mice , Humans , Female , Male , Amyloid beta-Peptides/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy/genetics , Brain/metabolism , Brain/pathology
9.
Alzheimers Dement ; 20(5): 3406-3415, 2024 05.
Article in English | MEDLINE | ID: mdl-38567735

ABSTRACT

INTRODUCTION: Aducanumab selectively targets aggregated forms of amyloid beta (Aß), a neuropathological hallmark of Alzheimer's disease (AD). METHODS: PRIME was a Phase 1b, double-blind, randomized clinical trial of aducanumab. During the 12-month placebo-controlled period, participants with prodromal AD or mild AD dementia were randomized to receive aducanumab or placebo. At week 56, participants could enroll in a long-term extension (LTE), in which all participants received aducanumab. The primary endpoint was safety and tolerability. RESULTS: Amyloid-related imaging abnormalities-edema (ARIA-E) were the most common adverse event. Dose titration was associated with a decrease in the incidence of ARIA-E. Over 48 months, aducanumab decreased brain amyloid levels in a dose- and time-dependent manner. Exploratory endpoints suggested a continued benefit in the reduction of clinical decline over 48 months. DISCUSSION: The safety profile of aducanumab remained unchanged in the LTE of PRIME. Amyloid plaque levels continued to decrease in participants treated with aducanumab. HIGHLIGHTS: PRIME was a Phase 1b, double-blind, randomized clinical trial of aducanumab. We report cumulative safety and 48-month efficacy results from PRIME. Amyloid-related imaging abnormalities-edema (ARIA-E) were the most common adverse event (AE); 61% of participants with ARIA-E were asymptomatic. Dose titration was associated with a decrease in the incidence of ARIA-E. Aducanumab decreased levels of amyloid beta (Aß) in a dose- and time-dependent manner.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Antibodies, Monoclonal, Humanized , Humans , Double-Blind Method , Antibodies, Monoclonal, Humanized/therapeutic use , Alzheimer Disease/drug therapy , Male , Female , Aged , Amyloid beta-Peptides/metabolism , Brain/diagnostic imaging , Brain/drug effects , Brain/pathology , Treatment Outcome , Plaque, Amyloid/drug therapy , Dose-Response Relationship, Drug
10.
J Clin Med ; 13(5)2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38592119

ABSTRACT

Anti-amyloid immunotherapies have recently emerged as treatments for Alzheimer's disease. While these therapies have demonstrated efficacy in clearing amyloid-ß and slowing cognitive decline, they have also been associated with amyloid-related imaging abnormalities (ARIA) which include both edema (ARIA-E) and hemorrhage (ARIA-H). Given that ARIA have been associated with significant morbidity in cases of antithrombotic or thrombolytic therapy, an understanding of mechanisms of and risk factors for ARIA is of critical importance for stroke care. We discuss the latest data regarding mechanisms of ARIA, including the role of underlying cerebral amyloid angiopathy, and implications for ischemic stroke prevention and management.

11.
Neurol Ther ; 13(3): 677-695, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38581616

ABSTRACT

INTRODUCTION: Donanemab, a monoclonal antibody directed against an insoluble, modified, N-terminal truncated form of amyloid beta, demonstrated efficacy and safety in patients with early, symptomatic Alzheimer's disease (AD) in the phase 3 TRAILBLAZER-ALZ 2 trial. Here, we report clinical outcomes, biomarkers, and safety results for the Japanese subpopulation. METHODS: TRAILBLAZER-ALZ 2 (N = 1736) was conducted in eight countries, including Japan (enrollment June 2020-November 2021; database lock April 2023). Participants (60-85 years) with early, symptomatic AD (mild cognitive impairment/mild dementia), Mini-Mental State Examination score 20-28, and confirmed amyloid and tau pathology were randomized 1:1 (stratified by tau status) to intravenous donanemab (700 mg for three doses, then 1400 mg/dose) or placebo every 4 weeks for 72 weeks. Primary outcome was change from baseline to week 76 in integrated Alzheimer's Disease Rating Scale (iADRS) score. Other outcomes included clinical measures of cognitive and functional impairment, biomarkers, and safety. RESULTS: Of 88 Japanese participants (43 placebo, 45 donanemab), 7 in each group discontinued. Least-squares mean (LSM) change from baseline in iADRS score at week 76 was smaller with donanemab than with placebo in the combined (low-medium tau and high tau) and low-medium tau (N = 76) subpopulations (LSM change difference: 4.43 and 3.99, representing 38.8% and 40.2% slowing of disease progression, respectively). Slowing of AD progression with donanemab was also observed for other clinical outcomes. Marked decreases in amyloid plaque and plasma phosphorylated tau 217 were observed; amyloid clearance (< 24.1 Centiloids) was observed in 83.3% of the combined donanemab and 0% of the combined placebo groups. Amyloid-related imaging abnormalities of edema/effusions occurred in ten (22.2%) donanemab-treated participants (one [2.2%] symptomatic) and one (2.3%) placebo-treated participant. CONCLUSIONS: The overall efficacy and safety of donanemab in Japanese participants were similar to the global TRAILBLAZER-ALZ 2 population. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT04437511.

