1
|
Shanks HRC, Chen K, Reiman EM, Blennow K, Cummings JL, Massa SM, Longo FM, Börjesson-Hanson A, Windisch M, Schmitz TW. p75 neurotrophin receptor modulation in mild to moderate Alzheimer disease: a randomized, placebo-controlled phase 2a trial. Nat Med 2024; 30:1761-1770. [PMID: 38760589 PMCID: PMC11186782 DOI: 10.1038/s41591-024-02977-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/04/2024] [Indexed: 05/19/2024]
Abstract
p75 neurotrophin receptor (p75NTR) signaling pathways substantially overlap with degenerative networks active in Alzheimer disease (AD). Modulation of p75NTR with the first-in-class small molecule LM11A-31 mitigates amyloid-induced and pathological tau-induced synaptic loss in preclinical models. Here we conducted a 26-week randomized, placebo-controlled, double-blinded phase 2a safety and exploratory endpoint trial of LM11A-31 in 242 participants with mild to moderate AD with three arms: placebo, 200 mg LM11A-31 and 400 mg LM11A-31, administered twice daily by oral capsules. This trial met its primary endpoint of safety and tolerability. Within the prespecified secondary and exploratory outcome domains (structural magnetic resonance imaging, fluorodeoxyglucose positron-emission tomography and cerebrospinal fluid biomarkers), significant drug-placebo differences were found, consistent with the hypothesis that LM11A-31 slows progression of pathophysiological features of AD; no significant effect of active treatment was observed on cognitive tests. Together, these results suggest that targeting p75NTR with LM11A-31 warrants further investigation in larger-scale clinical trials of longer duration. EU Clinical Trials registration: 2015-005263-16 ; ClinicalTrials.gov registration: NCT03069014 .
Collapse
Grants
- R35 AG071476 NIA NIH HHS
- P30 AG072980 NIA NIH HHS
- SG-23-1038904 QC Alzheimer's Association
- 2022-00732 Vetenskapsrådet (Swedish Research Council)
- P20 GM109025 NIGMS NIH HHS
- R01 AG053798 NIA NIH HHS
- R35AG71476 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- ZEN-21-848495 Alzheimer's Association
- R01 AG051596 NIA NIH HHS
- P20GM109025 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- 453677 Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de Recherche en Santé du Canada)
- P20 AG068053 NIA NIH HHS
- 2017-00915 Vetenskapsrådet (Swedish Research Council)
- U01 AG024904 NIA NIH HHS
- R01AG053798 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R25 AG083721-01 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R25 AG083721 NIA NIH HHS
- Jonathan and Joshua Memorial Foundation Government of Ontario
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- State of Arizona
- Alzheimer’s Association
- the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986 and #ALFGBG-965240), the Swedish Alzheimer Foundation (#AF-930351, #AF-939721 and #AF-968270), Hjärnfonden, Sweden (#FO2017-0243 and #ALZ2022-0006), La Fondation Recherche Alzheimer (FRA), Paris, France, the Kirsten and Freddy Johansen Foundation, Copenhagen, Denmark, and Familjen Rönströms Stiftelse, Stockholm, Sweden.
- U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- Alzheimer’s Drug Discovery Foundation (ADDF)
- Ted and Maria Quirk Endowment; Joy Chambers-Grundy Endowment.
- San Francisco VA Health Care System
- National Institutes of Aging (NIA AD Pilot Trial 1R01AG051596) PharmatrophiX (Menlo Park, California)
- Alzheimer’s Society of Canada (176677)
Collapse
Affiliation(s)
- Hayley R C Shanks
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
- Robarts Research Institute, Western University, London, Ontario, Canada.
- Western Institute for Neuroscience, Western University, London, Ontario, Canada.
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- College of Health Solutions, Arizona State University, Downtown, Phoenix, AZ, USA
| | - Eric M Reiman
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Translational Genomics Research Institute, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Stephen M Massa
- San Francisco Veterans Affairs Health Care System, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Anne Börjesson-Hanson
- Clinical Trials, Department of Aging, Karolinska University Hospital, Stockholm, Sweden
| | | | - Taylor W Schmitz
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
- Robarts Research Institute, Western University, London, Ontario, Canada.
