51
|
Peter J, Schumacher LV, Landerer V, Abdulkadir A, Kaller CP, Lahr J, Klöppel S. Biological Factors Contributing to the Response to Cognitive Training in Mild Cognitive Impairment. J Alzheimers Dis 2018; 61:333-345. [PMID: 29154279 PMCID: PMC5734129 DOI: 10.3233/jad-170580] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In mild cognitive impairment (MCI), small benefits from cognitive training were observed for memory functions but there appears to be great variability in the response to treatment. Our study aimed to improve the characterization and selection of those participants who will benefit from cognitive intervention. We evaluated the predictive value of disease-specific biological factors for the outcome after cognitive training in MCI (n = 25) and also considered motivation of the participants. We compared the results of the cognitive intervention group with two independent control groups of MCI patients (local memory clinic, n = 20; ADNI cohort, n = 302). The primary outcome measure was episodic memory as measured by verbal delayed recall of a 10-word list. Episodic memory remained stable after treatment and slightly increased 6 months after the intervention. In contrast, in MCI patients who did not receive an intervention, episodic memory significantly decreased during the same time interval. A larger left entorhinal cortex predicted more improvement in episodic memory after treatment and so did higher levels of motivation. Adding disease-specific biological factors significantly improved the prediction of training-related change compared to a model based simply on age and baseline performance. Bootstrapping with resampling (n = 1000) verified the stability of our finding. Cognitive training might be particularly helpful in individuals with a bigger left entorhinal cortex as individuals who did not benefit from intervention showed 17% less volume in this area. When extended to alternative treatment options, stratification based on disease-specific biological factors is a useful step towards individualized medicine.
Collapse
Affiliation(s)
- Jessica Peter
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Switzerland.,Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Freiburg, Germany
| | - Lena V Schumacher
- Medical Psychology and Medical Sociology, Faculty of Medicine, University of Freiburg, Germany
| | - Verena Landerer
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Freiburg, Germany
| | - Ahmed Abdulkadir
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Switzerland.,Department of Computer Science, University of Freiburg, Germany
| | - Christoph P Kaller
- Department of Neurology, Faculty of Medicine, University of Freiburg, Germany.,BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Freiburg, Germany
| | - Jacob Lahr
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Freiburg, Germany
| | - Stefan Klöppel
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Switzerland.,Centre for Geriatric Medicine and Gerontology, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
52
|
Harrison JE. Cognition comes of age: comments on the new FDA draft guidance for early Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2018; 10:61. [PMID: 29958538 PMCID: PMC6026341 DOI: 10.1186/s13195-018-0386-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background The FDA have recently published draft guidance for the development of treatments for early Alzheimer’s disease. Key features of this guidance are the advocacy of sensitive cognitive measures and a taxonomy of disease severity. Whilst desirable patterns of cognitive-functional improvement are included, specific measures, and the magnitude of required effects, are not described. Main section We describe key elements of the guidance content, especially with regard targeting key cognitive domains and the means by which they might be efficiently indexed in the disease stages included in the guidance. We discuss also the opportunities to assess cognitive performance in ‘Stage 2’ and ‘Stage 3’ patients, as well as the possibilities for effectively assessing function in the latter category. In this section we review candidate cognitive assessments that we judge are capable of delivering on the guidance specification for sensitive neuropsychological measures. This includes detailed consideration of the ADCS-PACC and Catch-Cog initiatives. With respect to the magnitude of effects, we propose that standardised effect sizes of 0.3 represent a reasonable level of efficacy based on the observation that already marketed drugs on average deliver this level of improvement. Conclusions We propose the use of cognitive measures in stage 2 patients to index the cognitive skills known to be compromised early in the Alzheimer’s disease process. We recommend extending the traditional interest in episodic memory to include sensitive, reliable and valid measures of attention, working memory and aspects of executive function. We propose a focus on these additional cognitive abilities based on evidence that performance on tests of these domains is moderately well related to functional skills.
Collapse
Affiliation(s)
- John E Harrison
- Metis Cognition Ltd., Kilmington Common, Wiltshire, BA12 6QY, UK. .,Alzheimer Center, Amsterdam, The Netherlands. .,IoPPN, King's College London, London, UK.
| |
Collapse
|
53
|
Sparrow G, Hurd R, Carlson R, Molina A. Exploring the effects of galantamine paired with meditation and dream reliving on recalled dreams: Toward an integrated protocol for lucid dream induction and nightmare resolution. Conscious Cogn 2018; 63:74-88. [PMID: 29960246 DOI: 10.1016/j.concog.2018.05.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 10/28/2022]
Abstract
An experimental home study examined the impact of a pre-sleep protocol for enhancing self-awareness, lucidity, and responsiveness in dreams. It included ingesting the cholinesterase inhibitor galantamine--which is widely reported to increase the frequency of lucid dreaming--prior to engaging in middle-of-the-night meditation and the imaginary reliving of a distressing dream while exercising new responses. Thirty-five participants completed an eight-night study, which included pre- and post-baseline nights and six conditions: waking for 40 min before returning to bed, called Wake-Back-to-Bed (WBTB); Wake-Back-to-Bed plus placebo (WBTB + P); Wake-Back-to-Bed plus galantamine (WBTB + G); meditation and dream reliving (MDR); meditation and dream reliving plus placebo (MDR + P); and meditation and dream reliving plus galantamine (MDR + G). The outcome measures included lucidity, reflectiveness, interactive behavior, role change, constructive action, and fear and threat, as measured by the participants' self-ratings. The results support the use of this protocol in further studies of lucid dream induction and nightmare/trauma resolution.
Collapse
Affiliation(s)
- Gregory Sparrow
- Dept. of Counseling, University of Texas Rio Grande Valley, 1201 W. University Dr., Edinburg, TX 78539, United States.
| | - Ryan Hurd
- Dept. of Psychology, John F. Kennedy University, Orinda, CA, United States
| | - Ralph Carlson
- Dept. of Educational Psychology, 1201 W. University Dr., University of Texas Rio Grande Valley, Edinburg, TX 78539, United States
| | - Ana Molina
- Dept. of Educational Psychology, 1201 W. University Dr., University of Texas Rio Grande Valley, Edinburg, TX 78539, United States
| |
Collapse
|
54
|
Ilhan Algin D, Dagli Atalay S, Ozkan S, Ozbabalik Adapinar D, Ak Sivrioz I. Memantine improves semantic memory in patients with amnestic mild cognitive impairment: A single-photon emission computed tomography study. J Int Med Res 2017; 45:2053-2064. [PMID: 28661262 PMCID: PMC5805216 DOI: 10.1177/0300060517715166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 05/23/2017] [Indexed: 12/20/2022] Open
Abstract
Objective This study was performed to assess the efficacy of memantine in patients with amnestic mild cognitive impairment (aMCI). Methods Thirty healthy controls and 45 patients diagnosed with aMCI based on the Petersen criteria were classified into 3 groups. Group 1 comprised patients who received a single memantine dose following examination (n = 25), Group 2 comprised patients who did not receive memantine treatment following examination (n = 20), and Group 3 comprised healthy age-matched volunteers (n = 30). Neuropsychological testing was performed, and the response to memantine was examined at baseline and at 12, 24, and 48 weeks. Single-photon emission computed tomography was performed at baseline and at 48 weeks in patients who received memantine treatment. Results Memantine treatment significantly improved the symptoms of aMCI according to the Wechsler Adult Intelligence Scale-Revised vocabulary subtest, backward digit span, and Blessed Dementia Rating Scale, all of which were recorded for the duration of the study. Conclusion These data indicate that patients with aMCI receiving memantine develop an improved semantic memory compared with no treatment. Further studies including larger patient cohorts are necessary to validate these findings.
Collapse
Affiliation(s)
- Demet Ilhan Algin
- Assistant Professor, Eskişehir Osmangazi
University Faculty of Medicine Department of Neurology, Eskişehir, Turkey
| | - Suna Dagli Atalay
- Assistant Professor, Eskişehir Osmangazi
University Faculty of Medicine Department of Neurology, Eskişehir, Turkey
| | - Serhat Ozkan
- Assistant Professor, Eskişehir Osmangazi
University Faculty of Medicine Department of Neurology, Eskişehir, Turkey
| | - Demet Ozbabalik Adapinar
- Assistant Professor, Eskişehir Osmangazi
University Faculty of Medicine Department of Neurology, Eskişehir, Turkey
| | - Ilknur Ak Sivrioz
- Professor, Eskişehir Osmangazi
University Faculty of Medicine Department of Nuclear Medicine, Eskişehir,
Turkey
| |
Collapse
|
55
|
Savulich G, Piercy T, Fox C, Suckling J, Rowe JB, O'Brien JT, Sahakian BJ. Cognitive Training Using a Novel Memory Game on an iPad in Patients with Amnestic Mild Cognitive Impairment (aMCI). Int J Neuropsychopharmacol 2017; 20:624-633. [PMID: 28898959 PMCID: PMC5569993 DOI: 10.1093/ijnp/pyx040] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/12/2017] [Accepted: 05/19/2017] [Indexed: 11/14/2022] Open
Abstract
Background Cognitive training is effective in patients with mild cognitive impairment but does not typically address the motivational deficits associated with older populations with memory difficulties. Methods We conducted a randomized controlled trial of cognitive training using a novel memory game on an iPad in 42 patients with a diagnosis of amnestic mild cognitive impairment assigned to either the cognitive training (n=21; 8 hours of gameplay over 4 weeks) or control (n=21; clinic visits as usual) groups. Results Significant time-by-pattern-by-group interactions were found for cognitive performance in terms of the number of errors made and trials needed on the Cambridge Neuropsychological Test Automated Battery Paired Associates Learning task (P=.044; P=.027). Significant time-by-group interactions were also found for the Cambridge Neuropsychological Test Automated Battery Paired Associates Learning first trial memory score (P=.002), Mini-Mental State Examination (P=.036), the Brief Visuospatial Memory Test (P=.032), and the Apathy Evaluation Scale (P=.026). Within-group comparisons revealed highly specific effects of cognitive training on episodic memory. The cognitive training group maintained high levels of enjoyment and motivation to continue after each hour of gameplay, with self-confidence and self-rated memory ability improving over time. Conclusions Episodic memory robustly improved in the cognitive training group. "Gamified" cognitive training may also enhance visuospatial abilities in patients with amnestic mild cognitive impairment. Gamification maximizes engagement with cognitive training by increasing motivation and could complement pharmacological treatments for amnestic mild cognitive impairment and mild Alzheimer's disease. Larger, more controlled trials are needed to replicate and extend these findings.