12.
Alzheimers Dement ; 20(4): 3127-3140, 2024 04.
Article in English | MEDLINE | ID: mdl-38323738

ABSTRACT

The Centers for Medicare & Medicaid Services (CMS) established a class-based National Coverage Determination (NCD) for monoclonal antibodies directed against amyloid for Alzheimer's disease (AD) with patient access through Coverage with Evidence Development (CED) based on three questions. This review, focused on donanemab, answers each of these CED questions with quality evidence. TRAILBLAZER-ALZ registration trials are presented with supporting literature and real-world data to answer CED questions for donanemab. TRAILBLAZER-ALZ registration trials demonstrated that donanemab significantly slowed cognitive and functional decline in amyloid-positive early symptomatic AD participants, and lowered their risk of disease progression while key safety risks occurred primarily within the first 6 months and then declined. Donanemab meaningfully improved health outcomes with a manageable safety profile in an early symptomatic AD population, representative of Medicare populations across diverse practice settings. The donanemab data provide the necessary level of evidence for CMS to open a reconsideration of their NCD. HIGHLIGHTS: Donanemab meaningfully improved outcomes in trial participants with early symptomatic Alzheimer's disease. Comorbidities in trial participants were consistent with the Medicare population. Co-medications in trial participants were consistent with the Medicare population. Risks associated with treatment tended to occur in the first 6 months. Risks of amyloid-related imaging abnormalities were managed with careful observation and magnetic resonance imaging monitoring.


Subject(s)
Alzheimer Disease , Antibodies, Monoclonal, Humanized , Noncommunicable Diseases , Aged , Humans , United States , Alzheimer Disease/pathology , Medicare , Amyloid , Amyloidogenic Proteins , Amyloid beta-Peptides
14.
EBioMedicine ; 94: 104665, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37392597

ABSTRACT

BACKGROUND: Anti-amyloid vaccines may offer a convenient, affordable, and accessible means of preventing and treating Alzheimer's disease. UB-311 is an anti-amyloid-ß active immunotherapeutic vaccine shown to be well-tolerated and to have a durable antibody response in a phase 1 trial. This phase 2a study assessed the safety, immunogenicity, and preliminary efficacy of UB-311 in participants with mild Alzheimer's disease. METHODS: A 78-week, randomised, double-blind, placebo-controlled, parallel-group, multicentre, phase 2a study was conducted in Taiwan. Participants were randomised in a 1:1:1 ratio to receive seven intramuscular injections of UB-311 (Q3M arm), or five doses of U311 with two doses of placebo (Q6M arm), or seven doses of placebo (placebo arm). The primary endpoints were safety, tolerability, and immunogenicity of UB-311. Safety was assessed in all participants who received at least one dose of investigational product. This study was registered at ClinicalTrials.gov (NCT02551809). FINDINGS: Between 7 December 2015 and 28 August 2018, 43 participants were randomised. UB-311 was safe, well-tolerated, and generated a robust immune response. The three treatment-emergent adverse events (TEAEs) with the highest incidence were injection-site pain (14 TEAEs in seven [16%] participants), amyloid-related imaging abnormality with microhaemorrhages and haemosiderin deposits (12 TEAEs in six [14%] participants), and diarrhoea (five TEAEs in five [12%] participants). A 97% antibody response rate was observed and maintained at 93% by the end of the study across both UB-311 arms. INTERPRETATION: These results support the continued development of UB-311. FUNDING: Vaxxinity, Inc. (Formerly United Neuroscience Ltd.).


