12 results on '"Tariot, Pierre N."'
Search Results
2. BACE inhibition causes rapid, regional, and non-progressive volume reduction in Alzheimer's disease brain.
- Author
-
Sur, Cyrille, Kost, James, Scott, David, Adamczuk, Katarzyna, Fox, Nick C, Cummings, Jeffrey L, Tariot, Pierre N, Aisen, Paul S, Vellas, Bruno, Voss, Tiffini, Mahoney, Erin, Mukai, Yuki, Kennedy, Matthew E, Lines, Christopher, Michelson, David, and Egan, Michael F
- Subjects
ALZHEIMER'S disease ,BRAIN diseases ,AMYLOID ,ALZHEIMER'S patients ,BRAIN metabolism ,BRAIN ,SULFUR compounds ,RESEARCH ,NERVE tissue proteins ,HIPPOCAMPUS (Brain) ,CLINICAL trials ,RESEARCH methodology ,PROTEOLYTIC enzymes ,MAGNETIC resonance imaging ,MEDICAL cooperation ,EVALUATION research ,TREATMENT effectiveness ,COMPARATIVE studies ,BLIND experiment ,ENZYME inhibitors ,PEPTIDES ,CHEMICAL inhibitors - Abstract
In the phase 3 EPOCH trial (Clinicaltrials.gov; NCT01739348), treatment with the BACE inhibitor verubecestat failed to improve cognition in patients with mild-to-moderate Alzheimer's disease, but was associated with reduced hippocampal volume after 78 weeks as assessed by MRI. The aims of the present exploratory analyses were to: (i) characterize the effect of verubecestat on brain volume by evaluating the time course of volumetric MRI changes for a variety of brain regions; and (ii) understand the mechanism through which verubecestat might cause hippocampal (and other brain region) volume loss by assessing its relationship to measures of amyloid, neurodegeneration, and cognition. Participants were aged 55-85 years with probable Alzheimer's disease dementia and a Mini Mental State Examination score ≥15 and ≤26. MRIs were obtained at baseline and at Weeks 13, 26, 52 and 78 of treatment. MRIs were segmented using Freesurfer and analysed using a tensor-based morphometry method. PET amyloid data were obtained with 18F-flutemetamol (Vizamyl®) at baseline and Week 78. Standardized uptake value ratios were generated with subcortical white matter as a reference region. Neurofilament light chain in the CSF was assessed as a biomarker of neurodegeneration. Compared with placebo, verubecestat showed increased MRI brain volume loss at Week 13 with no evidence of additional loss through Week 78. The verubecestat-related volumetric MRI loss occurred predominantly in amyloid-rich brain regions. Correlations between amyloid burden at baseline and verubecestat-related volumetric MRI reductions were not significant (r = 0.05 to 0.26, P-values > 0.27). There were no significant differences between verubecestat and placebo in changes from baseline in CSF levels of neurofilament light chain at Week 78 (increases of 7.2 and 14.6 pg/ml for verubecestat versus 19.7 pg/ml for placebo, P-values ≥ 0.1). There was a moderate correlation between volumetric MRI changes and cognitive decline in all groups including placebo at Week 78 (e.g. r = -0.45 to -0.55, P < 0.001 for whole brain), but the correlations were smaller at Week 13 and significant only for the verubecestat groups (e.g. r = -0.15 and -0.11, P < 0.04 for whole brain). Our results suggest that the verubecestat-associated MRI brain volume loss is not due to generalized, progressive neurodegeneration, but may be mediated by specific effects on BACE-related amyloid processes. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
3. GeneMatch: A novel recruitment registry using at‐home APOE genotyping to enhance referrals to Alzheimer's prevention studies.
- Author
-
Langbaum, Jessica B., Karlawish, Jason, Roberts, J. Scott, Wood, Elisabeth M., Bradbury, Angela, High, Nellie, Walsh, Trisha L., Gordon, David, Aggarwal, Raj, Davis, Peter, Stowell, Carter, Trisko, Lane, Langlois, Carolyn M., Reiman, Eric M., and Tariot, Pierre N.
