Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Alzheimers Dement ; 18(5): 988-1007, 2022 05.
Article in English | MEDLINE | ID: mdl-34581500

ABSTRACT

Studies supporting a strong association between tau deposition and neuronal loss, neurodegeneration, and cognitive decline have heightened the allure of tau and tau-related mechanisms as therapeutic targets. In February 2020, leading tau experts from around the world convened for the first-ever Tau2020 Global Conference in Washington, DC, co-organized and cosponsored by the Rainwater Charitable Foundation, the Alzheimer's Association, and CurePSP. Representing academia, industry, government, and the philanthropic sector, presenters and attendees discussed recent advances and current directions in tau research. The meeting provided a unique opportunity to move tau research forward by fostering global partnerships among academia, industry, and other stakeholders and by providing support for new drug discovery programs, groundbreaking research, and emerging tau researchers. The meeting also provided an opportunity for experts to present critical research-advancing tools and insights that are now rapidly accelerating the pace of tau research.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Biomarkers , Drug Discovery , Humans , tau Proteins
2.
J Alzheimers Dis ; 67(2): 685-705, 2019.
Article in English | MEDLINE | ID: mdl-30689585

ABSTRACT

BACKGROUND: Little is known with respect to behavioral markers of subjective cognitive decline (SCD), a condition initially described in association with Global Deterioration Scale (GDS) stage 2. OBJECTIVE: Two-year interval behavioral markers were investigated herein. METHODS: Subjects from a published 7-year outcome study of GDS stage 2 subjects were selected. This study had demonstrated a hazard ratio of 4.5 for progression of GDS stage 2, in comparison with GDS stage 1 (no subjective or objective cognitive decline) subjects, after controlling for demographic and temporal variables. Because GDS 2 subjects have previously demonstrated impairment in comparison with healthy persons free of complaints, we herein suggest the terminology "SCD(I)" for these persons. 98 SCD(I) persons, 63 women and 35 men, mean baseline age, 67.12±8.75 years, with a mean educational background of 15.55±2.60 years, and mean baseline MMSE scores of 28.9±1.24 were followed for 2.13±0.30 years. RESULTS: Observed annual decline on the GDS was 6.701% per annum, very close to a 1986 published estimate. At follow up, the MMSE, and 7 of 8 psychometric tests did not decline significantly. Of 21 Hamilton Depression Scale items, 2 improved and the remainder were unchanged. Anxieties declined from multiple perspectives. The Brief Cognitive Rating Scale (BCRS) declined significantly (p < 0.001), with component declines in Remote memory (p < 0.01), and Functioning/self-care (p = 0.01). CONCLUSION: SCD(I) persons decline at an annual rate of approximately 6.7% /year from several recent studies. The BCRS assessments and the Digit Symbol Substitution Test can be sensitive measures for future studies of progression mitigation.


Subject(s)
Behavior , Cognition Disorders/psychology , Cognitive Dysfunction/psychology , Neuropsychological Tests , Affect , Aged , Anxiety/complications , Anxiety/psychology , Biomarkers , Depression/complications , Depression/psychology , Disease Progression , Female , Humans , Longitudinal Studies , Male , Mental Status and Dementia Tests , Middle Aged , Psychometrics , Self Care , Treatment Outcome
3.
Alzheimers Dement ; 15(3): 465-476, 2019 03.
Article in English | MEDLINE | ID: mdl-30555032

ABSTRACT

INTRODUCTION: In this multicenter study on subjective cognitive decline (SCD) in community-based and memory clinic settings, we assessed the (1) incidence of Alzheimer's disease (AD) and non-AD dementia and (2) determinants of progression to dementia. METHODS: Eleven cohorts provided 2978 participants with SCD and 1391 controls. We estimated dementia incidence and identified risk factors using Cox proportional hazards models. RESULTS: In SCD, incidence of dementia was 17.7 (95% Poisson confidence interval 15.2-20.3)/1000 person-years (AD: 11.5 [9.6-13.7], non-AD: 6.1 [4.7-7.7]), compared with 14.2 (11.3-17.6) in controls (AD: 10.1 [7.7-13.0], non-AD: 4.1 [2.6-6.0]). The risk of dementia was strongly increased in SCD in a memory clinic setting but less so in a community-based setting. In addition, higher age (hazard ratio 1.1 [95% confidence interval 1.1-1.1]), lower Mini-Mental State Examination (0.7 [0.66-0.8]), and apolipoprotein E ε4 (1.8 [1.3-2.5]) increased the risk of dementia. DISCUSSION: SCD can precede both AD and non-AD dementia. Despite their younger age, individuals with SCD in a memory clinic setting have a higher risk of dementia than those in community-based cohorts.