- Western Institute for Neuroscience, Western University, London, Ontario, Canada.
| |
Collapse
|
2
|
Hajós M, Boasso A, Hempel E, Shpokayte M, Konisky A, Seshagiri CV, Fomenko V, Kwan K, Nicodemus-Johnson J, Hendrix S, Vaughan B, Kern R, Megerian JT, Malchano Z. Safety, tolerability, and efficacy estimate of evoked gamma oscillation in mild to moderate Alzheimer's disease. Front Neurol 2024; 15:1343588. [PMID: 38515445 PMCID: PMC10957179 DOI: 10.3389/fneur.2024.1343588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/05/2024] [Indexed: 03/23/2024] Open
Abstract
Background Alzheimer's Disease (AD) is a multifactorial, progressive neurodegenerative disease that disrupts synaptic and neuronal activity and network oscillations. It is characterized by neuronal loss, brain atrophy and a decline in cognitive and functional abilities. Cognito's Evoked Gamma Therapy System provides an innovative approach for AD by inducing EEG-verified gamma oscillations through sensory stimulation. Prior research has shown promising disease-modifying effects in experimental AD models. The present study (NCT03556280: OVERTURE) evaluated the feasibly, safety and efficacy of evoked gamma oscillation treatment using Cognito's medical device (CogTx-001) in participants with mild to moderate AD. Methods The present study was a randomized, double blind, sham-controlled, 6-months clinical trial in participants with mild to moderate AD. The trial enrolled 76 participants, aged 50 or older, who met the clinical criteria for AD with baseline MMSE scores between 14 and 26. Participants were randomly assigned 2:1 to receive self-administered daily, one-hour, therapy, evoking EEG-verified gamma oscillations or sham treatment. The CogTx-001 device was use at home with the help of a care partner, over 6 months. The primary outcome measures were safety, evaluated by physical and neurological exams and monthly assessments of adverse events (AEs) and MRI, and tolerability, measured by device use. Although the trial was not statistically powered to evaluate potential efficacy outcomes, primary and secondary clinical outcome measures included several cognitive and functional endpoints. Results Total AEs were similar between groups, there were no unexpected serious treatment related AEs, and no serious treatment-emergent AEs that led to study discontinuation. MRI did not show Amyloid-Related Imaging Abnormalities (ARIA) in any study participant. High adherence rates (85-90%) were observed in sham and treatment participants. There was no statistical separation between active and sham arm participants in primary outcome measure of MADCOMS or secondary outcome measure of CDR-SB or ADAS-Cog14. However, some secondary outcome measures including ADCS-ADL, MMSE, and MRI whole brain volume demonstrated reduced progression in active compared to sham treated participants, that achieved nominal significance. Conclusion Our results demonstrate that 1-h daily treatment with Cognito's Evoked Gamma Therapy System (CogTx-001) was safe and well-tolerated and demonstrated potential clinical benefits in mild to moderate AD.Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT03556280.
Collapse
Affiliation(s)
- Mihály Hajós
- Cognito Therapeutics, Inc., Cambridge, MA, United States
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Alyssa Boasso
- Cognito Therapeutics, Inc., Cambridge, MA, United States
| | - Evan Hempel
- Cognito Therapeutics, Inc., Cambridge, MA, United States
| | | | - Alex Konisky
- Cognito Therapeutics, Inc., Cambridge, MA, United States
| | | | | | - Kim Kwan
- Cognito Therapeutics, Inc., Cambridge, MA, United States
| | | | | | - Brent Vaughan
- Cognito Therapeutics, Inc., Cambridge, MA, United States
| | - Ralph Kern
- Cognito Therapeutics, Inc., Cambridge, MA, United States
| | | | - Zach Malchano
- Cognito Therapeutics, Inc., Cambridge, MA, United States
| |
Collapse
|
3
|
Li T, Zhu H, Li T, Zhu H. Asynchronous functional linear regression models for longitudinal data in reproducing kernel Hilbert space. Biometrics 2023; 79:1880-1895. [PMID: 36205584 DOI: 10.1111/biom.13767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/21/2022] [Indexed: 11/02/2022]
Abstract
Motivated by the analysis of longitudinal neuroimaging studies, we study the longitudinal functional linear regression model under asynchronous data setting for modeling the association between clinical outcomes and functional (or imaging) covariates. In the asynchronous data setting, both covariates and responses may be measured at irregular and mismatched time points, posing methodological challenges to existing statistical methods. We develop a kernel weighted loss function with roughness penalty to obtain the functional estimator and derive its representer theorem. The rate of convergence, a Bahadur representation, and the asymptotic pointwise distribution of the functional estimator are obtained under the reproducing kernel Hilbert space framework. We propose a penalized likelihood ratio test to test the nullity of the functional coefficient, derive its asymptotic distribution under the null hypothesis, and investigate the separation rate under the alternative hypotheses. Simulation studies are conducted to examine the finite-sample performance of the proposed procedure. We apply the proposed methods to the analysis of multitype data obtained from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study, which reveals significant association between 21 regional brain volume density curves and the cognitive function. Data used in preparation of this paper were obtained from the ADNI database (adni.loni.usc.edu).