- Abstract
Introduction: Recruitment for Alzheimer's disease (AD) prevention research studies is challenging because of lack of awareness among cognitively healthy adults coupled with the high screen fail rate due to participants not having a genetic risk factor or biomarker evidence of the disease. Participant recruitment registries offer one solution for efficiently and effectively identifying, characterizing, and connecting potential eligible volunteers to studies. Methods: Individuals aged 55‐75 years who live in the United States and self‐report not having a diagnosis of cognitive impairment such as MCI or dementia are eligible to join GeneMatch. Participants enroll online and are provided a cheek swab kit for DNA extraction and apolipoprotein E (APOE) genotyping. Participants are not told their APOE results, although the results may be used in part to help match participants to AD prevention studies. Results: As of August 2018, 75,351 participants had joined GeneMatch. Nearly 30% of participants have one APOE4 allele, and approximately 3% have two APOE4 alleles. The percentages of APOE4 heterozygotes and homozygotes are inversely associated with age (P <.001). Discussion: GeneMatch, the first trial‐independent research enrollment program designed to recruit and refer cognitively healthy adults to AD prevention studies based in part on APOE test results, provides a novel mechanism to accelerate prescreening and enrollment for AD prevention trials. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
4. Effect of Idalopirdine as Adjunct to Cholinesterase Inhibitors on Change in Cognition in Patients With Alzheimer Disease: Three Randomized Clinical Trials.
- Author
-
Atri, Alireza, Frölich, Lutz, Ballard, Clive, Tariot, Pierre N., Molinuevo, José Luis, Boneva, Neli, Windfeld, Kristian, Raket, Lars L., Cummings, Jeffrey L., Frölich, Lutz, and Molinuevo, José Luis
- Subjects
ALZHEIMER'S disease treatment ,SEROTONIN antagonists ,CHOLINESTERASE inhibitors ,CLINICAL trials ,DRUG dosage ,ALZHEIMER'S disease research ,THERAPEUTICS ,HYDROCARBONS ,PIPERIDINE ,GALANTHAMINE ,ALZHEIMER'S disease ,AMINES ,COMBINATION drug therapy ,COGNITION ,COMPARATIVE studies ,DOSE-effect relationship in pharmacology ,ACCIDENTAL falls ,RESEARCH methodology ,MEDICAL cooperation ,RESEARCH ,STATISTICAL sampling ,EVALUATION research ,RANDOMIZED controlled trials ,TREATMENT effectiveness ,BLIND experiment ,INDOLE compounds ,PSYCHOLOGY - Abstract
Importance: New therapeutic approaches for Alzheimer disease (AD) are needed.Objective: To assess whether idalopirdine, a selective 5-hydroxytryptamine-6 receptor antagonist, is effective for symptomatic treatment of mild to moderate AD.Design, Setting, and Participants: Three randomized clinical trials that included 2525 patients aged 50 years or older with mild to moderate AD (study 1: n = 933 patients at 119 sites; study 2: n = 858 at 158 sites; and study 3: n = 734 at 126 sites). The 24-week studies were conducted from October 2013 to January 2017; final follow-up on January 12, 2017.Interventions: Idalopirdine (10, 30, or 60 mg/d) or placebo added to cholinesterase inhibitor treatment (donepezil in studies 1 and 2; donepezil, rivastigmine, or galantamine in study 3).Main Outcomes and Measures: Primary end point in all 3 studies: change in cognition total score (range, 0-70; a lower score indicates less impairment) from baseline to 24 weeks measured by the 11-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-Cog); key secondary end points: Alzheimer's Disease Cooperative Study-Clinical Global Impression of Change Scale and 23-item Activities of Daily Living Inventory scores. Dose group efficacy required a significant benefit over placebo for the primary end point and 1 or more key secondary end points. Safety data and adverse event profiles were recorded.Results: Among 2525 patients randomized in the 3 trials (mean age, 74 years; mean baseline ADAS-Cog total score, 26; between 62% and 65% of participants were women), 2254 (89%) completed the studies. In study 1, the mean change in ADAS-Cog total score between baseline and 24 weeks was 0.37 for the 60-mg dose of idalopirdine group, 0.61 for the 30-mg dose group, and 0.41 for the placebo group (adjusted mean difference vs placebo, 0.05 [95% CI, -0.88 to 0.98] for the 60-mg dose group and 0.33 [95% CI, -0.59 to 1.26] for the 30-mg dose group). In study 2, the mean change in ADAS-Cog total score between baseline and 24 weeks was 1.01 for the 30-mg dose of idalopirdine group, 0.53 for the 10-mg dose group, and 0.56 for the placebo group (adjusted mean difference vs placebo, 0.63 [95% CI, -0.38 to 1.65] for the 30-mg dose group; given the gated testing strategy and the null findings at the 30-mg dose, statistical comparison of the 10-mg dose was not performed). In study 3, the mean change in ADAS-Cog total score between baseline and 24 weeks was 0.38 for the 60-mg dose of idalopirdine group and 0.82 for the placebo group (adjusted mean difference vs placebo, -0.55 [95% CI, -1.45 to 0.36]). Treatment-emergent adverse events occurred in between 55.4% and 69.7% of participants in the idalopirdine groups vs between 56.7% and 61.4% of participants in the placebo groups.Conclusions and Relevance: In patients with mild to moderate AD, the use of idalopirdine compared with placebo did not improve cognition over 24 weeks of treatment. These findings do not support the use of idalopirdine for the treatment of AD.Trial Registration: clinicaltrials.gov Identifiers: NCT01955161, NCT02006641, and NCT02006654. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
5. An empirically derived composite cognitive test score with improved power to track and evaluate treatments for preclinical Alzheimer's disease.
- Author
-
Langbaum, Jessica B., Hendrix, Suzanne B., Ayutyanont, Napatkamon, Chen, Kewei, Fleisher, Adam S., Shah, Raj C., Barnes, Lisa L., Bennett, David A., Tariot, Pierre N., and Reiman, Eric M.
- Abstract
Background There is growing interest in the evaluation of preclinical Alzheimer's disease (AD) treatments. As a result, there is a need to identify a cognitive composite that is sensitive to track preclinical AD decline to be used as a primary endpoint in treatment trials. Methods Longitudinal data from initially cognitively normal, 70- to 85-year-old participants in three cohort studies of aging and dementia from the Rush Alzheimer's Disease Center were examined to empirically define a composite cognitive endpoint that is sensitive to detect and track cognitive decline before the onset of cognitive impairment. The mean-to-standard deviation ratios (MSDRs) of change over time were calculated in a search for the optimal combination of cognitive tests/subtests drawn from the neuropsychological battery in cognitively normal participants who subsequently progressed to clinical stages of AD during 2- and 5-year periods, using data from those who remained unimpaired during the same period to correct for aging and practice effects. Combinations that performed well were then evaluated for representation of relevant cognitive domains, robustness across individual years before diagnosis, and occurrence of selected items within top performing combinations. Results The optimal composite cognitive test score comprised seven cognitive tests/subtests with an MSDR = 0.964. By comparison, the most sensitive individual test score was Logical Memory Delayed Recall with an MSDR = 0.64. Conclusions We have identified a composite cognitive test score representing multiple cognitive domains that has improved power compared with the most sensitive single test item to track preclinical AD decline and evaluate preclinical AD treatments. We are confirming the power of the composite in independent cohorts and with other analytical approaches, which may result in refinements, have designated it as the primary endpoint in the Alzheimer's Prevention Initiative's preclinical treatment trials for individuals at high imminent risk for developing symptoms due to late-onset AD. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
6. Alzheimer's Prevention Initiative: A Plan to Accelerate the Evaluation of Presymptomatic Treatments.
- Author
-
Ashford, Rosen, Adamson, Bayley, Sabri, Furst, Black, Weiner, Reiman, Eric M., Tariot, Pierre N., Langbaum, Jessica B.S., Ayutyanont, Napatkamon, Fleisher, Adam S., Chen, Kewei, Kosik, Kenneth S., Lopera, Francisco, Caselli, Richard J., and Quiroz, Yakeel T.