Collapse
Affiliation(s)
- George Savulich
- Department of Psychiatry (Dr Savulich, Mr Piercy, and Profs Suckling, O'Brien, and Sahakian), and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute (Dr Savulich, Mr Piercy, and Profs Suckling, Rowe, and Sahakian) University of Cambridge, Cambridge, United Kingdom; Department of Psychological Sciences, University of East Anglia, Norwich, United Kingdom (Prof Fox); Department of Clinical Neurosciences and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom (Prof Rowe)
| | - Thomas Piercy
- Department of Psychiatry (Dr Savulich, Mr Piercy, and Profs Suckling, O'Brien, and Sahakian), and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute (Dr Savulich, Mr Piercy, and Profs Suckling, Rowe, and Sahakian) University of Cambridge, Cambridge, United Kingdom; Department of Psychological Sciences, University of East Anglia, Norwich, United Kingdom (Prof Fox); Department of Clinical Neurosciences and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom (Prof Rowe)
| | - Chris Fox
- Department of Psychiatry (Dr Savulich, Mr Piercy, and Profs Suckling, O'Brien, and Sahakian), and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute (Dr Savulich, Mr Piercy, and Profs Suckling, Rowe, and Sahakian) University of Cambridge, Cambridge, United Kingdom; Department of Psychological Sciences, University of East Anglia, Norwich, United Kingdom (Prof Fox); Department of Clinical Neurosciences and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom (Prof Rowe)
| | - John Suckling
- Department of Psychiatry (Dr Savulich, Mr Piercy, and Profs Suckling, O'Brien, and Sahakian), and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute (Dr Savulich, Mr Piercy, and Profs Suckling, Rowe, and Sahakian) University of Cambridge, Cambridge, United Kingdom; Department of Psychological Sciences, University of East Anglia, Norwich, United Kingdom (Prof Fox); Department of Clinical Neurosciences and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom (Prof Rowe)
| | - James B Rowe
- Department of Psychiatry (Dr Savulich, Mr Piercy, and Profs Suckling, O'Brien, and Sahakian), and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute (Dr Savulich, Mr Piercy, and Profs Suckling, Rowe, and Sahakian) University of Cambridge, Cambridge, United Kingdom; Department of Psychological Sciences, University of East Anglia, Norwich, United Kingdom (Prof Fox); Department of Clinical Neurosciences and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom (Prof Rowe)
| | - John T O'Brien
- Department of Psychiatry (Dr Savulich, Mr Piercy, and Profs Suckling, O'Brien, and Sahakian), and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute (Dr Savulich, Mr Piercy, and Profs Suckling, Rowe, and Sahakian) University of Cambridge, Cambridge, United Kingdom; Department of Psychological Sciences, University of East Anglia, Norwich, United Kingdom (Prof Fox); Department of Clinical Neurosciences and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom (Prof Rowe)
| | - Barbara J Sahakian
- Department of Psychiatry (Dr Savulich, Mr Piercy, and Profs Suckling, O'Brien, and Sahakian), and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute (Dr Savulich, Mr Piercy, and Profs Suckling, Rowe, and Sahakian) University of Cambridge, Cambridge, United Kingdom; Department of Psychological Sciences, University of East Anglia, Norwich, United Kingdom (Prof Fox); Department of Clinical Neurosciences and MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom (Prof Rowe)
| |
Collapse
|
56
|
Bordet R, Ihl R, Korczyn AD, Lanza G, Jansa J, Hoerr R, Guekht A. Towards the concept of disease-modifier in post-stroke or vascular cognitive impairment: a consensus report. BMC Med 2017; 15:107. [PMID: 28539119 PMCID: PMC5444106 DOI: 10.1186/s12916-017-0869-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 05/06/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Vascular cognitive impairment (VCI) is a complex spectrum encompassing post-stroke cognitive impairment (PSCI) and small vessel disease-related cognitive impairment. Despite the growing health, social, and economic burden of VCI, to date, no specific treatment is available, prompting the introduction of the concept of a disease modifier. CONSENSUS AND SUGGESTIONS Within this clinical spectrum, VCI and PSCI remain advancing conditions as neurodegenerative diseases with progression of both vascular and degenerative lesions accounting for cognitive decline. Disease-modifying strategies should integrate both pharmacological and non-pharmacological multimodal approaches, with pleiotropic effects targeting (1) endothelial and brain-blood barrier dysfunction; (2) neuronal death and axonal loss; (3) cerebral plasticity and compensatory mechanisms; and (4) degenerative-related protein misfolding. Moreover, pharmacological and non-pharmacological treatment in PSCI or VCI requires valid study designs clearly stating the definition of basic methodological issues, such as the instruments that should be used to measure eventual changes, the biomarker-based stratification of participants to be investigated, and statistical tests, as well as the inclusion and exclusion criteria that should be applied. CONCLUSION A consensus emerged to propose the development of a disease-modifying strategy in VCI and PSCI based on pleiotropic pharmacological and non-pharmacological approaches.
Collapse
Affiliation(s)
- Régis Bordet
- University of Lille, Inserm, CHU, U1171 'Degenerative and vascular cognitive disorders', Lille, France.
- Département de Pharmacologie Médicale, Faculté de Médecine, 1 place Verdun, 59045, Lille Cedex, France.
| | - Ralf Ihl
- University of Duesseldorf, Alexian Research Center, Krefeld, Germany
| | - Amos D Korczyn
- Department of Neurology, Tel Aviv University, Ramat Aviv, Israel
| | - Giuseppe Lanza
- Department of Neurology IC, Oasi Institute for Research on Mental Retardation and Brain Aging (IRCCS), Troina, Italy
| | - Jelka Jansa
- University Medical Centre Ljubljana, Neurologic Hospital, Neurorehabilitation Unit, Ljubljana, Slovenia
| | - Robert Hoerr
- Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany
| | - Alla Guekht
- Department of Neurology, Neurosurgery and Genetics, Russian National Research Medical University, Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
57
|
Tian J, Shi J, Li T, Li L, Wang Z, Li X, Lv Z, Zheng Q, Wei M, Wang Y. Efficacy and Safety of an Herbal Therapy in Patients with Amnestic Mild Cognitive Impairment: A 24-Week Randomized Phase III Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2017; 2017:4251747. [PMID: 28596794 PMCID: PMC5449752 DOI: 10.1155/2017/4251747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/30/2017] [Accepted: 04/16/2017] [Indexed: 11/17/2022]
Abstract
OBJECTIVE In the 24-week randomized, double-blind, double-placebo, parallel-controlled trial, we aimed to test the effects of herbal therapy with amnestic mild cognitive impairment (aMCI). METHODS A total of 324 patients with aMCI entered a 2-week placebo run-in period followed by 24 weeks' treatment of either (a) herbal capsule (5 shenwu capsules/administration, 3 times/day) and placebo identical to donepezil tablets (n = 216) or (b) donepezil (5 mg/day) and placebo identical to herbal capsule (n = 108). RESULTS Herbal therapy showed a significant improvement on the primary efficacy measure, measured by Alzheimer Disease Assessment Scale-cognitive subscale (ADAS-cog), and showed a mean decrease from baseline of 4.23 points at the endpoint, without a significant difference from the donepezil group. Secondary efficacy measurement of the Logical Memory II Delayed Story Recall subtest (DSR) showed modest improvement in those taking herbal capsule compared to baseline, and there was no significant difference from donepezil group. The frequency of adverse events was much less in the herbal therapy group than the donepezil. CONCLUSION Herbal therapy demonstrated a significant improvement in cognition and memory, which were similar to the donepezil in patients with aMCI. Herbal therapy was safe and well tolerated. TRIAL REGISTRATION This study is registered with clinicaltrials.gov NCT01451749.
Collapse
Affiliation(s)
- Jinzhou Tian
- Third Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jing Shi
- Third Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Tao Li
- Department of Neurology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lin Li
- Laboratory of Pharmacy, Xuanwu Hospital, Capital University of Medical Sciences, Beijing 100053, China
| | - Zhiliang Wang
- North China Pharmaceutical Group Corporation, Hebei 050015, China
| | - Xiaobin Li
- North China Pharmaceutical Group Corporation, Hebei 050015, China
| | - Zhu Lv
- North China Pharmaceutical Group Corporation, Hebei 050015, China
| | - Qingshan Zheng
- Drug Clinical Trial Centre, Shanghai University of Chinese Medicine, Shanghai 200032, China
| | - Mingqing Wei
- Third Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yongyan Wang
- Institute of Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
58
|
Petersen RC, Thomas RG, Aisen PS, Mohs RC, Carrillo MC, Albert MS. Randomized controlled trials in mild cognitive impairment: Sources of variability. Neurology 2017; 88:1751-1758. [PMID: 28381516 PMCID: PMC5409843 DOI: 10.1212/wnl.0000000000003907] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 01/05/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To examine the variability in performance among placebo groups in randomized controlled trials for mild cognitive impairment (MCI). METHODS Placebo group data were obtained from 2 National Institute on Aging (NIA) MCI randomized controlled trials, the Alzheimer's Disease Cooperative Study (ADCS) MCI trial and the Alzheimer's Disease Neuroimaging Initiative (ADNI), which is a simulated clinical trial, in addition to industry-sponsored clinical trials involving rivastigmine, galantamine, rofecoxib, and donepezil. The data were collated for common measurement instruments. The performance of the placebo participants from these studies was tracked on the Alzheimer's Disease Assessment Scale-cognitive subscale, Mini-Mental State Examination, and Clinical Dementia Rating-sum of boxes, and for progression on these measures to prespecified clinical study endpoints. APOE status, where available, was also analyzed for its effects. RESULTS The progression to clinical endpoints varied a great deal among the trials. The expected performances were seen for the participants in the 2 NIA trials, ADCS and ADNI, with generally worsening of performance over time; however, the industry-sponsored trials largely showed stable or improved performance in their placebo participants. APOE4 carrier status influenced results in an expected fashion on the study outcomes, including rates of progression and cognitive subscales. CONCLUSIONS In spite of apparently similar criteria for MCI being adopted by the 7 studies, the implementation of the criteria varied a great deal. Several explanations including instruments used to characterize participants and variability among study populations contributed to the findings.
Collapse
Affiliation(s)
- Ronald C Petersen
- From the Department of Neurology (R.C.P.), Mayo Clinic College of Medicine, Rochester, MN; Department of Neurosciences (R.G.T.), University of California, San Diego, La Jolla; University of Southern California (P.S.A.), Los Angeles; Eli Lilly and Company (R.C.M.), Indianapolis, IN; Division of Medical & Scientific Relations (M.C.C.), Alzheimer's Association, Chicago, IL; and Johns Hopkins School of Medicine (M.S.A.), Baltimore, MD. R.C.M. is currently with Global Alzheimer's Platform Foundation, Washington, DC.