Collapse
Affiliation(s)
- Ting Li
- School of Statistics and Management, Shanghai University of Finance and Economics, Shanghai, China
- Shanghai Institute of International Finance and Economics, Shanghai University of Finance and Economics, Shanghai, China
| | - Huichen Zhu
- Department of Statistics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tengfei Li
- Department of Radiology and Biomedical Research Imaging Center (BRIC), University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
4
|
Mirkin S, Albensi BC. Should artificial intelligence be used in conjunction with Neuroimaging in the diagnosis of Alzheimer's disease? Front Aging Neurosci 2023; 15:1094233. [PMID: 37187577 PMCID: PMC10177660 DOI: 10.3389/fnagi.2023.1094233] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/27/2023] [Indexed: 05/17/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disorder that affects memory, thinking, behavior, and other cognitive functions. Although there is no cure, detecting AD early is important for the development of a therapeutic plan and a care plan that may preserve cognitive function and prevent irreversible damage. Neuroimaging, such as magnetic resonance imaging (MRI), computed tomography (CT), and positron emission tomography (PET), has served as a critical tool in establishing diagnostic indicators of AD during the preclinical stage. However, as neuroimaging technology quickly advances, there is a challenge in analyzing and interpreting vast amounts of brain imaging data. Given these limitations, there is great interest in using artificial Intelligence (AI) to assist in this process. AI introduces limitless possibilities in the future diagnosis of AD, yet there is still resistance from the healthcare community to incorporate AI in the clinical setting. The goal of this review is to answer the question of whether AI should be used in conjunction with neuroimaging in the diagnosis of AD. To answer the question, the possible benefits and disadvantages of AI are discussed. The main advantages of AI are its potential to improve diagnostic accuracy, improve the efficiency in analyzing radiographic data, reduce physician burnout, and advance precision medicine. The disadvantages include generalization and data shortage, lack of in vivo gold standard, skepticism in the medical community, potential for physician bias, and concerns over patient information, privacy, and safety. Although the challenges present fundamental concerns and must be addressed when the time comes, it would be unethical not to use AI if it can improve patient health and outcome.