- Subjects
ALZHEIMER'S disease prevention ,ALZHEIMER'S disease research ,BRAIN imaging ,CEREBROSPINAL fluid ,AMYLOID ,THERAPEUTICS - Abstract
There is an urgent need to find effective presymptomatic Alzheimer's disease (AD) treatments that reduce the risk of AD symptoms or prevent them completely. It currently takes too many healthy people, too much money and too many years to evaluate the range of promising presymptomatic treatments using clinical endpoints. We have used brain imaging and other measurements to track some of the earliest changes associated with the predisposition to AD. We have proposed the Alzheimer's Prevention Initiative (API) to evaluate investigational amyloid-modifying treatments in healthy people who, based on their age and genetic background, are at the highest imminent risk of developing symptomatic AD using brain imaging, cerebrospinal fluid (CSF), and cognitive endpoints. In one trial, we propose to study AD-causing presenilin 1 [PS1] mutation carriers from the world's largest early-onset AD kindred in Antioquia, Colombia, close to their estimated average age at clinical onset. In another trial, we propose to study apolipoprotein E (APOE) ε4 homozygotes (and possibly heterozygotes) close to their estimated average age at clinical onset. The API has several goals: 1) to evaluate investigational AD-modifying treatments sooner than otherwise possible; 2) to determine the extent to which the treatment's brain imaging and other biomarker effects predict a clinical benefit—information needed to help qualify biomarker endpoints for use in pivotal prevention trials; 3) to provide a better test of the amyloid hypothesis than clinical trials in symptomatic patients, when these treatments may be too little too late to exert their most profound effect; 4) to establish AD prevention registries needed to support these and other presymptomatic AD trials; and 5) to give those individuals at highest imminent risk of AD symptoms access to the most promising investigational treatments in clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
7. Meta-analysis of six-month memantine trials in Alzheimer’s disease.
- Author
-
Doody, Rachelle Smith, Tariot, Pierre N., Pfeiffer, Eric, Olin, Jason T., and Graham, Stephen M.
- Subjects
CLINICAL trials ,ALZHEIMER'S disease ,CLINICAL drug trials ,INDUSTRIAL safety ,SENSITIVITY (Personality trait) - Abstract
Abstract: Background: Clinical trials have suggested benefits of memantine for all stages of Alzheimer’s disease (AD). A meta-analysis of memantine trials in outpatients with probable AD was conducted. Methods: Six randomized, placebo-controlled trials fulfilled inclusion criteria. Cognitive, global, functional, behavioral, and safety measures were evaluated by using fixed or random effects models. Sensitivity analysis was performed to determine potential effects of disease severity and concomitant cholinesterase inhibitor use. Results: Overall, memantine showed statistically significant, homogenous benefits on global and functional outcomes and statistically significant but heterogeneous benefit on cognition. In pooled moderate to severe AD studies, effects were significant and homogeneous on global, functional, and behavioral measures; in pooled mild to moderate studies, significant and homogeneous effects were found on global rating and cognition. Safety profiles suggested excellent safety and tolerability, with no differences between the severity groups. Conclusions: Memantine consistently benefited patients in all stages of AD and was well-tolerated. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
8. Memantine Treatment in Patients With Moderate to Severe Alzheimer Disease Already Receiving Donepezil: A Randomized Controlled Trial.