Subject(s)
Cognitive Dysfunction/epidemiology , Dementia/epidemiology , Aged , Aged, 80 and over , Cognitive Dysfunction/psychology , Cohort Studies , Dementia/psychology , Diagnostic Self Evaluation , Disease Progression , Female , Humans , Incidence , Male , Middle Aged , Risk Factors , Self Concept
4.
Alzheimers Dement ; 10(6): 844-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24798886

ABSTRACT

There is increasing evidence that subjective cognitive decline (SCD) in individuals with unimpaired performance on cognitive tests may represent the first symptomatic manifestation of Alzheimer's disease (AD). The research on SCD in early AD, however, is limited by the absence of common standards. The working group of the Subjective Cognitive Decline Initiative (SCD-I) addressed this deficiency by reaching consensus on terminology and on a conceptual framework for research on SCD in AD. In this publication, research criteria for SCD in pre-mild cognitive impairment (MCI) are presented. In addition, a list of core features proposed for reporting in SCD studies is provided, which will enable comparability of research across different settings. Finally, a set of features is presented, which in accordance with current knowledge, increases the likelihood of the presence of preclinical AD in individuals with SCD. This list is referred to as SCD plus.


Subject(s)
Alzheimer Disease/physiopathology , Cognition Disorders/diagnosis , Cognition Disorders/physiopathology , Disease Progression , Prodromal Symptoms , Age of Onset , Female , Humans , Male , Neuropsychological Tests , Psychiatric Status Rating Scales , Terminology as Topic
5.
Dement Geriatr Cogn Disord ; 38(1-2): 89-146, 2014.
Article in English | MEDLINE | ID: mdl-24714384

ABSTRACT

BACKGROUND: Behavioral and psychological symptoms of dementia (BPSD) and associated disturbances in Alzheimer's disease (AD) are a source of distress and burden for spouses, professional caregivers, and others with responsibilities for the care of individuals with AD. BPSD with behavioral disturbances are also associated with more rapid institutionalization and increased morbidity and mortality for persons with AD. OBJECTIVES: In this review and commentary, we discuss the history of the development of BPSD and behavioral disturbance assessments, which are distinct from those evaluating cognitive and functional symptoms of AD. In particular, we review the informant-based Behavioral Pathology in Alzheimer's Disease Rating Scale (BEHAVE-AD), the related, potentially more sensitive, BEHAVE-AD Frequency-Weighted Severity Scale (BEHAVE-AD-FW), and the direct subject evaluation-based Empirical BEHAVE-AD Rating Scale (E-BEHAVE-AD). The kinds of medications that alleviate behavioral symptoms on these measures as well as the problems and possibilities for further advances with these medications are discussed. Finally, the importance of distinguishing BPSD and behavioral disturbance remediation in AD from the treatment of cognitive decline and other aspects of AD is emphasized in the context of appropriate assessment methodology. The objective of this paper is to provide a framework for further advances in the treatment of BPSD and associated behavioral disturbances in AD and, consequently, a framework for continuing improvements in the lives of individuals with AD and those who share the burden of the disease with the AD person.


Subject(s)
Alzheimer Disease , Behavioral Symptoms , Cost of Illness , Dementia , Psychotropic Drugs/therapeutic use , Symptom Assessment/methods , Alzheimer Disease/diagnosis , Alzheimer Disease/psychology , Alzheimer Disease/therapy , Behavioral Symptoms/diagnosis , Behavioral Symptoms/therapy , Dementia/diagnosis , Dementia/psychology , Dementia/therapy , Disease Management , Humans , Neuropsychological Tests , Patient Outcome Assessment , Psychiatric Status Rating Scales
6.
Neurology ; 81(13): 1114-21, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-23966249

ABSTRACT

OBJECTIVE: To inform whether the Alzheimer's Disease Neuroimaging Initiative (ADNI) should change its policy of not returning research results to ADNI participants, we surveyed investigators and research staff about disclosing ADNI biomarker information to research participants, with particular emphasis on amyloid imaging results. METHODS: In April 2012, just before Food and Drug Administration approval of the amyloid-binding radiotracer, florbetapir, all ADNI investigators and personnel were recruited to complete an anonymous online survey that contained fixed choice and free-text questions. RESULTS: Although ADNI participants often requested amyloid imaging results (the proportions of investigators who reported requests from more than half of their participants with normal cognition or mild cognitive impairment were 20% and 22%, respectively), across all diagnostic groups, the majority of ADNI investigators (approximately 90%) did not return amyloid imaging results to ADNI participants. However, the majority of investigators reported that, if the Food and Drug Administration approved florbetapir, they would support the return of amyloid imaging results to participants with mild cognitive impairment and normal cognition, but they emphasized the need for guidance on how to provide these results to participants and for research to assess the value of returning results as well as how returning results will affect study validity and participant well-being. CONCLUSIONS: A majority of ADNI investigators support returning amyloid imaging results to ADNI participants. The findings that they want guidance on how to do this and research on the impact of disclosure suggest how to develop and monitor a disclosure process.