Collapse
Affiliation(s)
- Sophia Mirkin
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Benedict C. Albensi
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
- St. Boniface Hospital Research, Winnipeg, MB, Canada
- University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
5
|
Li Y, Wang M, Cong L, Hou T, Song L, Wang X, Shi L, Dekhtyar S, Wang Y, Du Y, Qiu C. Lifelong Cognitive Reserve, Imaging Markers of Brain Aging, and Cognitive Function in Dementia-Free Rural Older Adults: A Population-Based Study. J Alzheimers Dis 2023; 92:261-272. [PMID: 36710675 PMCID: PMC10041437 DOI: 10.3233/jad-220864] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND Cognitive reserve (CR) partly explains cognitive variability in the presence of pathological brain aging. OBJECTIVE We investigated the interplay of lifelong CR with age, sex, and brain aging markers in cognitive phenotypes among older adults with very limited education. METHODS This population-based cross-sectional study included 179 dementia-free participants (age ≥65 years; 39.7% women; 67.0% had no or elementary education) examined in 2014-2016. We assessed lacunes and volumes of hippocampus, ventricles, grey matter, white matter (WM), and white matter hyperintensities. Lifelong CR score was generated from six lifespan intellectual factors (e.g., education and social support). We used Mini-Mental State Examination (MMSE) score to assess cognition and Petersen's criteria to define mild cognitive impairment (MCI). Data were analyzed using general linear and logistic models. RESULTS The association of higher lifelong CR score (range: -4.0-5.0) with higher MMSE score was stronger in women (multivariable-adjusted β-coefficient and 95% CI: 1.75;0.99-2.51) than in men (0.68;0.33-1.03) (pinteraction = 0.006). The association of higher CR with MCI (multivariable-adjusted odds ratio and 95% CI: 0.77;0.60-0.99) did not vary by age or sex. Among participants with low CR (<1.4[median]), greater hippocampal and WM volumes were related to higher MMSE scores with multivariable-adjusted β-coefficients being 1.77(0.41-3.13) and 0.44(0.15-0.74); the corresponding figures in those with high CR were 0.15(-0.76-1.07) and -0.17(-0.41-0.07) (pinteraction <0.01). There was no statistical interaction of CR with MRI markers on MCI. CONCLUSION Greater lifelong CR capacity is associated with better late-life cognition among people with limited education, possibly by compensating for impact of neurodegeneration.
Collapse
Affiliation(s)
- Yuanjing Li
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Mingqi Wang
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
| | - Lin Cong
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
| | - Tingting Hou
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
| | - Lin Song
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
| | - Xiang Wang
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong SAR, P.R. China
| | - Serhiy Dekhtyar
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Chengxuan Qiu
- Department of Neurology, Shandong Provincial Hospital, Jinan, Shandong, P.R. China
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| |
Collapse
|
6
|
Iversen WL, Monroe TB, Atalla S, Anderson AR, Cowan RL, Wright KD, Failla MD, Moss KO. Promoting successful participation of people living with Alzheimer's disease and related dementias in pain-related neuroimaging research studies. FRONTIERS IN PAIN RESEARCH 2022; 3:926459. [PMID: 36061416 PMCID: PMC9437430 DOI: 10.3389/fpain.2022.926459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Recruitment and retention of participants for pain-related neuroimaging research is challenging and becomes increasingly so when research participants have a diagnosis of Alzheimer's disease and related dementias (ADRD). This article shares the authors' recommendations from several years of successful recruitment and completion of pain-related neuroimaging studies of people living with ADRD and includes supportive literature. While not an exhaustive list, this review covers several topics related to recruitment and retention of participants living with ADRD, including community engagement, capacity to consent, dementia diagnostic criteria, pain medication and other study exclusion criteria, participant and caregiver burden, communication concerns, and relationships with neuroimaging facilities. Threaded throughout the paper are important cultural considerations. Additionally, we discuss implications of the coronavirus (COVID-19) pandemic for recruitment. Once tailored to specific research study protocols, these proven strategies may assist researchers with successfully recruiting and retaining participants living with ADRD for pain-related neuroimaging research studies toward improving overall health outcomes.