| | - Ronald G Thomas
- From the Department of Neurology (R.C.P.), Mayo Clinic College of Medicine, Rochester, MN; Department of Neurosciences (R.G.T.), University of California, San Diego, La Jolla; University of Southern California (P.S.A.), Los Angeles; Eli Lilly and Company (R.C.M.), Indianapolis, IN; Division of Medical & Scientific Relations (M.C.C.), Alzheimer's Association, Chicago, IL; and Johns Hopkins School of Medicine (M.S.A.), Baltimore, MD. R.C.M. is currently with Global Alzheimer's Platform Foundation, Washington, DC
| | - Paul S Aisen
- From the Department of Neurology (R.C.P.), Mayo Clinic College of Medicine, Rochester, MN; Department of Neurosciences (R.G.T.), University of California, San Diego, La Jolla; University of Southern California (P.S.A.), Los Angeles; Eli Lilly and Company (R.C.M.), Indianapolis, IN; Division of Medical & Scientific Relations (M.C.C.), Alzheimer's Association, Chicago, IL; and Johns Hopkins School of Medicine (M.S.A.), Baltimore, MD. R.C.M. is currently with Global Alzheimer's Platform Foundation, Washington, DC
| | - Richard C Mohs
- From the Department of Neurology (R.C.P.), Mayo Clinic College of Medicine, Rochester, MN; Department of Neurosciences (R.G.T.), University of California, San Diego, La Jolla; University of Southern California (P.S.A.), Los Angeles; Eli Lilly and Company (R.C.M.), Indianapolis, IN; Division of Medical & Scientific Relations (M.C.C.), Alzheimer's Association, Chicago, IL; and Johns Hopkins School of Medicine (M.S.A.), Baltimore, MD. R.C.M. is currently with Global Alzheimer's Platform Foundation, Washington, DC
| | - Maria C Carrillo
- From the Department of Neurology (R.C.P.), Mayo Clinic College of Medicine, Rochester, MN; Department of Neurosciences (R.G.T.), University of California, San Diego, La Jolla; University of Southern California (P.S.A.), Los Angeles; Eli Lilly and Company (R.C.M.), Indianapolis, IN; Division of Medical & Scientific Relations (M.C.C.), Alzheimer's Association, Chicago, IL; and Johns Hopkins School of Medicine (M.S.A.), Baltimore, MD. R.C.M. is currently with Global Alzheimer's Platform Foundation, Washington, DC
| | - Marilyn S Albert
- From the Department of Neurology (R.C.P.), Mayo Clinic College of Medicine, Rochester, MN; Department of Neurosciences (R.G.T.), University of California, San Diego, La Jolla; University of Southern California (P.S.A.), Los Angeles; Eli Lilly and Company (R.C.M.), Indianapolis, IN; Division of Medical & Scientific Relations (M.C.C.), Alzheimer's Association, Chicago, IL; and Johns Hopkins School of Medicine (M.S.A.), Baltimore, MD. R.C.M. is currently with Global Alzheimer's Platform Foundation, Washington, DC
| | | |
Collapse
|
59
|
Cheng YW, Chen TF, Chiu MJ. From mild cognitive impairment to subjective cognitive decline: conceptual and methodological evolution. Neuropsychiatr Dis Treat 2017; 13:491-498. [PMID: 28243102 PMCID: PMC5317337 DOI: 10.2147/ndt.s123428] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Identification of subjects at the early stages of Alzheimer's disease (AD) is fundamental for drug development and possible intervention or prevention of cognitive decline. The concept of mild cognitive impairment (MCI) evolved during the past two decades to define subjects at the transitional stage between normal aging and dementia. Evidence from cross-sectional and longitudinal studies has shown that MCI is associated with an increased risk of positive AD biomarkers and an increased annual conversion rate of 5%-17% to AD. The presence of AD biomarkers in subjects with MCI was associated with an even higher risk of progression to dementia. However, earlier clinical trials for pharmacotherapy in subjects with MCI were disappointing. To extend the spectrum of AD to an earlier stage before MCI, subjective cognitive decline (SCD) was introduced and was defined as self-reported cognitive decline before the deficits could be detected by cognitive tests. Subjects with SCD have an increased risk of underlying AD pathology. However, SCD can also develop secondary to other heterogeneous etiologies, including other neurodegenerative and psychiatric diseases, personality traits, physical conditions, and medication use. Several clinical and biomarker features were proposed to predict risk of conversion to AD in subjects with SCD. Further longitudinal studies are needed to support the validity of these high-risk features.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- Department of Neurology, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Psychology, College of Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
60
|
Wang J, Tan L, Yu JT. Prevention Trials in Alzheimer's Disease: Current Status and Future Perspectives. J Alzheimers Dis 2016; 50:927-45. [PMID: 26836177 DOI: 10.3233/jad-150826] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly. Over the past 20 years, both pharmacological and lifestyle interventions have been studied for AD prevention, but the overall results have been disappointing. The majority of disappointing results have raised questions and great challenges for the future of AD prevention trials. Ongoing advances in the knowledge of pathogenesis, in the identification of novel targets, in improved outcome measures, and in identification and validation of biomarkers may lead to effective strategies for AD prevention. In this paper, we review the selection of participants and interventions, trial design, outcome assessments, and promising biomarkers in prevention trials, and summarize the lessons learned from completed trials and perspectives from ongoing trials in AD prevention. Selection of optimal participants and interventions, coupled with more refined outcomes and more efficient trial design, may have the capacity to deliver a new era of preventive discovery in this challenging area.
Collapse
Affiliation(s)
- Jun Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China.,Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Yuzhong District, Chongqing, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
61
|
Does mild cognitive impairment always lead to dementia? A review. J Neurol Sci 2016; 369:57-62. [DOI: 10.1016/j.jns.2016.07.055] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 11/20/2022]
|
62
|
Subjective cognitive impairment: Towards early identification of Alzheimer disease. NEUROLOGÍA (ENGLISH EDITION) 2016. [DOI: 10.1016/j.nrleng.2013.02.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
63
|
Clinical Predictors for Mild Cognitive Impairment Progression in a Korean Cohort. Dement Neurocogn Disord 2016; 15:68-74. [PMID: 30906345 PMCID: PMC6427964 DOI: 10.12779/dnd.2016.15.3.68] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 09/23/2016] [Accepted: 09/23/2016] [Indexed: 11/27/2022] Open
Abstract
Background and Purpose Patients with mild cognitive impairment (MCI) and their caregivers are concerned with the likelihood and time course of progression to dementia. This study was performed to identify the clinical predictors of the MCI progression in a Korean registry, and investigated the effects of medications without evidence, frequently prescribed in clinical practice. Methods Using a Korean cohort that included older adults with MCI who completed at least one follow-up visit, clinical characteristics and total medical expenses including prescribed medications were compared between two groups: progressed to dementia or not. Cox proportional hazards regression analysis was conducted. Results During the mean 1.42±0.72 years, 215 (27.63%) of 778 participants progressed to dementia. The best predictors were age [hazard ratio (HR), 1.036; 95% confidence interval (CI), 1.006–1.067; p=0.018], apolipoprotein ε4 allele (HR, 2.247; 95% CI, 1.512–3.337; p<0.001), Clinical Dementia Rating scale-sum of boxes scores (HR, 1.367; 95% CI, 1.143–1.636; p=0.001), Instrumental Activities of Daily Living scores (HR, 1.035; 95% CI, 1.003–1.067; p=0.029), and lower Mini-Mental State Examination scores (HR, 0.892; 95% CI, 0.839–0.949; p<0.001). Total medical expenses were not different. Conclusions Our data are in accordance with previous reports about clinical predictors for the progression from MCI to dementia. Total medical expenses were not different between groups with and without progression.
Collapse
|
64
|
Kim KH, Go HY, Lee JA, Choi J, Park S, Lee MS, Ko SG. The Effect of Dangguijagyag-san on Mild Cognitive Impairment. J Altern Complement Med 2016; 22:509-14. [DOI: 10.1089/acm.2015.0261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Kyeong Han Kim
- Korea Health Promotion Foundation, Toegye-ro, Korea
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ho-Yeon Go
- Internal Medicine College of Korean Medicine, Semyung University, Chungju, Republic of Korea
| | - Ju Ah Lee
- KM Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Jiae Choi
- Clinical Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Sunju Park
- Department of Preventive Medicine, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| | - Myeong Soo Lee
- Clinical Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
65
|
Pelton GH, Harper OL, Roose SP, Marder K, D'Antonio K, Devanand DP. Combined treatment with memantine/es-citalopram for older depressed patients with cognitive impairment: a pilot study. Int J Geriatr Psychiatry 2016; 31:648-55. [PMID: 26559790 DOI: 10.1002/gps.4375] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/18/2015] [Accepted: 09/21/2015] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The objective of the study is to assess combined antidepressant and memantine treatment in older patients presenting with depression and cognitive impairment. METHODS Thirty-five depressed patients with cognitive impairment participated in this open-label pilot study. We evaluated whether, over a 48-week period, combined antidepressant (primarily es-citalopram) and memantine treatment was effective in the treatment of cognitive impairment and depression. Neuropsychological testing was performed, and antidepressant response monitored at baseline and at the 12, 24, and 48-week time points. RESULTS Treatment with escitalopram (mean daily dose 18.62 mg, SD 5.15) and memantine (mean daily dose 13.62 mg, SD 6.67) was associated with improvement in Hamilton Depression Rating Scale scores over the 48-week study period. Patients demonstrated significant improvement in the primary outcome of cognitive performance (Selective Reminding Test total immediate recall; SRT-IR) over the 48-week treatment period (p = 0.0147). Significant improvement was also observed in measures of naming and verbal fluency but not in the other cognitive domains. One of the 35 patients (2.9%) converted to Alzheimer's disease over the 48-week treatment period. In the amnestic mild cognitive impairment subsample (n = 22), the conversion rate was 4.5%, a rate lower than in other reports of patients with DEP-CI. CONCLUSIONS In this open-label trial, combined antidepressant and memantine treatment in patients with DEP-CI was associated with improved cognition and a low rate of conversion to dementia compared with published studies in patients with DEP-CI. Although limited by the open-label study design that incorporates practice effects that can improve cognitive test performance, the findings suggest the need for a larger randomized placebo-controlled trial.
Collapse
Affiliation(s)
- Gregory H Pelton
- Late Life Depression Clinic, The Memory Disorders Center, and The Division of Geriatric Psychiatry, New York State Psychiatric Institute, New York, NY, USA
- The College of Physicians and Surgeons of Columbia University, New York, NY, USA
- Department of Neurology, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Oliver L Harper
- The College of Physicians and Surgeons of Columbia University, New York, NY, USA
| | - Steven P Roose
- Late Life Depression Clinic, The Memory Disorders Center, and The Division of Geriatric Psychiatry, New York State Psychiatric Institute, New York, NY, USA
- The College of Physicians and Surgeons of Columbia University, New York, NY, USA
| | - Karen Marder
- The College of Physicians and Surgeons of Columbia University, New York, NY, USA
- Department of Neurology, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Kristina D'Antonio
- Late Life Depression Clinic, The Memory Disorders Center, and The Division of Geriatric Psychiatry, New York State Psychiatric Institute, New York, NY, USA
| | - D P Devanand
- Late Life Depression Clinic, The Memory Disorders Center, and The Division of Geriatric Psychiatry, New York State Psychiatric Institute, New York, NY, USA
- The College of Physicians and Surgeons of Columbia University, New York, NY, USA
- Department of Neurology, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
66
|
Patient-Centred Care of Older Adults With Cardiovascular Disease and Multiple Chronic Conditions. Can J Cardiol 2016; 32:1097-107. [PMID: 27378591 DOI: 10.1016/j.cjca.2016.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/29/2016] [Accepted: 04/03/2016] [Indexed: 12/21/2022] Open
Abstract
Multimorbidity, defined as the presence of 2 or more chronic conditions, is common among older adults with cardiovascular disease. These individuals are at increased risk for poor health outcomes and account for a large proportion of health care utilization. Clinicians are challenged with the heterogeneity of this population, the complexity of the treatment regimen, limited high-quality evidence, and fragmented health care systems. Each treatment recommended by a clinical practice guideline for a single cardiovascular disease might be rational, but the combination of all evidence-based recommendations can be impractical or even harmful to individuals with multimorbidity. These challenges can be overcome with a patient-centred approach that incorporates the individual's preferences, relevant evidence, the overall and condition-specific prognosis, clinical feasibility of treatments, and interactions with other treatments and coexisting chronic conditions. The ultimate goal is to maximize benefits and minimize harms by optimizing adherence to the most essential treatments, while acknowledging trade-offs between treatments for different health conditions. It might be necessary to discontinue therapies that are not essential or potentially harmful to decrease the risk of drug-drug and drug-disease interactions from polypharmacy. A decision to initiate, withhold, or stop a treatment should be on the basis of the time horizon to benefits vs the individual's prognosis. In this review, we illustrate how cardiologists and general practitioners can adopt a patient-centred approach to focus on the aspects of cardiovascular and noncardiovascular health that have the greatest effect on functioning and quality of life in older adults with cardiovascular disease and multimorbidity.