- Author
-
Tariot, Pierre N., Farlow, Martin R., Grossberg, George T., Graham, Stephen M., McDonald, Scott, and Gergel, Ivan
- Subjects
- *
ALZHEIMER'S disease treatment , *CLINICAL trials , *MEDICAL research - Abstract
Context: Memantine is a low- to moderate-affinity, uncompetitive N-methyl-
D -aspartate receptor antagonist. Controlled trials have demonstrated the safety and efficacy of memantine monotherapy for patients with moderate to severe Alzheimer disease (AD) but no controlled trials of memantine in patients receiving a cholinesterase inhibitor have been performed. Objective: To compare the efficacy and safety of memantine vs placebo in patients with moderate to severe AD already receiving stable treatment with donepezil. Design, Setting, and Participants: A randomized, double-blind, placebo-controlled clinical trial of 404 patients with moderate to severe AD and Mini-Mental State Examination scores of 5 to 14, who received stable doses of donepezil, conducted at 37 US sites between June 11, 2001, and June 3, 2002. A total of 322 patients (80%) completed the trial. Interventions: Participants were randomized to receive memantine (starting dose 5 mg/d, increased to 20 mg/d, n = 203) or placebo (n = 201) for 24 weeks. Main Outcome Measures: Change from baseline on the Severe Impairment Battery (SIB), a measure of cognition, and on a modified 19-item AD Cooperative Study–Activities of Daily Living Inventory (ADCS-ADL[sub 19]). Secondary outcomes included a Clinician's Interview-Based Impression of Change Plus Caregiver Input (CIBIC-Plus), the Neuropsychiatric Inventory, and the Behavioral Rating Scale for Geriatric Patients (BGP Care Dependency Subscale). Results: The change in total mean (SE) scores favored memantine vs placebo treatment for SIB (possible score range, 0-100), 0.9 (0.67) vs –2.5 (0.69), respectively (P<.001); ADCS-ADL[sub 19] (possible score range, 0-54), –2.0 (0.50) vs –3.4 (0.51), respectively (P = .03); and the CIBIC-Plus (possible score range, 1-7), 4.41 (0.074) vs 4.66 (0.075), respectively (P = .03). All other secondary measures showed significant benefits of memantine treatment. Treatment discontinuations because of adverse events for memantine vs placebo were 15 (7.4%) vs 25 (12.4%), respectively. Conclusions: In patients with moderate to severe AD receiving stable doses of donepezil, memantine resulted in significantly better outcomes than placebo on measures of cognition, activities of daily living, global outcome, and behavior and was well tolerated. These results, together with previous studies, suggest that memantine represents a new approach for the treatment of patients with moderate to severe AD. [ABSTRACT FROM AUTHOR]- Published
- 2004
- Full Text
- View/download PDF
9. Medical Management of Advanced Dementia.
- Author
-
Tariot, Pierre N.
- Subjects
- *
ALZHEIMER'S disease , *DEMENTIA , *BRAIN diseases , *PSYCHIATRIC drugs , *CLINICAL trials , *ACETYLCHOLINESTERASE - Abstract
A cure for Alzheimer's disease (AD) is still far off, and clinicians face the burden of caring for patients at all stages of dementia for the foreseeable future. Those with advanced disease suffer neurological symptoms and signs that include incontinence; problems with gait and mobility; marked cognitive, language, and functional impairment; and in about 90% of patients, significant behavior problems. Dementia precludes the ability to initiate meaningful activities or social interactions. Whether patients are resident in the community or living in a nursing home, this composite reflects a highly complex medical and neuropsychiatric management challenge. Predictable medical conditions also must be addressed (i.e., those that accompany dementia, such as parkinsonism, and those that are prevalent in any aging population, such as hypertension). Clinicians can better address these problems with awareness of current treatment options. Placebo-controlled trials of some psychotropic agents have shown modest favorable effects on behavior problems. Use of acetylcholinesterase inhibitors (AChEIs) to treat cognitive impairment and secondary behavioral symptoms derives primarily from results of placebo-controlled clinical trials. Trials in patients with moderate to severe AD, outpatients as well as nursing home residents, show overall effects similar to those seen in outpatients with milder dementia. Treatment with AChEIs may delay institutional placement. Memantine has shown benefit in trials in moderate to severe dementia, although it is not yet approved in the United States. Emerging data have expanded physicians' ability to use pharmacotherapy in patients with advanced dementia. Physicians need to enact the principle that something can be done for our afflicted parents and grandparents. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