Subject(s)
Alzheimer Disease/diagnosis , Attitude , Biomarkers/metabolism , Brain/metabolism , Neuroimaging , Research Personnel/psychology , Alzheimer Disease/complications , Alzheimer Disease/diagnostic imaging , Aniline Compounds , Brain/diagnostic imaging , Brain/pathology , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/etiology , Educational Status , Ethylene Glycols , Female , Fluorine Radioisotopes , Health Surveys , Humans , Male , Neuroimaging/psychology , Radionuclide Imaging , United States , United States Food and Drug Administration
7.
Alzheimers Dement ; 6(1): 11-24, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20129317

ABSTRACT

BACKGROUND: Subjective cognitive impairment (SCI) in older persons without manifest symptomatology is a common condition with a largely unclear prognosis. We hypothesized that (1) examining outcome for a sufficient period by using conversion to mild cognitive impairment (MCI) or dementia would clarify SCI prognosis, and (2) with the aforementioned procedures, the prognosis of SCI subjects would differ significantly from that of demographically matched healthy subjects, free of SCI, termed no cognitive impairment (NCI) subjects. METHODS: A consecutive series of healthy subjects, aged > or =40 years, presenting with NCI or SCI to a brain aging and dementia research center during a 14-year interval, were studied and followed up during an 18-year observation window. The study population (60 NCI, 200 SCI, 60% female) had a mean age of 67.2 +/- 9.1 years, was well-educated (mean, 15.5 +/- 2.7 years), and cognitively normal (Mini-Mental State Examination, 29.1 +/- 1.2). RESULTS: A total of 213 subjects (81.9% of the study population) were followed up. Follow-up occurred during a mean period of 6.8 +/- 3.4 years, and subjects had a mean of 2.9 +/- 1.6 follow-up visits. Seven NCI (14.9%) and 90 SCI (54.2%) subjects declined (P < .0001). Of NCI decliners, five declined to MCI and two to probable Alzheimer's disease. Of SCI decliners, 71 declined to MCI and 19 to dementia diagnoses. Controlling for baseline demographic variables and follow-up time, Weibull proportional hazards model revealed increased decline in SCI subjects (hazard ratio, 4.5; 95% confidence interval, 1.9-10.3), whereas the accelerated failure time model analysis with an underlying Weibull survival function showed that SCI subjects declined more rapidly, at 60% of the rate of NCI subjects (95% confidence interval, 0.45-0.80). Furthermore, mean time to decline was 3.5 years longer for NCI than for SCI subjects (P = .0003). CONCLUSIONS: These results indicate that SCI in subjects with normal cognition is a harbinger of further decline in most subjects during a 7-year mean follow-up interval. Relevance for community populations should be investigated, and prevention studies in this at-risk population should be explored.


Subject(s)
Cognition Disorders/diagnosis , Cognition Disorders/epidemiology , Geriatric Assessment , Age Factors , Aged , Disability Evaluation , Disease Progression , Female , Humans , Longitudinal Studies , Male , Middle Aged , Neuropsychological Tests , Predictive Value of Tests , Psychiatric Status Rating Scales , Retrospective Studies
10.
Epilepsy Behav ; 3(5S): 30-34, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12609318

ABSTRACT

Impaired memory is a common and often debilitating complaint in patients with epilepsy. Overlapping variables such as seizure control, attentional dysfunction, and mood disorders further complicate diagnosis and management. Direct therapy for memory deficits associated with epilepsy is rarely attempted. The varied pharmacological (AED selection, cholinesterase inhibitors, stimulants, antidepressants, and herbal supplements) and nonpharmacological approaches to cognitive remediation in epilepsy patients are reviewed.

11.
Epilepsy Behav ; 3(4): 393-394, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12609339
12.
Epilepsy Behav ; 1(6): 384-395, 2000 Dec.
Article in English | MEDLINE | ID: mdl-12737828

ABSTRACT

The frontal lobes have been overshadowed by the temporal lobes in the vast literature addressing the neurobehavioral and psychological perspectives of epilepsy. The purpose of this review is to summarize contemporary anatomicobehavioral correlations and to highlight the frontal lobe contributions to the neurology, neuropsychology, and neuropsychiatry of epilepsy, in general, and to temporal lobe epilepsy (TLE) and frontal lobe epilepsy (FLE), in particular. Much evidence has accumulated suggesting that focal epileptogenic tissue may have effects on distant neural systems. Data supporting the case that the frontal regions are preferentially affected in TLE are presented. Emphasis is placed on the results of numerous functional imaging studies demonstrating correlations between frontal hypoperfusion and cognitive or mood impairments in patients with TLE.

SELECTION OF CITATIONS
SEARCH DETAIL
...