Subject(s)
Alzheimer Disease , Vaccines , Humans , Alzheimer Disease/therapy , Amyloid beta-Peptides , Vaccination , Antibody Formation , Double-Blind Method
15.
J Prev Alzheimers Dis ; 10(3): 362-377, 2023.
Article in English | MEDLINE | ID: mdl-37357276

ABSTRACT

Lecanemab (Leqembi®) is approved in the United States for the treatment of Alzheimer's disease (AD) to be initiated in early AD (mild cognitive impairment [MCI] due to AD or mild AD dementia) with confirmed brain amyloid pathology. Appropriate Use Recommendations (AURs) are intended to help guide the introduction of new therapies into real-world clinical practice. Community dwelling patients with AD differ from those participating in clinical trials. Administration of lecanemab at clinical trial sites by individuals experienced with monoclonal antibody therapy also differs from the community clinic-based administration of lecanemab. These AURs use clinical trial data as well as research and care information regarding AD to help clinicians administer lecanemab with optimal safety and opportunity for effectiveness. Safety and efficacy of lecanemab are known only for patients like those participating in the phase 2 and phase 3 lecanemab trials, and these AURs adhere closely to the inclusion and exclusion criteria of the trials. Adverse events may occur with lecanemab including amyloid related imaging abnormalities (ARIA) and infusion reactions. Monitoring guidelines for these events are detailed in this AUR. Most ARIA with lecanemab is asymptomatic, but a few cases are serious or, very rarely, fatal. Microhemorrhages and rare macrohemorrhages may occur in patients receiving lecanemab. Anticoagulation increases the risk of hemorrhage, and the AUR recommends that patients requiring anticoagulants not receive lecanemab until more data regarding this interaction are available. Patients who are apolipoprotein E ε4 (APOE4) gene carriers, especially APOE4 homozygotes, are at higher risk for ARIA, and the AUR recommends APOE genotyping to better inform risk discussions with patients who are lecanemab candidates. Clinician and institutional preparedness are mandatory for use of lecanemab, and protocols for management of serious events should be developed and implemented. Communication between clinicians and therapy candidates or those on therapy is a key element of good clinical practice for the use of lecanemab. Patients and their care partners must understand the potential benefits, the potential harms, and the monitoring requirements for treatment with this agent. Culture-specific communication and building of trust between clinicians and patients are the foundation for successful use of lecanemab.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Humans , Apolipoprotein E4/genetics , Alzheimer Disease/genetics , Antibodies, Monoclonal/therapeutic use , Amyloid
16.
Brain ; 146(11): 4414-4424, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37280110

ABSTRACT

Excess accumulation and aggregation of toxic soluble and insoluble amyloid-ß species in the brain are a major hallmark of Alzheimer's disease. Randomized clinical trials show reduced brain amyloid-ß deposits using monoclonal antibodies that target amyloid-ß and have identified MRI signal abnormalities called amyloid-related imaging abnormalities (ARIA) as possible spontaneous or treatment-related adverse events. This review provides a comprehensive state-of-the-art conceptual review of radiological features, clinical detection and classification challenges, pathophysiology, underlying biological mechanism(s) and risk factors/predictors associated with ARIA. We summarize the existing literature and current lines of evidence with ARIA-oedema/effusion (ARIA-E) and ARIA-haemosiderosis/microhaemorrhages (ARIA-H) seen across anti-amyloid clinical trials and therapeutic development. Both forms of ARIA may occur, often early, during anti-amyloid-ß monoclonal antibody treatment. Across randomized controlled trials, most ARIA cases were asymptomatic. Symptomatic ARIA-E cases often occurred at higher doses and resolved within 3-4 months or upon treatment cessation. Apolipoprotein E haplotype and treatment dosage are major risk factors for ARIA-E and ARIA-H. Presence of any microhaemorrhage on baseline MRI increases the risk of ARIA. ARIA shares many clinical, biological and pathophysiological features with Alzheimer's disease and cerebral amyloid angiopathy. There is a great need to conceptually link the evident synergistic interplay associated with such underlying conditions to allow clinicians and researchers to further understand, deliberate and investigate on the combined effects of these multiple pathophysiological processes. Moreover, this review article aims to better assist clinicians in detection (either observed via symptoms or visually on MRI), management based on appropriate use recommendations, and general preparedness and awareness when ARIA are observed as well as researchers in the fundamental understanding of the various antibodies in development and their associated risks of ARIA. To facilitate ARIA detection in clinical trials and clinical practice, we recommend the implementation of standardized MRI protocols and rigorous reporting standards. With the availability of approved amyloid-ß therapies in the clinic, standardized and rigorous clinical and radiological monitoring and management protocols are required to effectively detect, monitor, and manage ARIA in real-world clinical settings.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/complications , Antibodies, Monoclonal, Humanized/therapeutic use , Amyloid beta-Peptides/metabolism , Brain/metabolism , Amyloid , Amyloidogenic Proteins
17.
J Alzheimers Dis ; 93(2): 803-813, 2023.
Article in English | MEDLINE | ID: mdl-37125554