Collapse
Affiliation(s)
- Wm. Larkin Iversen
- College of Nursing, The Ohio State University, Columbus, OH, United States
| | - Todd B. Monroe
- College of Nursing, The Ohio State University, Columbus, OH, United States
| | - Sebastian Atalla
- Department of Physics and Astronomy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alison R. Anderson
- College of Nursing, The Ohio State University, Columbus, OH, United States
| | - Ronald L. Cowan
- University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kathy D. Wright
- College of Nursing, The Ohio State University, Columbus, OH, United States
| | - Michelle D. Failla
- College of Nursing, The Ohio State University, Columbus, OH, United States
| | - Karen O. Moss
- College of Nursing, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
7
|
Zarzar TG, Lee B, Coughlin R, Kim D, Shen L, Hall MA. Sex Differences in the Metabolome of Alzheimer's Disease Progression. FRONTIERS IN RADIOLOGY 2022; 2:782864. [PMID: 35445209 PMCID: PMC9014653 DOI: 10.3389/fradi.2022.782864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia; however, men and women face differential AD prevalence, presentation, and progression risks. Characterizing metabolomic profiles during AD progression is fundamental to understand the metabolic disruptions and the biological pathways involved. However, outstanding questions remain of whether peripheral metabolic changes occur equally in men and women with AD. Here, we evaluated differential effects of metabolomic and brain volume associations between sexes. We used three cohorts from the Alzheimer's Disease Neuroimaging Initiative (ADNI), evaluated 1,368 participants, two metabolomic platforms with 380 metabolites in total, and six brain segment volumes. Using dimension reduction techniques, we took advantage of the correlation structure of the brain volume phenotypes and the metabolite concentration values to reduce the number of tests while aggregating relevant biological structures. Using WGCNA, we aggregated modules of highly co-expressed metabolites. On the other hand, we used partial least squares regression-discriminant analysis (PLS-DA) to extract components of brain volumes that maximally co-vary with AD diagnosis as phenotypes. We tested for differences in effect sizes between sexes in the association between single metabolite and metabolite modules with the brain volume components. We found five metabolite modules and 125 single metabolites with significant differences between sexes. These results highlight a differential lipid disruption in AD progression between sexes. Men showed a greater negative association of phosphatidylcholines and sphingomyelins and a positive association of VLDL and large LDL with AD progression. In contrast, women showed a positive association of triglycerides in VLDL and small and medium LDL with AD progression. Explicitly identifying sex differences in metabolomics during AD progression can highlight particular metabolic disruptions in each sex. Our research study and strategy can lead to better-tailored studies and better-suited treatments that take sex differences into account.
Collapse
Affiliation(s)
- Tomás González Zarzar
- Department of Veterinary and Biomedical Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA, United States.,Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Brian Lee
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rory Coughlin
- Department of Veterinary and Biomedical Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Molly A Hall
- Department of Veterinary and Biomedical Sciences, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA, United States.,Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States.,Penn State Cancer Institute, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
8
|
Zou H, Li K, Zeng D, Luo S. Bayesian inference and dynamic prediction of multivariate joint model with functional data: An application to Alzheimer's disease. Stat Med 2021; 40:6855-6872. [PMID: 34649301 PMCID: PMC8671252 DOI: 10.1002/sim.9214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 08/03/2021] [Accepted: 09/20/2021] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder impairing multiple domains, for example, cognition and behavior. Assessing the risk of AD progression and initiating timely interventions at early stages are critical to improve the quality of life for AD patients. Due to the heterogeneous nature and complex mechanisms of AD, one single longitudinal outcome is insufficient to assess AD severity and disease progression. Therefore, AD studies collect multiple longitudinal outcomes, including cognitive and behavioral measurements, as well as structural brain images such as magnetic resonance imaging (MRI). How to utilize the multivariate longitudinal outcomes and MRI data to make efficient statistical inference and prediction is an open question. In this article, we propose a multivariate joint model with functional data (MJM-FD) framework that relates multiple correlated longitudinal outcomes to a survival outcome, and use the scalar-on-function regression method to include voxel-based whole-brain MRI data as functional predictors in both longitudinal and survival models. We adopt a Bayesian paradigm to make statistical inference and develop a dynamic prediction framework to predict an individual's future longitudinal outcomes and risk of a survival event. We validate the MJM-FD framework through extensive simulation studies and apply it to the motivating Alzheimer's Disease Neuroimaging Initiative (ADNI) study.