10. CAP--advancing the evaluation of preclinical Alzheimer disease treatments.
- Author
-
Reiman, Eric M., Langbaum, Jessica B., Tariot, Pierre N., Lopera, Francisco, Bateman, Randall J., Morris, John C., Sperling, Reisa A., Aisen, Paul S., Roses, Allen D., Welsh-Bohmer, Kathleen A., Carrillo, Maria C., and Weninger, Stacie
- Subjects
- *
ALZHEIMER'S disease , *BASAL ganglia diseases , *METHODOLOGY , *SCIENTIFIC knowledge , *SOCIOLOGY of knowledge , *ALZHEIMER'S disease diagnosis , *COGNITION disorders diagnosis , *NOOTROPIC agents , *CLINICAL trials , *COGNITION disorders , *COOPERATIVENESS , *DRUG design , *CLINICAL drug trials , *INTERPROFESSIONAL relations , *NEUROPSYCHOLOGICAL tests , *MEDICAL protocols , *TREATMENT effectiveness , *DISEASE progression , *EARLY diagnosis , *THERAPEUTICS - Abstract
If we are to find treatments to postpone, reduce the risk of, or completely prevent the clinical onset of Alzheimer disease (AD), we need faster methods to evaluate promising preclinical AD treatments, new ways to work together in support of common goals, and a determination to expedite the initiation and performance of preclinical AD trials. In this article, we note some of the current challenges, opportunities and emerging strategies in preclinical AD treatment. We describe the Collaboration for Alzheimer's Prevention (CAP)-a convening, harmonizing and consensus-building initiative to help stakeholders advance AD prevention research with rigour, care and maximal impact-and we demonstrate the impact of CAP on the goals and design of new preclinical AD trials. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
11. Pimavanserin for psychosis in Parkinson's disease dementia: Subgroup analysis of the HARMONY Trial.
- Author
-
Weintraub, Daniel, Espay, Alberto J., Sharma, Vibhash D., Tariot, Pierre N., Abler, Victor, Pathak, Sanjeev, and Stankovic, Srdjan
- Subjects
- *
PARKINSON'S disease , *PSYCHOSES , *DEMENTIA , *CLINICAL trials , *SUBGROUP analysis (Experimental design) , *VASCULAR dementia - Abstract
Pimavanserin is FDA-approved to treat Parkinson's disease (PD) psychosis. We analyzed the effect of pimavanserin on psychosis in the PD dementia (PDD) subgroup from the phase 3 HARMONY trial. This subgroup analysis included PDD patients enrolled in an international, multicenter, randomized discontinuation study of pimavanserin for dementia-related psychosis. PDD patients with moderate-to-severe psychosis, age 50–90 years, received pimavanserin 34 mg/day for 12 weeks (open-label period). Those with a sustained psychosis response to pimavanserin at weeks 8 and 12 were randomized during the double-blind period to continue pimavanserin or receive placebo. Primary efficacy endpoint was time to psychosis relapse as measured by the SAPS-H + D and CGI-I. Safety was assessed, as were effects on motor symptoms and cognitive abilities using the ESRS-A and MMSE. 392 patients were enrolled in HARMONY (mean age: 72.6 years; 38.8 % female): 59 had PDD; 49/59 remained on pimavanserin during the open-label period (safety analysis set), and 36/49 were randomized to pimavanserin (n = 16) or placebo (n = 20) in the double-blind phase (intent-to-treat analysis set). Risk of psychosis relapse was lower with pimavanserin 34 mg compared with placebo in the double-blind phase (HR = 0.052; 95 % CI 0.016–0.166; 1-sided nominal p < 0.001). During the open-label period, 46.9 % experienced a treatment-emergent adverse event; event incidence was similar across arms in the double-blind period. Pimavanserin did not adversely affect motor or cognitive function in either treatment phase. Pimavanserin significantly reduced risk of psychosis relapse in patients with PDD, was well tolerated, and did not worsen motor or cognitive function. • Pimavanserin is FDA-approved for hallucinations and delusions associated with PDP. • This subgroup analysis evaluated pimavanserin in patients with PDP + dementia. • Pimavanserin reduced the risk of psychosis relapse. • Treatment was well tolerated, and did not worsen motor or cognitive function. • These findings extend the known safety and efficacy of pimavanserin in PDP. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Randomized Trial of Verubecestat for Prodromal Alzheimer's Disease.