ABSTRACT

Host responses to anti-amyloid-ß (Aß) antibody therapy are evident in neuroimaging changes and clinical symptoms in a subset of clinical trial subjects receiving such therapy. The pathological basis for the imaging changes and clinical symptoms is not known, nor is the precise mechanism of Aß clearing. We report the autopsy findings in a 65-year-old woman who received three open label infusions of the experimental anti-Aß drug lecanemab over about one month. Four days after the last infusion, she was treated with tissue plasminogen activator for acute stroke symptoms and died several days later with multifocal hemorrhage. Neuropathological examination demonstrated histiocytic vasculitis involving blood vessels with cerebral amyloid angiopathy. Fragmentation and phagocytosis of vascular Aß were present throughout the cerebral cortex. Phagocytosis of parenchymal Aß plaques was noted. Changes suggestive of Aß and phosphorylated tau "clearing" were also noted. The findings overall suggest that anti-Aß treatment stimulated a host response to Aß, i.e., target engagement. The findings also provide evidence that amyloid-related imaging abnormalities might be indicative of an Aß phagocytic syndrome within cerebral vasculature and parenchymal brain tissue in some cases.


Subject(s)
Alzheimer Disease , Cerebral Amyloid Angiopathy , Female , Humans , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/therapy , Tissue Plasminogen Activator , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/diagnostic imaging , Cerebral Amyloid Angiopathy/therapy , Cerebral Amyloid Angiopathy/etiology , Brain/pathology , Immunotherapy/adverse effects
18.
J Alzheimers Dis ; 91(3): 1173-1183, 2023.
Article in English | MEDLINE | ID: mdl-36565118

ABSTRACT

BACKGROUND: Cerebrospinal fluid (CSF) biomarkers in patients with cerebral amyloid angiopathy-related inflammation (CAA-ri) have demonstrated inconsistent results. OBJECTIVE: We investigated the relationship between CSF amyloid-ß protein (Aß) and vascular pathological findings to elucidate the mechanisms of Aß elimination from the brain in CAA-ri. METHODS: We examined Aß40 and Aß42 levels in CSF samples in 15 patients with CAA-ri and 15 patients with Alzheimer's disease and cerebral amyloid angiopathy (AD-CAA) using ELISA as a cross-sectional study. Furthermore, we pathologically examined Aß40 and Aß42 depositions on the leptomeningeal blood vessels (arteries, arterioles, and veins) using brain biopsy samples from six patients with acute CAA-ri and brain tissues of two autopsied patients with CAA-ri. RESULTS: The median Aß40 and Aß42 levels in the CSF showed no significant difference between pre-treatment CAA-ri (Aß40, 6837 pg/ml; Aß42, 324 pg/ml) and AD-CAA (Aß40, 7669 pg/ml, p = 0.345; Aß42, 355 pg/ml, p = 0.760). Aß40 and Aß42 levels in patients with post-treatment CAA-ri (Aß40, 1770 pg/ml, p = 0.056; Aß42, 167 pg/ml, p = 0.006) were lower than those in patients with pre-treatment CAA-ri. Regarding Aß40 and Aß42 positive arteries, acute CAA-ri cases showed a higher frequency of partially Aß-deposited blood vessels than postmortem CAA-ri cases (Aß40, 20.8% versus 3.9%, p = 0.0714; Aß42, 27.4% versus 2.0%, p = 0.0714, respectively). CONCLUSION: Lower levels of CSF Aß40 and Aß42 could be biomarkers for the cessation of inflammation in CAA-ri reflecting the recovery of the intramural periarterial drainage pathway and vascular function.