Collapse
Affiliation(s)
- Haotian Zou
- Gillings School of Global Public Health, Department of Biostatistics, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Kan Li
- Merck Research Lab, Merck & Co, North Wales, Pennsylvania
| | - Donglin Zeng
- Department of Biostatistics, Gillings School of Global Public Health, CB#7420, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Sheng Luo
- Corresponding author: Sheng Luo, Department of Biostatistics and Informatics, Duke University, 2424 Erwin Rd, Durham, NC 27705, USA ()
| | | |
Collapse
|
9
|
Sapkota S, McFall GP, Masellis M, Dixon RA, Black SE. Differential Cognitive Decline in Alzheimer's Disease Is Predicted by Changes in Ventricular Size but Moderated by Apolipoprotein E and Pulse Pressure. J Alzheimers Dis 2021; 85:545-560. [PMID: 34864669 DOI: 10.3233/jad-215068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Differential cognitive trajectories in Alzheimer's disease (AD) may be predicted by biomarkers from multiple domains. OBJECTIVE In a longitudinal sample of AD and AD-related dementias patients (n = 312), we tested whether 1) change in brain morphometry (ventricular enlargement) predicts differential cognitive trajectories, 2) further risk is contributed by genetic (Apolipoprotein E [APOE] ɛ4+) and vascular (pulse pressure [PP]) factors separately, and 3) the genetic + vascular risk moderates this pattern. METHODS We applied a dynamic computational approach (parallel process models) to test both concurrent and change-related associations between predictor (ventricular size) and cognition (executive function [EF]/attention). We then tested these associations as stratified by APOE (ɛ4-/ɛ4+), PP (low/high), and APOE+ PP (low/intermediate/high) risk. RESULTS First, concurrently, higher ventricular size predicted lower EF/attention performance and, longitudinally, increasing ventricular size predicted steeper EF/attention decline. Second, concurrently, higher ventricular size predicted lower EF/attention performance selectively in APOEɛ4+ carriers, and longitudinally, increasing ventricular size predicted steeper EF/attention decline selectively in the low PP group. Third, ventricular size and EF/attention associations were absent in the high APOE+ PP risk group both concurrently and longitudinally. CONCLUSION As AD progresses, a threshold effect may be present in which ventricular enlargement in the context of exacerbated APOE+ PP risk does not produce further cognitive decline.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - G Peggy McFall
- Department of Psychology (Science), University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Mario Masellis
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Medicine (Neurology), University of Toronto, Toronto, ON, Canada
| | - Roger A Dixon
- Department of Psychology (Science), University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Sandra E Black
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Medicine (Neurology), University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Bottiroli S, Bernini S, Cavallini E, Sinforiani E, Zucchella C, Pazzi S, Cristiani P, Vecchi T, Tost D, Sandrini G, Tassorelli C. The Smart Aging Platform for Assessing Early Phases of Cognitive Impairment in Patients With Neurodegenerative Diseases. Front Psychol 2021; 12:635410. [PMID: 33790839 PMCID: PMC8005545 DOI: 10.3389/fpsyg.2021.635410] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/05/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Smart Aging is a serious game (SG) platform that generates a 3D virtual reality environment in which users perform a set of screening tasks designed to allow evaluation of global cognition. Each task replicates activities of daily living performed in a familiar environment. The main goal of the present study was to ascertain whether Smart Aging could differentiate between different types and levels of cognitive impairment in patients with neurodegenerative disease. Methods: Ninety-one subjects (mean age = 70.29 ± 7.70 years)—healthy older adults (HCs, n = 23), patients with single-domain amnesic mild cognitive impairment (aMCI, n = 23), patients with single-domain executive Parkinson's disease MCI (PD-MCI, n = 20), and patients with mild Alzheimer's disease (mild AD, n = 25)—were enrolled in the study. All participants underwent cognitive evaluations performed using both traditional neuropsychological assessment tools, including the Mini-Mental State Examination (MMSE), Montreal Overall Cognitive Assessment (MoCA), and the Smart Aging platform. We analyzed global scores on Smart Aging indices (i.e., accuracy, time, distance) as well as the Smart Aging total score, looking for differences between the four groups. Results: The findings revealed significant between-group differences in all the Smart Aging indices: accuracy (p < 0.001), time (p < 0.001), distance (p < 0.001), and total Smart Aging score (p < 0.001). The HCs outperformed the mild AD, aMCI, and PD-MCI patients in terms of accuracy, time, distance, and Smart Aging total score. In addition, the mild AD group was outperformed both by the HCs and by the aMCI and PD-MCI patients on accuracy and distance. No significant differences were found between aMCI and PD-MCI patients. Finally, the Smart Aging scores significantly correlated with the results of the neuropsychological assessments used. Conclusion: These findings, although preliminary due to the small sample size, suggest the validity of Smart Aging as a screening tool for the detection of cognitive impairment in patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Sara Bottiroli