Collapse
|
67
|
Abstract
PURPOSE OF REVIEW As individuals age, the quality of cognitive function becomes an increasingly important topic. The concept of mild cognitive impairment (MCI) has evolved over the past 2 decades to represent a state of cognitive function between that seen in normal aging and dementia. As such, it is important for health care providers to be aware of the condition and place it in the appropriate clinical context. RECENT FINDINGS Numerous international population-based studies have been conducted to document the frequency of MCI, estimating its prevalence to be between 15% and 20% in persons 60 years and older, making it a common condition encountered by clinicians. The annual rate in which MCI progresses to dementia varies between 8% and 15% per year, implying that it is an important condition to identify and treat. In those MCI cases destined to develop Alzheimer disease, biomarkers are emerging to help identify etiology and predict progression. However, not all MCI is due to Alzheimer disease, and identifying subtypes is important for possible treatment and counseling. If treatable causes are identified, the person with MCI might improve. SUMMARY MCI is an important clinical entity to identify, and while uncertainties persist, clinicians need to be aware of its diagnostic features to enable them to counsel patients. MCI remains an active area of research as numerous randomized controlled trials are being conducted to develop effective treatments.
Collapse
|
68
|
Podhorna J, Krahnke T, Shear M, Harrison JE. Alzheimer's Disease Assessment Scale-Cognitive subscale variants in mild cognitive impairment and mild Alzheimer's disease: change over time and the effect of enrichment strategies. ALZHEIMERS RESEARCH & THERAPY 2016; 8:8. [PMID: 26868820 PMCID: PMC4751673 DOI: 10.1186/s13195-016-0170-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/04/2016] [Indexed: 12/18/2022]
Abstract
Background Development of new treatments for Alzheimer’s disease (AD) has broadened into early interventions in individuals with modest cognitive impairment and a slow decline. The 11-item version of the Alzheimer’s Disease Assessment Scale–Cognitive subscale (ADAS-Cog) was originally developed to measure cognition in patients with mild to moderate AD. Attempts to improve its properties for early AD by removing items prone to ceiling and/or by adding cognitive measures known to be impaired early have yielded a number of ADAS-Cog variants. Using Alzheimer’s Disease Neuroimaging Initiative data, we compared the performance of the 3-, 5-, 11- and 13-item ADAS-Cog variants in subjects with early AD. Given the interest in enrichment strategies, we also examined this aspect with a focus on cerebrospinal fluid (CSF) markers. Methods Subjects with mild cognitive impairment (MCI) and mild AD with available ADAS-Cog 13 and CSF data were analysed. The decline over time was defined by change from baseline. Direct cross-comparison of the ADAS-Cog variants was performed using the signal-to-noise ratio (SNR), with higher values reflecting increased sensitivity to detect change over time. Results The decline over time on any of the ADAS-Cog variants was minimal in subjects with MCI. Approximately half of subjects with MCI fulfilled enrichment criteria for positive AD pathology. The impact of enrichment was detectable but subtle in MCI. The annual decline in mild AD was more pronounced but still modest. More than 90 % of subjects with mild AD had positive AD pathology. SNRs were low in MCI but greater in mild AD. The numerically largest SNRs were seen for the ADAS-Cog 5 in MCI and for both the 5- and 13-item ADAS-Cog variants in mild AD, although associated confidence intervals were large. Conclusions The possible value of ADAS-Cog expansion or reduction is less than compelling, particularly in MCI. In mild AD, adding items known to be impaired at early stages seems to provide more benefit than removing items on which subjects score close to ceiling. Electronic supplementary material The online version of this article (doi:10.1186/s13195-016-0170-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jana Podhorna
- Boehringer Ingelheim Pharma GmbH & Co. KG, Binger Strasse 173, Ingelheim/Rhein, 55218, Germany.
| | | | - Michael Shear
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany.
| | - John E Harrison
- VU University Medical Center Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
69
|
Steiner GZ, Yeung A, Liu JX, Camfield DA, Blasio FMD, Pipingas A, Scholey AB, Stough C, Chang DH. The effect of Sailuotong (SLT) on neurocognitive and cardiovascular function in healthy adults: a randomised, double-blind, placebo controlled crossover pilot trial. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:15. [PMID: 26762282 PMCID: PMC4712609 DOI: 10.1186/s12906-016-0989-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 01/07/2016] [Indexed: 11/10/2022]
Abstract
Background Sailuotong (SLT) is a standardised herbal medicine formula consisting of Panax ginseng, Ginkgo biloba, and Crocus sativus, and has been designed to enhance cognitive and cardiovascular function. Methods Using a randomised, double-blind, placebo controlled crossover design, this pilot study assessed the effect of treatment for 1 week with SLT and placebo (1 week washout period) on neurocognitive and cardiovascular function in healthy adults. Sixteen adults completed a computerised neuropsychological test battery (Compass), and had their electroencephalographic (EEG) activity and cardiovascular system function assessed. Primary outcome measures were cognitive test scores and oddball task event-related potential (ERP) component amplitudes. Secondary outcome measures were resting EEG spectral band amplitudes, and cardiovascular parameters. Results Treatment with SLT, compared to placebo, resulted in small improvements in working memory, a slight increase in auditory target (cf. nontarget) P3a amplitude, and a decrease in auditory N1 target (cf. nontarget) amplitude. There was no effect of SLT on EEG amplitude in delta, theta, alpha, or beta bands in both eyes open and eyes closed resting conditions, or on aortic and peripheral pulse pressure, and resting heartrate. Conclusions Findings suggest that SLT has the potential to improve working memory performance in healthy adults; a larger sample size is needed to confirm this. Trial registration Australia New Zealand Clinical Trials Registry Trial Registration Id: ACTRN12610000947000.
Collapse
|
70
|
Ströhle A, Schmidt DK, Schultz F, Fricke N, Staden T, Hellweg R, Priller J, Rapp MA, Rieckmann N. Drug and Exercise Treatment of Alzheimer Disease and Mild Cognitive Impairment: A Systematic Review and Meta-Analysis of Effects on Cognition in Randomized Controlled Trials. Am J Geriatr Psychiatry 2015; 23:1234-1249. [PMID: 26601726 DOI: 10.1016/j.jagp.2015.07.007] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 07/01/2015] [Accepted: 07/07/2015] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Demographic changes are increasing the pressure to improve therapeutic strategies against cognitive decline in Alzheimer disease (AD) and mild cognitive impairment (MCI). Besides drug treatment, physical activity seems to be a promising intervention target as epidemiological and clinical studies suggest beneficial effects of exercise training on cognition. Using comparable inclusion and exclusion criteria, we analyzed the efficacy of drug therapy (cholinesterase inhibitors, memantine, and Ginkgo biloba) and exercise interventions for improving cognition in AD and MCI populations. METHODS We searched The Cochrane Library, EBSCO, OVID, Web of Science, and U.S Food and Drug Administration data from inception through October 30, 2013. Randomized controlled trials in which at least one treatment arm consisted of an exercise or a pharmacological intervention for AD or MCI patients, and which had either a non-exposed control condition or a control condition that received another intervention. Treatment discontinuation rates and Standardized Mean Change score using Raw score standardization (SMCR) of cognitive performance were calculated. RESULTS Discontinuation rates varied substantially and ranged between 0% and 49% with a median of 18%. Significantly increased discontinuation rates were found for galantamine and rivastigmine as compared to placebo in AD studies. Drug treatments resulted in a small pooled effect on cognition (SMCR: 0.23, 95% CI: 0.20 to 0.25) in AD studies (N = 45, 18,434 patients) and no effect in any of the MCI studies (N = 5, 3,693 patients; SMCR: 0.03, 95% CI: 0.00 to 0.005). Exercise interventions had a moderate to strong pooled effect size (SMCR: 0.83, 95% CI: 0.59 to 1.07) in AD studies (N = 4, 119 patients), and a small effect size (SMCR: 0.20, 95% CI: 0.11 to 0.28) in MCI (N = 6, 443 patients). CONCLUSIONS Drug treatments have a small but significant impact on cognitive functioning in AD and exercise has the potential to improve cognition in AD and MCI. Head-to-head trials with sufficient statistical power are necessary to directly compare efficacy, safety, and acceptability. Combining these two approaches might further increase the efficacy of each individual intervention. IDENTIFIER PROSPERO (2013:CRD42013003910).
Collapse
Affiliation(s)
- Andreas Ströhle
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Germany.
| | - Dietlinde K Schmidt
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Germany
| | | | | | - Theresa Staden
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Germany
| | - Rainer Hellweg
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Germany; Department of Neuropsychiatry, Charité - Universitätsmedizin Berlin, Germany; Cluster of Excellence, NeuroCure, German Center for Neurodegenerative Diseases and BIH, Berlin, Germany
| | - Michael A Rapp
- Social and Preventive Medicine, University of Potsdam, Potsdam, Germany
| | - Nina Rieckmann
- Berlin School of Public Health, Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
71
|
Pottie K, Rahal R, Jaramillo A, Birtwhistle R, Thombs BD, Singh H, Gorber SC, Dunfield L, Shane A, Bacchus M, Bell N, Tonelli M. Recommendations on screening for cognitive impairment in older adults. CMAJ 2015; 188:37-46. [PMID: 26622001 DOI: 10.1503/cmaj.141165] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
| | - Kevin Pottie
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Rana Rahal
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Alejandra Jaramillo
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Richard Birtwhistle
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Brett D Thombs
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Harminder Singh
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Sarah Connor Gorber
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Lesley Dunfield
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Amanda Shane
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Maria Bacchus
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Niel Bell
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| | - Marcello Tonelli
- Departments of Family Medicine, and of Epidemiology and Community Medicine, University of Ottawa, and Bruyère Research Institute (Pottie), Ottawa, Ont.; Public Health Agency of Canada (Rahal, Jaramillo, Connor Gorber, Dunfield, Shane), Ottawa, Ont.; Departments of Family Medicine and of Community Health and Epidemiology, and Centre for Studies in Primary Care (Birtwhistle), Queen's University, Kingston, Ont.; Department of Medicine, Lady Davis Institute, Jewish General Hospital and Faculty of Medicine, McGill University (Thombs), Montréal, Que.; Department of Internal Medicine and Community Health Sciences (Singh), University of Manitoba, Winnipeg, Man.; Department of Family Medicine (Bell), University of Alberta, Edmonton, Alta.; Department of Medicine (Bacchus, Tonelli), University of Calgary, Calgary, Alta
| |
Collapse
|
72
|
Fitzpatrick-Lewis D, Warren R, Ali MU, Sherifali D, Raina P. Treatment for mild cognitive impairment: a systematic review and meta-analysis. CMAJ Open 2015; 3:E419-27. [PMID: 26770964 PMCID: PMC4701654 DOI: 10.9778/cmajo.20150057] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The effectiveness of treatments for mild cognitive impairment is uncertain. The aim of this review was to evaluate the effectiveness and harms of treatment for mild cognitive impairment in adults 65 years of age and older. METHODS We searched MEDLINE, Embase and Cochrane Central (December 2012-December 2014); citations from 2 systematic reviews were considered for inclusion. We included randomized controlled trials involving community-dwelling adults aged 65 years and older with a diagnosis of mild cognitive impairment. Studies reporting on cognition, function, behaviour, global status, mortality and adverse events for treatment with pharmacologic and nonpharmacologic interventions were included. RESULTS Seventeen studies were included. Cholinesterase inhibitor studies evaluating cognition (Alzheimer's Disease Assessment Scale, cognition subscale) showed no difference between intervention and control groups (mean difference [MD] -0.33, 95% CI -0.73 to 0.06]; one behavioural study showed no significant effect on cognition (Alzheimer's Disease Assessment Scale, cognition subscale) for the intervention group when compared to controls (MD -0.60, (95% CI -1.44 to 0.24), and one study on vitamin E showed no difference between intervention and control groups (MD 0.85, 95% CI -0.32 to 2.02). With the Mini-Mental State Examination, cholinesterase inhibitors showed no difference between intervention and control groups (MD 0.17, 95% CI -0.13 to 0.47); behavioural studies showed a significant difference favouring intervention (MD 1.01, 95% CI 0.25 to 1.77), and studies of dietary supplements and/or vitamins showed no difference between intervention and control groups (MD 0.20, 95% CI -0.04 to 0.43). Pharmacologic studies showed no difference in serious adverse events (risk ratio 0.98, 95% CI 0.86 to 1.10). No serious adverse events were reported for nonpharmacologic interventions. INTERPRETATION Treatment of mild cognitive impairment with cholinesterase inhibitors showed no benefit when compared with a control group. A small cognitive benefit was observed using behavioural therapies when compared with the control group. However, the clinical significance of this small benefit remains uncertain. The current evidence does not support the use of cholinesterase inhibitors for treating mild cognitive impairment, and future high-quality research using a standardized approach is needed to affirm the finding of a small benefit on cognition that was observed for behavioural interventions. REGISTRATION PROSPERO no. CRD42014015431.