- Author
-
Egan, Michael F., Kost, James, Voss, Tiffini, Mukai, Yuki, Ying Zhang, Wen Li, Furtek, Christine, Mahoney, Erin, Mozley, Lyn Harper, Yi Mo, Sur, Cyrille, Michelson, David, Aisen, Paul S., Cummings, Jeffrey L., Tariot, Pierre N., Vellas, Bruno, van Dyck, Christopher H., Boada, Merce, Zhang, Ying, and Li, Wen
- Subjects
- *
ALZHEIMER'S disease , *ALZHEIMER'S patients , *ZYMOGENS , *ALZHEIMER'S disease prevention , *PEPTIDE analysis , *ANTHROPOMETRY , *BIOCHEMISTRY , *CLINICAL trials , *COMPARATIVE studies , *ENZYME inhibitors , *HIPPOCAMPUS (Brain) , *MAGNETIC resonance imaging , *PHENOMENOLOGY , *RESEARCH methodology , *MEDICAL cooperation , *PROTEIN precursors , *RESEARCH , *POSITRON emission tomography , *SULFUR compounds , *EVALUATION research , *RANDOMIZED controlled trials , *TREATMENT effectiveness , *BLIND experiment , *DISEASE progression , *EARLY diagnosis , *THERAPEUTICS - Abstract
Background: Prodromal Alzheimer's disease offers an opportunity to test the effect of drugs that modify the deposition of amyloid in the brain before the onset of dementia. Verubecestat is an orally administered β-site amyloid precursor protein-cleaving enzyme 1 (BACE-1) inhibitor that blocks production of amyloid-beta (Aβ). The drug did not prevent clinical progression in a trial involving patients with mild-to-moderate dementia due to Alzheimer's disease.Methods: We conducted a randomized, double-blind, placebo-controlled, 104-week trial to evaluate verubecestat at doses of 12 mg and 40 mg per day, as compared with placebo, in patients who had memory impairment and elevated brain amyloid levels but whose condition did not meet the case definition of dementia. The primary outcome was the change from baseline to week 104 in the score on the Clinical Dementia Rating Scale-Sum of Boxes (CDR-SB; scores range from 0 to 18, with higher scores indicating worse cognition and daily function). Secondary outcomes included other assessments of cognition and daily function.Results: The trial was terminated for futility after 1454 patients had been enrolled; 485 had been assigned to receive verubecestat at a dose of 12 mg per day (the 12-mg group), 484 to receive verubecestat at a dose of 40 mg per day (the 40-mg group), and 485 to receive placebo. A total of 234 patients, 231 patients, and 239 patients per group, respectively, completed 104 weeks of the trial regimen. The estimated mean change from baseline to week 104 in the CDR-SB score was 1.65 in the 12-mg group, 2.02 in the 40-mg group, and 1.58 in the placebo group (P = 0.67 for the comparison between the 12-mg group and the placebo group and P = 0.01 for the comparison between the 40-mg group and the placebo group), suggesting a worse outcome in the higher-dose group than in the placebo group. The estimated rate of progression to dementia due to Alzheimer's disease was 24.5, 25.5, and 19.3 events per 100 patient-years in the 12-mg group, the 40-mg group, and the placebo group, respectively (hazard ratio for 40 mg vs. placebo, 1.38; 97.51% confidence interval, 1.07 to 1.79, not adjusted for multiple comparisons), favoring placebo. Adverse events were more common in the verubecestat groups than in the placebo group.Conclusions: Verubecestat did not improve clinical ratings of dementia among patients with prodromal Alzheimer's disease, and some measures suggested that cognition and daily function were worse among patients who received verubecestat than among those who received placebo. (Funded by Merck Sharp & Dohme; ClinicalTrials.gov number, NCT01953601.). [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.