- Faculty of Law, Giustino Fortunato University, Benevento, Italy.,National Neurological Institute C. Mondino Foundation, Pavia, Italy
| | - Sara Bernini
- National Neurological Institute C. Mondino Foundation, Pavia, Italy
| | - Elena Cavallini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Elena Sinforiani
- National Neurological Institute C. Mondino Foundation, Pavia, Italy
| | - Chiara Zucchella
- Neurology Unit, Department of Neurosciences, Verona University Hospital, Verona, Italy
| | - Stefania Pazzi
- Consorzio di Bioingegneria Medica e Informatica CBIM, Pavia, Italy
| | - Paolo Cristiani
- Consorzio di Bioingegneria Medica e Informatica CBIM, Pavia, Italy
| | - Tomaso Vecchi
- National Neurological Institute C. Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Daniela Tost
- Computer Graphics Division Research Centre for Biomedical Engineering (CREB), Universitat Politecnica de Catalunya, Barcelona, Spain
| | - Giorgio Sandrini
- National Neurological Institute C. Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Cristina Tassorelli
- National Neurological Institute C. Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
11
|
Thabtah F, Spencer R, Ye Y. The correlation of everyday cognition test scores and the progression of Alzheimer's disease: a data analytics study. Health Inf Sci Syst 2020; 8:24. [PMID: 32765845 DOI: 10.1007/s13755-020-00114-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/14/2020] [Indexed: 11/25/2022] Open
Abstract
The process of diagnosing dementia conditions, especially Alzheimer's disease, and the cognitive tests that are involved in this process, are important areas of study. Everyday Cognition (ECog) is one test that can be used as part of Alzheimer's disease diagnosis to measure cognitive decline in different areas. In this study, we investigate two versions of the ECog test: the study partner reported version (ECogSP), and the patient reported version (ECogPT). We compare these, using statistical analysis and machine learning techniques, to create classification models to demonstrate the progression in ECog scores over time by using the Alzheimer's Disease Neuroimaging Initiative longitudinal data repository (ADNI); participants are classed with having normal cognition, mild cognitive impairment, or Alzheimer's disease. We found that participants who are diagnosed with Alzheimer's disease at baseline, or during a subsequent visit, tend to self-report consistent ECogPT scores over time indicating no change in cognitive ability. However, study partners tend to report higher and increasing ECogSP scores on behalf of participants in the same diagnosis category; this would indicate a degradation in the participant's cognitive ability over time, consistent with the progress of Alzheimer's disease.
Collapse
Affiliation(s)
- Fadi Thabtah
- Digital Technologies, Manukau Institute of Technology, Auckland, New Zealand
| | - Robinson Spencer
- Digital Technologies, Manukau Institute of Technology, Auckland, New Zealand
| | - Yongsheng Ye
- Digital Technologies, Manukau Institute of Technology, Auckland, New Zealand
| |
Collapse
|
12
|
Volume estimation of brain ventricles using Cavalieri's principle and Atlas-based methods in Alzheimer disease: Consistency between methods. J Clin Neurosci 2020; 78:333-338. [PMID: 32360163 DOI: 10.1016/j.jocn.2020.04.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/15/2020] [Indexed: 11/20/2022]
Abstract
Automatic estimations of brain ventricles are needed to assess disease progression in neurodegenerative disorders such as Alzheimer Disease (AD). The objectives of this study are to evaluate the diagnostic performances of an automated volumetric assessment tool in estimating lateral ventricle volumes in AD and to compare this with Cavalieri's principle, which is accepted as the gold standard method. This is across-sectional volumetric study including 25 Alzheimer patients and 25 healthy subjects undergoing magnetic resonance images (MRI) with a 3D turbo spin echo sequence at 1.5 Tesla. The Atlas-based method incorporated MRIStudio software to automatically measure he volumes of brain ventricles. To compare the corresponding measurements, we used manual point-counting and semi-automatic planimetry methods based on Cavalieri's principle. Bland-Altman test results indicated an excellent agreement between Cavalieri's principle and the Atlas-based method in all volumetric measurements (p < 0.05). We obtained a 64% sensitivity and 92% specificity for lateral ventricular volumes according to the Atlas-based method. AD subjects had significantly larger left and right lateral ventricle volume (LVV) when compared to control subjects in respect to three volumetric methods (p < 0.01). Lateral ventricle-to-brain ratio (VBR) statistically increased 49.23% in measurements done with the point-counting method, 45.12% with the planimetry method, and 45.49% with the Atlas-based method in AD patients (p < 0.01). As a result, the Atlas-based method may be used instead of manual volumetry to estimate brain volumes. Additionally, this method provides rapid and accurate estimations of brain ventricular volumes in-vivo examination of MRI.