Collapse
|
73
|
Andrieu S, Coley N, Lovestone S, Aisen PS, Vellas B. Prevention of sporadic Alzheimer's disease: lessons learned from clinical trials and future directions. Lancet Neurol 2015. [PMID: 26213339 DOI: 10.1016/s1474-4422(15)00153-2] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Interventions that have even quite modest effects at the individual level could drastically reduce the future burden of dementia associated with Alzheimer's disease at the population level. In the past three decades, both pharmacological and lifestyle interventions have been studied for the prevention of cognitive decline or dementia in randomised controlled trials of individuals mostly aged older than 50-55 years with or without risk factors for Alzheimer's disease. Several trials testing the effects of physical activity, cognitive training, or antihypertensive interventions showed some evidence of efficacy on a primary cognitive endpoint. However, most of these trials had short follow-up periods, and further evidence is needed to confirm effectiveness and establish the optimum design or dose of interventions and ideal target populations. Important innovations in ongoing trials include the development of multidomain interventions, and the use of biomarker or genetic inclusion criteria. Challenges include the use of adaptive trial designs, the development of standardised, sensitive outcome measures, and the need for interventions that can be implemented in resource-poor settings.
Collapse
Affiliation(s)
- Sandrine Andrieu
- Inserm UMR1027, F-31073, Toulouse, France; University Toulouse III, Toulouse, France; Department of Epidemiology and Public Health, CHU Toulouse, Toulouse, France.
| | - Nicola Coley
- Inserm UMR1027, F-31073, Toulouse, France; University Toulouse III, Toulouse, France; Department of Epidemiology and Public Health, CHU Toulouse, Toulouse, France
| | | | - Paul S Aisen
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, CA, USA
| | - Bruno Vellas
- Inserm UMR1027, F-31073, Toulouse, France; University Toulouse III, Toulouse, France; Department of Geriatric Medicine, CHU Toulouse, Toulouse, France
| |
Collapse
|
74
|
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is a major cause of disability and dependency amongst older people. AD drugs approved so far are symptomatic treatments and are not thought to affect the underlying disease process. Trials conducted with agents aiming to slow or stop disease progression in patients with AD have all failed, perhaps because they were tested too late in the disease process. Therefore, there has been a move towards prevention of AD. This paper presents an overview of trials testing pharmacological interventions for sporadic AD prevention. Those tested to date were initially developed for the treatment of AD or for the treatment of other conditions, rather than being specifically developed for AD prevention. Associated issues, such as evidence of 'proof-of-concept,' doses and safety, are discussed. A major shift has taken place in the methodology of AD prevention trials since the results of the first trials were published in the 1990s. New directions that are currently being considered in ongoing or future prevention trials are discussed, in terms of endpoints, target populations, and study design. The use of AD-specific drugs to prevent AD in high-risk individuals is currently limited by a lack of validated predictive and surrogate markers. Population approaches, such as lifestyle changes, are an alternative strategy that could be of public health interest, but may provide only limited benefits for individuals. The best chance of preventing AD may come from a combination of individual and population prevention approaches.
Collapse
|
75
|
A Risk-Benefit Assessment of Dementia Medications: Systematic Review of the Evidence. Drugs Aging 2015; 32:453-67. [DOI: 10.1007/s40266-015-0266-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
76
|
Abstract
Mild cognitive impairment (MCI) is widely regarded as the intermediate stage of cognitive impairment between the changes seen in normal cognitive aging and those associated with dementia. Elderly patients with MCI constitute a high-risk population for developing dementia, in particular Alzheimer's disease (AD). Although the core clinical criteria for MCI have remained largely unchanged, the operational definition of MCI has undergone several revisions over the course of the last decade and remains an evolving diagnosis. Prognostic implications of this diagnosis are becoming clearer with regard to the risk of progressive cognitive deterioration. Although patients with MCI may represent an optimal target population for pharmacological and non-pharmacological interventions, results from clinical trials have been mixed and an effective treatment remains elusive. This article provides a brief overview of the evolution of the concept of MCI and reviews current diagnostic criteria, the longitudinal course of the disorder, and current and emerging treatments for MCI.
Collapse
Affiliation(s)
- Jennifer N Vega
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University School of Medicine, 1601 23rd Ave. S., Nashville, TN, 37212, USA
| | | |
Collapse
|
77
|
Prins ND, van der Flier WA, Knol DL, Fox NC, Brashear HR, Nye JS, Barkhof F, Scheltens P. The effect of galantamine on brain atrophy rate in subjects with mild cognitive impairment is modified by apolipoprotein E genotype: post-hoc analysis of data from a randomized controlled trial. ALZHEIMERS RESEARCH & THERAPY 2014; 6:47. [PMID: 25478019 PMCID: PMC4255389 DOI: 10.1186/alzrt275] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 06/27/2014] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The aim of this investigation was to assess the effect of galantamine, an acetylcholinesterase inhibitor and allosteric modulator of nicotinic receptors, on brain atrophy in individuals with mild cognitive impairment (MCI), and to assess effect modification by apolipoprotein E (APOE) genotype. METHODS We used data from the Galantamine-International-11 (Gal-Int-11) trial, a 24-month, randomized, double blind, placebo-controlled, flexible-dose (16 to 24 mg daily) study in patients with MCI. Brain magnetic resonance imaging (MRI), including a 3-dimensional T1-weighted gradient echo volumetric sequence, was performed at screening and at 24 months. We recorded whole brain and hippocampal volumes, and calculated annual atrophy rates. Linear regression analysis was used to calculate adjusted mean differences in the rate of whole brain and hippocampal atrophy, between MCI patients treated with galantamine and with placebo. Additionally, we performed stratified analyses according to APOE genotype. RESULTS Data from 364 MCI patients with 24-month MRI data (galantamine, n = 176; placebo, n = 188) were included in the volumetric analysis. Subjects treated with galantamine demonstrated a lower rate of whole brain atrophy compared to those treated with placebo (adjusted mean difference 0.18% per year (95% confidence interval (CI) 0.04; 0.30)). Stratified analyses according to APOE genotype, showed that this effect was confined to patients who carried an APOE ϵ4 allele (adjusted mean difference 0.28% per year (95% CI 0.07; 0.50)). Rates of hippocampal atrophy did not differ significantly between study groups. CONCLUSIONS Patients with MCI who were treated with galantamine demonstrated a lower rate of whole brain atrophy, but not of hippocampal atrophy, over a 24-month treatment period, compared to those treated with placebo. This protective effect of galantamine on whole brain atrophy rate in MCI was only present in APOE ϵ4 carriers.
Collapse
Affiliation(s)
- Niels D Prins
- Alzheimer Centre, Department of Neurology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, the Netherlands ; Alzheimer Research Center, Gebouw Cronenburg, Cronenburg 75, 1081 GM Amsterdam, the Netherlands
| | - Wiesje A van der Flier
- Alzheimer Centre, Department of Neurology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, the Netherlands ; Alzheimer Research Center, Gebouw Cronenburg, Cronenburg 75, 1081 GM Amsterdam, the Netherlands
| | - Dirk L Knol
- Department of Epidemiology & Biostatistics, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Nick C Fox
- Dementia Research Centre (N.C.F.), Department of Neurodegenerative Disease, Institute of Neurology, University College London, 8-11 Queen Square, London WC1N 3BG, UK
| | - H Robert Brashear
- Janssen Alzheimer Immunotherapy Research and Development, 700 Gateway Blvd, South San Francisco, CA 94080, USA
| | - Jeffrey S Nye
- Janssen Research and Development, LLC Johnson and Johnson Innovation, One Cambridge Center, 7th Floor, Cambridge, MA 02142, USA
| | - Frederik Barkhof
- Alzheimer Centre, Department of Neurology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, the Netherlands ; Department of Radiology, Image Analysis Centre, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Philip Scheltens
- Alzheimer Centre, Department of Neurology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| |
Collapse
|
78
|
Ghosh S, Libon D, Lippa C. Mild Cognitive Impairment: A Brief Review and Suggested Clinical Algorithm. Am J Alzheimers Dis Other Demen 2014; 29:293-302. [PMID: 24370618 PMCID: PMC10852630 DOI: 10.1177/1533317513517040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mild cognitive impairment (MCI) is a dynamic state between normal cognition and dementia, where interventions can be taken to stop or delay the progression to dementia. It is broadly of 2 types-amnestic, where memory loss is the chief concern and nonamnestic, where it is not. One variant of nonamnestic, dysexecutive, being more prevalent is sometimes known as a separate subtype by itself. Diagnosis of MCI is mostly clinical and is aided by various scales and neuropsychological testing. Functional imaging studies help in early detection and is superior to biomarkers or structural magnetic resonance imaging. Although there is no evidence supporting any pharmacological intervention, cognitive rehabilitation, memory training, and caregiver support play a strong role in limiting and sometimes reversing the ongoing cognitive decline. As the spectrum of MCI is heterogeneous, making the right diagnosis can be a challenging; hence, we need a systematic yet cost-effective algorithm for the timely management of MCI.
Collapse
Affiliation(s)
- Sayantani Ghosh
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - David Libon
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Carol Lippa
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
79
|
Forlenza OV, Diniz BS, Teixeira AL, Stella F, Gattaz W. Mild cognitive impairment. Part 2: Biological markers for diagnosis and prediction of dementia in Alzheimer's disease. BRAZILIAN JOURNAL OF PSYCHIATRY 2014; 35:284-94. [PMID: 24142092 DOI: 10.1590/1516-4446-2012-3505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 09/08/2012] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To present a critical review of publications reporting on the rationale and clinical implications of the use of biomarkers for the early diagnosis of Alzheimer's disease (AD). METHODS We conducted a systematic search of the PubMed and Web of Science electronic databases, limited to articles published in English between 1999 and 2012, and based on the following terms: mild cognitive impairment, Alzheimer's disease OR dementia, biomarkers. We retrieved 1,130 articles, of which 175 were reviews. Overall, 955 original articles were eligible. RESULTS The following points were considered relevant for the present review: a) rationale for biomarkers research in AD and mild cognitive impairment (MCI); b) usefulness of distinct biomarkers for the diagnosis and prediction of AD; c) the role of multimodality biomarkers for the diagnosis and prediction of AD; d) the role of biomarkers in clinical trials of patients with AD and MCI; and e) current limitations to the widespread use of biomarkers in research and clinical settings. CONCLUSION Different biomarkers are useful for the early diagnosis and prediction of AD in at-risk subjects. Nonetheless, important methodological limitations need to be overcome for widespread use of biomarkers in research and clinical settings.