Subject(s)
Alzheimer Disease , Cerebral Amyloid Angiopathy , Humans , Cross-Sectional Studies , Cerebral Amyloid Angiopathy/pathology , Amyloid beta-Peptides/metabolism , Brain/pathology , Alzheimer Disease/pathology , Inflammation/metabolism , Biomarkers/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid
19.
Radiol Case Rep ; 18(1): 275-279, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36388611

ABSTRACT

Patients with Alzheimer's disease who have been given monoclonal antibodies targeting amyloid-ß (Aß) (eg, gantenerumab, donanemab, lecanemab, and aducanumab) for scientific purposes may have a spectrum of imaging findings known as amyloid-related imaging abnormalities (ARIA), shown on brain magnetic resonance imaging (MRI) scans. These neuroimaging abnormalities are caused by antibody-mediated destruction of accumulated Aß aggregates in cerebral blood vessels and brain parenchyma. ARIA may demonstrate as brain edema or sulcal effusion (ARIA-E) or as hemosiderin deposits caused by brain parenchymal or pial hemorrhage (ARIA-H). The current study explores 2 cases with interval development of FLAIR hyper signal intensity along the bilateral corticospinal tracts in the motor cortex/precentral gyri after treatment by aducanumab. We believe this manifestation is a subtype of ARIA-A that has not been explored earlier. Our first case was a 72-year-old woman with a history of HTN and kidney transplant (polycystic kidney) who presented with mild cognitive impairment with clinical findings consistent with early Alzheimer's disease. After receiving 3 doses of aducunumab and experiencing cognition improvement, she underwent a brain MRI because of dizziness and vertigo. The brain MRI demonstrated new FLAIR hyper signal intensity in subcortical regions of precentral gyri (motor cortex) symmetrically as well as trace subarachnoid hemorrhage at the vertex compatible with ARIA-E and ARIA-H. Our second case was an 85-year-old woman with a history of small lymphocytic leukemia which was treated 20 years earlier. After orthopedic surgery 2 years ago, she developed dementia with anterograde amnesia. Since then, Aricept and Namenda have been started, but there have been no improvements in her subjective condition. The initial Amyloid PET/MR imaging showed diffuse cerebral Amyloid deposition. After tolerating 6 doses of aducanumab a safety MRI revealed new bilateral symmetric FLAIR hyper signal intensity in the subcortical motor cortex. Results of our study suggest that the subcortical corticospinal tract is another hotspot for ARIA findings. Hence, these regions might be an unknown site for both the action and adverse effects of aducanumab on amyloid plaques with secondary inflammation. In addition, radiologists must take this phenomenon into the account, and be cognizant that the FLAIR hyper signal intensities should not be misinterpreted as motor neuron disease (eg, amyotrophic lateral sclerosis).

20.
Clin Neuroradiol ; 33(2): 293-306, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36443509

ABSTRACT

Superficial siderosis (SS) of the central nervous system constitutes linear hemosiderin deposits in the leptomeninges and the superficial layers of the cerebrum and the spinal cord. Infratentorial (i) SS is likely due to recurrent or continuous slight bleeding into the subarachnoid space. It is assumed that spinal dural pathologies often resulting in cerebrospinal fluid (CSF) leakage is the most important etiological group which causes iSS and detailed neuroradiological assessment of the spinal compartment is necessary. Further etiologies are neurosurgical interventions, trauma and arteriovenous malformations. Typical neurological manifestations of this classical type of iSS are slowly progressive sensorineural hearing impairment and cerebellar symptoms, such as ataxia, kinetic tremor, nystagmus and dysarthria. Beside iSS, a different type of SS restricted to the supratentorial compartment can be differentiated, i.e. cortical (c) SS, especially in older people often due to cerebral amyloid angiopathy (CAA). Clinical presentation of cSS includes transient focal neurological episodes or "amyloid spells". In addition, spontaneous and amyloid beta immunotherapy-associated CAA-related inflammation may cause cSS, which is included in the hemorrhagic subgroup of amyloid-related imaging abnormalities (ARIA). Because a definitive diagnosis requires a brain biopsy, knowledge of neuroimaging features and clinical findings in CAA-related inflammation is essential. This review provides neuroradiological hallmarks of the two groups of SS and give an overview of neurological symptoms and differential diagnostic considerations.


Subject(s)
Cerebral Amyloid Angiopathy , Siderosis , Humans , Aged , Siderosis/diagnostic imaging , Siderosis/etiology , Amyloid beta-Peptides , Magnetic Resonance Imaging , Cerebral Hemorrhage , Cerebral Amyloid Angiopathy/complications , Cerebral Amyloid Angiopathy/diagnostic imaging , Inflammation
SELECTION OF CITATIONS
SEARCH DETAIL