Collapse
Affiliation(s)
- Orestes V Forlenza
- Universidade de São Paulo, Laboratory of Neuroscience, Department and Institute of Psychiatry, School of Medicine, São PauloSP, Brazil
| | | | | | | | | |
Collapse
|
80
|
Petersen RC, Caracciolo B, Brayne C, Gauthier S, Jelic V, Fratiglioni L. Mild cognitive impairment: a concept in evolution. J Intern Med 2014; 275:214-28. [PMID: 24605806 PMCID: PMC3967548 DOI: 10.1111/joim.12190] [Citation(s) in RCA: 1035] [Impact Index Per Article: 94.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The construct of mild cognitive impairment (MCI) has evolved over the past 10 years since the publication of the new MCI definition at the Key Symposium in 2003, but the core criteria have remained unchanged. The construct has been extensively used worldwide, both in clinical and in research settings, to define the grey area between intact cognitive functioning and clinical dementia. A rich set of data regarding occurrence, risk factors and progression of MCI has been generated. Discrepancies between studies can be mostly explained by differences in the operationalization of the criteria, differences in the setting where the criteria have been applied, selection of subjects and length of follow-up in longitudinal studies. Major controversial issues that remain to be further explored are algorithmic versus clinical classification, reliability of clinical judgment, temporal changes in cognitive performances and predictivity of putative biomarkers. Some suggestions to further develop the MCI construct include the tailoring of the clinical criteria to specific populations and to specific contexts. The addition of biomarkers to the clinical phenotypes is promising but requires deeper investigation. Translation of findings from the specialty clinic to the population setting, although challenging, will enhance uniformity of outcomes. More longitudinal population-based studies on cognitive ageing and MCI need to be performed to clarify all these issues.
Collapse
Affiliation(s)
- R C Petersen
- From the Mayo Alzheimer's Disease Research Center, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | | | | | | | |
Collapse
|
81
|
Schneider LS, Mangialasche F, Andreasen N, Feldman H, Giacobini E, Jones R, Mantua V, Mecocci P, Pani L, Winblad B, Kivipelto M. Clinical trials and late-stage drug development for Alzheimer's disease: an appraisal from 1984 to 2014. J Intern Med 2014; 275:251-83. [PMID: 24605808 PMCID: PMC3956752 DOI: 10.1111/joim.12191] [Citation(s) in RCA: 493] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The modern era of drug development for Alzheimer's disease began with the proposal of the cholinergic hypothesis of memory impairment and the 1984 research criteria for Alzheimer's disease. Since then, despite the evaluation of numerous potential treatments in clinical trials, only four cholinesterase inhibitors and memantine have shown sufficient safety and efficacy to allow marketing approval at an international level. Although this is probably because the other drugs tested were ineffective, inadequate clinical development methods have also been blamed for the failures. Here, we review the development of treatments for Alzheimer's disease during the past 30 years, considering the drugs, potential targets, late-stage clinical trials, development methods, emerging use of biomarkers and evolution of regulatory considerations in order to summarize advances and anticipate future developments. We have considered late-stage Alzheimer's disease drug development from 1984 to 2013, including individual clinical trials, systematic and qualitative reviews, meta-analyses, methods, commentaries, position papers and guidelines. We then review the evolution of drugs in late clinical development, methods, biomarkers and regulatory issues. Although a range of small molecules and biological products against many targets have been investigated in clinical trials, the predominant drug targets have been the cholinergic system and the amyloid cascade. Trial methods have evolved incrementally: inclusion criteria have largely remained focused on mild-to-moderate Alzheimer's disease criteria, recently extending to early or prodromal Alzheimer disease or 'mild cognitive impairment due to Alzheimer's disease', for drugs considered to be disease modifying. The duration of trials has remained at 6-12 months for drugs intended to improve symptoms; 18- to 24-month trials have been established for drugs expected to attenuate clinical course. Cognitive performance, activities of daily living, global change and severity ratings have persisted as the primary clinically relevant outcomes. Regulatory guidance and oversight have evolved to allow for enrichment of early-stage Alzheimer's disease trial samples using biomarkers and phase-specific outcomes. In conclusion, validated drug targets for Alzheimer's disease remain to be developed. Only drugs that affect an aspect of cholinergic function have shown consistent, but modest, clinical effects in late-phase trials. There is opportunity for substantial improvements in drug discovery and clinical development methods.
Collapse
Affiliation(s)
- Lon S. Schneider
- Departments of Psychiatry and the Behavioral Sciences, and Neurology, Keck School of Medicine, and the Leonard Davis School of Gerontology of the University of Southern California, Los Angeles, CA, USA
| | - Francesca Mangialasche
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Section of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Niels Andreasen
- Department of Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden
- Alzheimer Disease Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Howard Feldman
- Division of Neurology, University of British Columbia, Vancouver, Canada
| | - Ezio Giacobini
- Departments of Internal Medicine, Rehabilitation and Geriatrics, University of Geneva Hospitals, Geneva, Switzerland
| | - Roy Jones
- The Research Institute for the Care of Older People (RICE) and University of Bath, Bath, UK
| | - Valentina Mantua
- European Assessment Office, Italian Medicines Agency (AIFA), Rome, Italy
| | - Patrizia Mecocci
- Section of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Luca Pani
- European Assessment Office, Italian Medicines Agency (AIFA), Rome, Italy
| | - Bengt Winblad
- Alzheimer Disease Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Miia Kivipelto
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Alzheimer Disease Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
82
|
Harrison J, Rentz DM, McLaughlin T, Niecko T, Gregg KM, Black RS, Buchanan J, Liu E, Grundman M. Cognition in MCI and Alzheimer's disease: baseline data from a longitudinal study of the NTB. Clin Neuropsychol 2014; 28:252-68. [PMID: 24521259 DOI: 10.1080/13854046.2013.875595] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Baseline data are summarized from a study examining the psychometric properties of the Neuropsychological Test Battery (NTB) and its subtests, and correlating the NTB with other cognitive and functional assessments. A multicenter, longitudinal, non-interventional study included mild to moderate Alzheimer's disease (AD, n = 196), mild cognitive impairment (MCI, n = 70), or normal cognition participants (NC, n = 75). The NTB, other cognitive assessment tools, functional/behavioral questionnaires, and health outcome assessments were administered. At baseline composite NTB, NTB memory, and NTB executive function z-scores were significantly lower for participants with AD compared with MCI, and for participants with MCI compared with NC. The composite NTB z-score had high test-retest reliability between screening and baseline. The results of this study suggest that NTB exhibits good reliability in patients with mild to moderate AD and MCI.
Collapse
|
83
|
Larner AJ. Cholinesterase inhibitors: beyond Alzheimer’s disease. Expert Rev Neurother 2014; 10:1699-705. [DOI: 10.1586/ern.10.105] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
84
|
Oulhaj A, Martin ES. Generating data from improper distributions: application to Cox proportional hazards models with cure. J STAT COMPUT SIM 2014. [DOI: 10.1080/00949655.2012.700714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
85
|
|
86
|
Hager K, Baseman AS, Nye JS, Brashear HR, Han J, Sano M, Davis B, Richards HM. Effects of galantamine in a 2-year, randomized, placebo-controlled study in Alzheimer's disease. Neuropsychiatr Dis Treat 2014; 10:391-401. [PMID: 24591834 PMCID: PMC3937252 DOI: 10.2147/ndt.s57909] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Currently available treatments for Alzheimer's disease (AD) can produce mild improvements in cognitive function, behavior, and activities of daily living in patients, but their influence on long-term survival is not well established. This study was designed to assess patient survival and drug efficacy following a 2-year galantamine treatment in patients with mild to moderately severe AD. METHODS In this multicenter, double-blind study, patients were randomized 1:1 to receive galantamine or placebo. One primary end point was safety; mortality was assessed. An independent Data Safety Monitoring Board monitored mortality for the total deaths reaching prespecified numbers, using a time-to-event method and a Cox-regression model. The primary efficacy end point was cognitive change from baseline to month 24, as measured by the Mini-Mental State Examination (MMSE) score, analyzed using intent-to-treat analysis with the 'last observation carried forward' approach, in an analysis of covariance model. RESULTS In all, 1,024 galantamine- and 1,021 placebo-treated patients received study drug, with mean age ~73 years, and mean (standard deviation [SD]) baseline MMSE score of 19 (4.08). A total of 32% of patients (661/2,045) completed the study, 27% (554/2,045) withdrew, and 41% (830/2,045) did not complete the study and were discontinued due to a Data Safety Monitoring Board-recommended early study termination. The mortality rate was significantly lower in the galantamine group versus placebo (hazard ratio [HR] =0.58; 95% confidence interval [CI]: 0.37; 0.89) (P=0.011). Cognitive impairment, based on the mean (SD) change in MMSE scores from baseline to month 24, significantly worsened in the placebo (-2.14 [4.34]) compared with the galantamine group (-1.41 [4.05]) (P<0.001). Functional impairment, based on mean (SD) change in the Disability Assessment in Dementia score (secondary end point), at month 24 significantly worsened in the placebo (-10.81 [18.27]) versus the galantamine group (-8.16 [17.25]) (P=0.002). Incidences of treatment-emergent adverse events were 54.0% for the galantamine and 48.6% for the placebo group. CONCLUSION Long-term treatment with galantamine significantly reduced mortality and the decline in cognition and daily living activities, in mild to moderate AD patients. IDENTIFICATION This study is registered at ClinicalTrials.gov (NCT00679627).
Collapse
Affiliation(s)
- Klaus Hager
- Clinic for Medicine of the Elderly, Hannover, Germany
| | | | - Jeffrey S Nye
- Janssen Research and Development, LLC, Raritan, NJ, USA
| | | | - John Han
- Janssen Research and Development, LLC, Raritan, NJ, USA
| | - Mary Sano
- The Mount Sinai Medical Center, New York, NY, USA
| | | | | |
Collapse
|
87
|
Lu J, E L, Roy N, Hutfles L, Selfridge E, Funk E, Burns JM, Swerdlow RH. Effect of cholinergic signaling on neuronal cell bioenergetics. J Alzheimers Dis 2013; 33:1135-46. [PMID: 23099815 DOI: 10.3233/jad-2012-121822] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) patients have reduced brain acetylcholine and reversing this deficit yields clinical benefits. In this study we explored how increased cholinergic tone impacts cell bioenergetics, which are also perturbed in AD. We treated SH-SY5Y neuroblastoma cells with carbachol, a cholinergic agonist, and tested for bioenergetic flux and bioenergetic infrastructure changes. Carbachol rapidly increased both oxidative phosphorylation and glycolysis fluxes. ATP levels rose slightly, as did cell energy demand, and AMPK phosphorylation occurred. At least some of these effects depended on muscarinic receptor activation, ER calcium release, and ER calcium re-uptake. Our data show that increasing cholinergic signaling enhances cell bioenergetics, and reveal mechanisms that mediate this effect. Phenomena we observed could potentially explain why cholinesterase inhibitor therapy increases AD brain glucose utilization and N-acetyl aspartate levels. The question of whether cholinesterase inhibitors have a disease modifying effect in AD has long been debated; our data suggest a theoretical mechanism through which such an effect could potentially arise.
Collapse
Affiliation(s)
- Jianghua Lu
- University of Kansas Alzheimer's Disease Center, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Abstract
PURPOSE OF REVIEW The term mild cognitive impairment (MCI) is used to describe older subjects with demonstrable cognitive impairment who have not crossed the threshold for dementia. Because patients with MCI have an increased risk of developing dementia, especially Alzheimer disease (AD), there is significant interest in the clinical characterization of these subjects and in understanding the pathophysiology of the transition from MCI to AD. RECENT FINDINGS The MCI syndrome, as an expression of an incipient disorder that may lead to dementia, is extremely heterogeneous and may coexist with systemic, neurologic, or psychiatric disorders that can cause cognitive deficits. Recent clinical criteria were designed to take into account the different forms of clinical presentation of the syndrome, and introduced the possible contribution of biomarkers to the clinical diagnosis. Bedside diagnosis of MCI can be difficult, since patients who report having cognitive problems may have normal scores in global cognitive scales or in brief neuropsychological instruments. SUMMARY This article presents the evolution of the clinical concept of MCI, the operationalization of its current definitions, the development of biomarkers that can help to identify an underlying neurodegenerative process as the etiology of the syndrome, and its proposed treatments.
Collapse
|
89
|
Tricco AC, Soobiah C, Berliner S, Ho JM, Ng CH, Ashoor HM, Chen MH, Hemmelgarn B, Straus SE. Efficacy and safety of cognitive enhancers for patients with mild cognitive impairment: a systematic review and meta-analysis. CMAJ 2013; 185:1393-401. [PMID: 24043661 DOI: 10.1503/cmaj.130451] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Cognitive enhancers, including cholinesterase inhibitors and memantine, are used to treat dementia, but their effectiveness for mild cognitive impairment is unclear. We conducted a systematic review to examine the efficacy and safety of cognitive enhancers for mild cognitive impairment. METHODS Our eligibility criteria were studies of the effects of donepezil, rivastigmine, galantamine or memantine on mild cognitive impairment reporting cognition, function, behaviour, global status, and mortality or harms. We identified relevant material by searching electronic databases (e.g., MEDLINE, Embase), the references of included studies, trial registries and conference proceedings, and by contacting experts. Two reviewers independently screened the results of the literature search, abstracted data and appraised risk of bias using the Cochrane risk-of-bias tool. RESULTS We screened 15,554 titles and abstracts and 1384 full-text articles. Eight randomized clinical trials and 3 companion reports met our inclusion criteria. We found no significant effects of cognitive enhancers on cognition (Mini-Mental State Examination: 3 randomized clinical trials [RCTs], mean difference [MD] 0.14, 95% confidence interval [CI] -0.22 to 0.50; Alzheimer's Disease Assessment Scale - cognition subscale: 3 RCTs, standardized MD -0.07, 95% CI-0.16 to 0.01]) or function (Alzheimer's Disease Cooperative Study activities of daily living inventory: 2 RCTs, MD 0.30, 95% CI -0.26 to 0.86). Cognitive enhancers were associated with higher risks of nausea, diarrhea and vomiting than placebo. INTERPRETATION Cognitive enhancers did not improve cognition or function among patients with mild cognitive impairment and were associated with a greater risk of gastrointestinal harms. Our findings do not support the use of cognitive enhancers for mild cognitive impairment.
Collapse
|
90
|
Abstract
BACKGROUND More people are presenting with mild cognitive impairment (MCI), frequently a precursor to dementia, but we do not know how to reduce deterioration. AIMS To systematically review randomised controlled trials (RCTs) evaluating the effects of any intervention for MCI on cognitive, neuropsychiatric, functional, global outcomes, life quality or incident dementia. METHOD We reviewed 41 studies fitting predetermined criteria, assessed validity using a checklist, calculated standardised outcomes and prioritised primary outcome findings in placebo-controlled studies. RESULTS The strongest evidence was that cholinesterase inhibitors did not reduce incident dementia. Cognition improved in single trials of: a heterogeneous psychological group intervention over 6 months; piribedil, a dopamine agonist over 3 months; and donepezil over 48 weeks. Nicotine improved attention over 6 months. There was equivocal evidence that Huannao Yicong improved cognition and social functioning. CONCLUSIONS There was no replicated evidence that any intervention was effective. Cholinesterase inhibitors and rofecoxib are ineffective in preventing dementia. Further good-quality RCTs are needed and preliminary evidence suggests these should include trials of psychological group interventions and piribedil.
Collapse
Affiliation(s)
- Claudia Cooper
- Mental Health Sciences Unit, University College London, Holborn Union Building, Highgate Hill, London N19 5LW, United Kingdom
| | - Ryan Li
- Mental Health Sciences Unit, University College London, Holborn Union Building, Highgate Hill, London N19 5LW, United Kingdom
| | - Constantine Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine and Johns Hopkins Bayview, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Gill Livingston
- Mental Health Sciences Unit, University College London, Holborn Union Building, Highgate Hill, London N19 5LW, United Kingdom
| |
Collapse
|
91
|
Gauthier S, Leuzy A, Rosa-Neto P. How can we improve transfer of outcomes from randomized clinical trials to clinical practice with disease-modifying drugs in Alzheimer's disease? NEURODEGENER DIS 2013; 13:197-9. [PMID: 23942173 DOI: 10.1159/000353748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 06/11/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Randomized clinical trials (RCTs) for putative disease-modifying drugs in Alzheimer's disease (AD) are using cognitive outcomes, such as the Alzheimer's Disease Assessment Scale--cognitive subscale, activities of daily living scales, such as the Alzheimer's Disease Cooperative Study Activities of Daily Living, and time from mild cognitive impairment to AD dementia. OBJECTIVE It was the aim of this study to build clinically relevant outcomes for future use in clinical practice into RCT designs and help third-party payers to measure benefit. METHODS We used a literature review for analysis. RESULTS The Clinical Dementia Rating Scale Sum of Boxes (CDR-SB) appears to be the most reliable primary outcome for RCT at different stages of AD, with the Relevant Outcome Scale for Alzheimer's Disease (ROSA) as a suitable alternative. The importance of current AD biomarkers vis-à- vis determination of efficacy of disease-modifying drugs has yet to be established; however, it is likely that at least one amyloid-specific test will be required prior to treatment with a drug acting predominantly on β-amyloid (Aβ42). Furthermore, serial MRI may be required to monitor adverse side effects associated with such drugs. CONCLUSIONS Global clinical scales such as CDR-SB and ROSA should be considered for use with treatments aiming at slowing disease progression.
Collapse
Affiliation(s)
- S Gauthier
- McGill Center for Studies in Aging, Douglas Mental Health Research Institute, Montreal, Que., Canada
| | | | | |
Collapse
|
92
|
Karantzoulis S, Novitski J, Gold M, Randolph C. The Repeatable Battery for the Assessment of Neuropsychological Status (RBANS): Utility in detection and characterization of mild cognitive impairment due to Alzheimer's disease. Arch Clin Neuropsychol 2013; 28:837-44. [PMID: 23867976 DOI: 10.1093/arclin/act057] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Current diagnostic criteria for mild cognitive impairment (MCI) due to Alzheimer's disease (AD) require standardized tests that are capable of measuring a range of neurocognitive abilities in healthy elderly individuals and sensitive to detect change over time. There currently is no clearly-established "gold standard" for this purpose. The Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) is a widely used neuropsychological test battery for the clinical diagnosis/tracking of dementia also recently incorporated into clinical trials of new investigational medications for AD treatment. The RBANS has a number of design features that suggest possible utility in diagnosis/tracking of MCI. Eighty-one patients with MCI completed the RBANS and their scores were compared with 81 demographically matched healthy controls. RBANS Total Scale scores in both groups were normally distributed, demonstrating no floor/ceiling effects. The MCI group was most impaired on the Delayed Memory Index (DMI). Receiver operating characteristic analyses reflected good discrimination, with an area under the curve of 0.88 for the Total Scale score and 0.90 for the DMI score. The profile of performance for the MCI group was similar to that previously reported for mild AD patients. The RBANS may be a suitable neurocognitive battery for the detection and tracking of MCI presumed to be due to AD.
Collapse
Affiliation(s)
- Stella Karantzoulis
- Department of Neurology, New York University Langone Medical Center, New York, NY, USA
| | | | | | | |
Collapse
|
93
|
Garcia-Ptacek S, Eriksdotter M, Jelic V, Porta-Etessam J, Kåreholt I, Manzano Palomo S. Subjective cognitive impairment: Towards early identification of Alzheimer disease. Neurologia 2013; 31:562-71. [PMID: 23601758 DOI: 10.1016/j.nrl.2013.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 02/13/2013] [Accepted: 02/17/2013] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Neurodegeneration in Alzheimer disease (AD) begins decades before dementia and patients with mild cognitive impairment (MCI) already demonstrate significant lesion loads. Lack of information about the early pathophysiology in AD complicates the search for therapeutic strategies.Subjective cognitive impairment is the description given to subjects who have memory-related complaints without pathological results on neuropsychological tests. There is no consensus regarding this heterogeneous syndrome, but at least some of these patients may represent the earliest stage in AD. METHOD We reviewed available literature in order to summarise current knowledge on subjective cognitive impairment. RESULTS Although they may not present detectable signs of disease, SCI patients as a group score lower on neuropsychological tests than the general population does, and they also have a higher incidence of future cognitive decline. Depression and psychiatric co-morbidity play a role but cannot account for all cognitive complaints. Magnetic resonance imaging studies in these patients reveal a pattern of hippocampal atrophy similar to that of amnestic mild cognitive impairment and functional MRI shows increased activation during cognitive tasks which might indicate compensation for loss of function. Prevalence of an AD-like pattern of beta-amyloid (Aβ42) and tau proteins in cerebrospinal fluid is higher in SCI patients than in the general population. CONCLUSIONS Memory complaints are relevant symptoms and may predict AD. Interpatient variability and methodological differences between clinical studies make it difficult to assign a definition to this syndrome. In the future, having a standard definition and longitudinal studies with sufficient follow-up times and an emphasis on quantifiable variables may clarify aspects of early AD.
Collapse
Affiliation(s)
- S Garcia-Ptacek
- Servicio de Neurología, Hospital Clínico San Carlos, Madrid, España; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Estocolmo, Suecia.
| | - M Eriksdotter
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Estocolmo, Suecia; Department of Geriatric Medicine, Karolinska University Hospital, Karolinska Institutet/Stockholm University, Estocolomo, Suecia
| | - V Jelic
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Estocolmo, Suecia; Department of Geriatric Medicine, Karolinska University Hospital, Karolinska Institutet/Stockholm University, Estocolomo, Suecia
| | - J Porta-Etessam
- Servicio de Neurología, Hospital Clínico San Carlos, Madrid, España
| | - I Kåreholt
- Aging Research Center, Karolinska Institutet and Stockholm University, Estocolmo, Suecia; Institute of Gerontology, School of Health Sciences, Jönköping University, Jönköping, Suecia
| | - S Manzano Palomo
- Servicio de Neurología, Hospital Infanta Cristina, Parla, Madrid, España
| |
Collapse
|
94
|
Ritchie CW, Zhinchin G. Low dose, high dose, or no dose: better prescribing of cholinesterase inhibitors for Alzheimer's disease. Int Psychogeriatr 2013; 25:511-5. [PMID: 23422014 DOI: 10.1017/s1041610212002414] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
95
|
Christa Maree Stephan B, Minett T, Pagett E, Siervo M, Brayne C, McKeith IG. Diagnosing Mild Cognitive Impairment (MCI) in clinical trials: a systematic review. BMJ Open 2013; 3:bmjopen-2012-001909. [PMID: 23386579 PMCID: PMC3586181 DOI: 10.1136/bmjopen-2012-001909] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To describe how criteria for amnestic Mild Cognitive Impairment (aMCI) have been operationalised in randomised controlled clinical trials (RCTs). DESIGN Systematic review. INFORMATION SOURCES EMBASE, PubMed and PSYCHInfo were searched from their inception to February 2012. Electronic clinical trial registries were also searched (February 2012). STUDY SELECTION RCTs were included where participant selection was made using Petersen et al-defined aMCI. There was no restriction on intervention type or the outcome tested. DATA EXTRACTION For each trial, we extracted information on study design, demographics, exclusion criteria and the operationalisation strategy for the five aMCI diagnostic criteria including: (1) memory complaint, (2) normal general cognitive function, (3) memory impairment, (4) no functional impairment and (5) no dementia. RESULTS 223 articles and 278 registered trials were reviewed, of which 22 met inclusion criteria. Various methods were applied for operationalising aMCI criteria resulting in variability in participant selection. Memory complaint and assessment of general cognitive function were the most consistently measured criteria. There was large heterogeneity in the neuropsychological methods used to determine memory impairment. It was not possible to assess the impact of these differences on case selection accuracy for dementia prediction. Further limitations include selective and unclear reporting of how each of the criteria was measured. CONCLUSIONS The results highlight the urgent need for a standardised approach to map aMCI. Lack of uniformity in clinical diagnosis, however, is not exclusively a problem for MCI but also for other clinical states such as dementia including Alzheimer's disease, Lewy Body, frontotemporal or vascular dementia. Defining a uniform approach to MCI classification, or indeed for any classification concept within the field of dementia, should be a priority if further trials are to be undertaken in the older aged population based on these concepts.
Collapse
Affiliation(s)
| | - Thais Minett
- Department of Public Health and Primary Care, Cambridge University, Cambridge, UK
| | - Emma Pagett
- Department of Public Health and Primary Care, Cambridge University, Cambridge, UK
| | - Mario Siervo
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | - Carol Brayne
- Department of Public Health and Primary Care, Cambridge University, Cambridge, UK
| | - Ian G McKeith
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
96
|
Karakaya T, Fußer F, Schröder J, Pantel J. Pharmacological Treatment of Mild Cognitive Impairment as a Prodromal Syndrome of Alzheimer´s Disease. Curr Neuropharmacol 2013; 11:102-8. [PMID: 23814542 PMCID: PMC3580783 DOI: 10.2174/157015913804999487] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 07/24/2012] [Accepted: 08/06/2012] [Indexed: 01/08/2023] Open
Abstract
Mild cognitive impairment (MCI) is a syndrome which, depending on various neurobiological, psychological and social factors, carries a high risk of developing into dementia. As far as diagnostic uncertainty and the heterogeneous underlying pathophysiological mechanisms are concerned, only limited therapeutic options are currently available. Clinical trials involving a wide range of substances have failed to show efficacy on primary and secondary outcome parameters. Most results reflect not only a lack of effectiveness of drug therapy but also methodological constraints in true prodromal Alzheimer´s disease (AD) based on clinical criteria. Biomarkers may help to identify MCI as a prodromal phase of dementia, so it is important to use them to improve specificity of case selection in future studies. For MCI as a prodromal syndrome of AD, clinical trials with disease modifying drugs that target underlying pathological mechanisms such as amyloid-beta accumulation and neurofibrillary tangle formation may help develop effective treatment options in the future. Alternative pharmacological approaches are currently being evaluated in ongoing phase 1 and phase 2 studies. Nevertheless, a lack of approved pharmacotherapeutic options has led to specific interventions that focus on patient education and life-style related factors receiving increasing attention.
Collapse
Affiliation(s)
- Tarik Karakaya
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe-University, Frankfurt am Main, Germany
| | - Fabian Fußer
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe-University, Frankfurt am Main, Germany
| | - Johannes Schröder
- Department of Psychiatry, Section of Geriatric Psychiatry, University of Heidelberg, Heidelberg, Germany
| | - Johannes Pantel
- Institute of General Practice, Geriatric Medicine, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
97
|
Kashiwaya Y, Bergman C, Lee JH, Wan R, King MT, Mughal MR, Okun E, Clarke K, Mattson MP, Veech RL. A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer's disease. Neurobiol Aging 2012; 34:1530-9. [PMID: 23276384 DOI: 10.1016/j.neurobiolaging.2012.11.023] [Citation(s) in RCA: 266] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/23/2012] [Accepted: 11/26/2012] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) involves progressive accumulation of amyloid β-peptide (Aβ) and neurofibrillary pathologies, and glucose hypometabolism in brain regions critical for memory. The 3xTgAD mouse model was used to test the hypothesis that a ketone ester-based diet can ameliorate AD pathogenesis. Beginning at a presymptomatic age, 2 groups of male 3xTgAD mice were fed a diet containing a physiological enantiomeric precursor of ketone bodies (KET) or an isocaloric carbohydrate diet. The results of behavioral tests performed at 4 and 7 months after diet initiation revealed that KET-fed mice exhibited significantly less anxiety in 2 different tests. 3xTgAD mice on the KET diet also exhibited significant, albeit relatively subtle, improvements in performance on learning and memory tests. Immunohistochemical analyses revealed that KET-fed mice exhibited decreased Aβ deposition in the subiculum, CA1 and CA3 regions of the hippocampus, and the amygdala. KET-fed mice exhibited reduced levels of hyperphosphorylated tau deposition in the same regions of the hippocampus, amygdala, and cortex. Thus, a novel ketone ester can ameliorate proteopathic and behavioral deficits in a mouse AD model.
Collapse
Affiliation(s)
- Yoshihiro Kashiwaya
- Laboratory of Metabolic Control, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Schneider JS, Pioli EY, Jianzhong Y, Li Q, Bezard E. Effects of memantine and galantamine on cognitive performance in aged rhesus macaques. Neurobiol Aging 2012; 34:1126-32. [PMID: 23158762 DOI: 10.1016/j.neurobiolaging.2012.10.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 10/05/2012] [Accepted: 10/23/2012] [Indexed: 10/27/2022]
Abstract
Current pharmacotherapies for Alzheimer's disease (AD) are focused on improving performance of daily activities, personal care, and management of problematic behaviors. Both memantine, a noncompetitive N-methyl-D-aspartate channel blocker and galantamine, a selective acetylcholinesterase inhibitor, are currently prescribed as symptomatic therapies for AD. However, drugs that progressed directly from testing in rodent models to testing in AD patients in clinical trials failed to demonstrate consistent effects on cognitive symptoms. Considering the lack of nonhuman primate data on the effects of memantine and galantamine alone or in combination on cognitive dysfunction in aged nonhuman primates, the present study examined how closely data derived from aged nonhuman primates reflects data obtained in humans. Mild beneficial effects on aspects of cognitive performance in aged primates were found, in general agreement with the human clinical experience with these drugs but in contrast to the more positive effects reported in the rodent literature. These data suggest that the nonhuman primate might have more predictive validity for drug development in this area than comparable rodent assays.
Collapse
Affiliation(s)
- Jay S Schneider
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA19107, USA.
| | | | | | | | | |
Collapse
|
99
|
Abstract
BACKGROUND Mild cognitive impairment is hypothesised to represent a pre-clinical stage of dementia but forms a heterogeneous group with variable prognosis. OBJECTIVES To assess the safety and efficacy of cholinesterase inhibitors in people with mild cognitive impairment. SEARCH METHODS Trials were identified from the Cochrane Dementia and Cognitive Improvement Group's Specialised Register, which is frequently updated from the major healthcare databases (MEDLINE, EMBASE, CINAHL, PsycINFO and Lilacs) as well as trial registers and grey literature. SELECTION CRITERIA Double-blind, placebo-controlled randomised trials of any cholinesterase inhibitor in people with mild cognitive impairment. DATA COLLECTION AND ANALYSIS Data were extracted from the published reports of the included studies, combined by meta-analysis where appropriate, and treatment efficacy and risk of adverse events were estimated. MAIN RESULTS Nine studies (from eight published reports) of 5149 individuals with mild cognitive impairment (however defined) were included in the review. Limited pooling of results was possible owing to different lengths of trials. Meta-analysis of the three studies reporting conversion to dementia gives no strong evidence of a beneficial effect of cholinesterase inhibitors on the progression to dementia at one, two or three years. The risk ratio (RR) for conversion at two years was significantly different from unity (0.67; 95% confidence interval (CI) 0.55 to 0.83), but this is based on only two studies reported in the same article. There was essentially no effect of cholinesterase inhibitors on cognitive test scores.Based on the results from 4207 individuals, there were significantly more adverse events in the cholinesterase inhibitor groups (RR 1.09; 95% CI 1.02 to 1.16), but no more serious adverse events or deaths. Gastrointestinal side effects were much more common (diarrhoea: RR 2.10; 95% CI 1.30 to 3.39; nausea: RR 2.97; 95% CI 2.57 to 3.42; vomiting: RR 4.42; 95% CI 3.23 to 6.05). Cardiac problems were no more likely in either group (RR 0.71; 95% CI 0.25 to 2.02). Other side effects reported significantly more often in the cholinesterase inhibitor group were muscle spasms/leg cramps (RR 7.52; 95% CI 4.34 to 13.02), headache (RR 1.34; 95% CI 1.05 to 1.71), syncope or dizziness (RR 1.62; 95% CI 1.36 to 1.93), insomnia (RR 1.66; 95% CI 1.36 to 2.02) and abnormal dreams (RR 4.25; 95% CI 2.57 to 7.04). AUTHORS' CONCLUSIONS There is very little evidence that cholinesterase inhibitors affect progression to dementia or cognitive test scores in mild cognitive impairment. This weak evidence is overwhelmed by the increased risk of adverse events, particularly gastrointestinal. Cholinesterase inhibitors should not be recommended for mild cognitive impairment.
Collapse
Affiliation(s)
- Tom C Russ
- Alzheimer Scotland Dementia Research Centre, Edinburgh, UK.
| | | |
Collapse
|
100
|
Cedarbaum JM, Jaros M, Hernandez C, Coley N, Andrieu S, Grundman M, Vellas B. Rationale for use of the Clinical Dementia Rating Sum of Boxes as a primary outcome measure for Alzheimer's disease clinical trials. Alzheimers Dement 2012; 9:S45-55. [PMID: 22658286 DOI: 10.1016/j.jalz.2011.11.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 11/04/2011] [Accepted: 11/17/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND We used the database of the Alzheimer's Disease Neuroimaging Initiative (ADNI) to explore the psychometric properties of the Clinical Dementia Rating Sum of Boxes (CDR-SB) to consider its utility as an outcome measure for clinical trials in early and mild, as well as later, stages of Alzheimer's disease (AD). METHODS We assessed internal consistency, structural validity, convergent validity, and 2-year internal and external responsiveness of the CDR-SB using data from 382 subjects with early or mild AD at entry into the ADNI study. RESULTS The CDR-SB assesses both cognitive and functional domains of AD disability. Mean scores declined nearly linearly; CDR-SB cognitive and functional subsums contributed equally to total scores at both very mild (early) and mild stages of the disease. CONCLUSIONS The CDR-SB has psychometric properties that make it attractive as a primary outcome measure that comprehensively assesses both cognitive and functional disability in AD patients. It may prove particularly useful for studies in early, predementia stages of AD.
Collapse
|