Collapse
|
13
|
Lombardi G, Crescioli G, Cavedo E, Lucenteforte E, Casazza G, Bellatorre A, Lista C, Costantino G, Frisoni G, Virgili G, Filippini G. Structural magnetic resonance imaging for the early diagnosis of dementia due to Alzheimer's disease in people with mild cognitive impairment. Cochrane Database Syst Rev 2020; 3:CD009628. [PMID: 32119112 PMCID: PMC7059964 DOI: 10.1002/14651858.cd009628.pub2] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Mild cognitive impairment (MCI) due to Alzheimer's disease is the symptomatic predementia phase of Alzheimer's disease dementia, characterised by cognitive and functional impairment not severe enough to fulfil the criteria for dementia. In clinical samples, people with amnestic MCI are at high risk of developing Alzheimer's disease dementia, with annual rates of progression from MCI to Alzheimer's disease estimated at approximately 10% to 15% compared with the base incidence rates of Alzheimer's disease dementia of 1% to 2% per year. OBJECTIVES To assess the diagnostic accuracy of structural magnetic resonance imaging (MRI) for the early diagnosis of dementia due to Alzheimer's disease in people with MCI versus the clinical follow-up diagnosis of Alzheimer's disease dementia as a reference standard (delayed verification). To investigate sources of heterogeneity in accuracy, such as the use of qualitative visual assessment or quantitative volumetric measurements, including manual or automatic (MRI) techniques, or the length of follow-up, and age of participants. MRI was evaluated as an add-on test in addition to clinical diagnosis of MCI to improve early diagnosis of dementia due to Alzheimer's disease in people with MCI. SEARCH METHODS On 29 January 2019 we searched Cochrane Dementia and Cognitive Improvement's Specialised Register and the databases, MEDLINE, Embase, BIOSIS Previews, Science Citation Index, PsycINFO, and LILACS. We also searched the reference lists of all eligible studies identified by the electronic searches. SELECTION CRITERIA We considered cohort studies of any size that included prospectively recruited people of any age with a diagnosis of MCI. We included studies that compared the diagnostic test accuracy of baseline structural MRI versus the clinical follow-up diagnosis of Alzheimer's disease dementia (delayed verification). We did not exclude studies on the basis of length of follow-up. We included studies that used either qualitative visual assessment or quantitative volumetric measurements of MRI to detect atrophy in the whole brain or in specific brain regions, such as the hippocampus, medial temporal lobe, lateral ventricles, entorhinal cortex, medial temporal gyrus, lateral temporal lobe, amygdala, and cortical grey matter. DATA COLLECTION AND ANALYSIS Four teams of two review authors each independently reviewed titles and abstracts of articles identified by the search strategy. Two teams of two review authors each independently assessed the selected full-text articles for eligibility, extracted data and solved disagreements by consensus. Two review authors independently assessed the quality of studies using the QUADAS-2 tool. We used the hierarchical summary receiver operating characteristic (HSROC) model to fit summary ROC curves and to obtain overall measures of relative accuracy in subgroup analyses. We also used these models to obtain pooled estimates of sensitivity and specificity when sufficient data sets were available. MAIN RESULTS We included 33 studies, published from 1999 to 2019, with 3935 participants of whom 1341 (34%) progressed to Alzheimer's disease dementia and 2594 (66%) did not. Of the participants who did not progress to Alzheimer's disease dementia, 2561 (99%) remained stable MCI and 33 (1%) progressed to other types of dementia. The median proportion of women was 53% and the mean age of participants ranged from 63 to 87 years (median 73 years). The mean length of clinical follow-up ranged from 1 to 7.6 years (median 2 years). Most studies were of poor methodological quality due to risk of bias for participant selection or the index test, or both. Most of the included studies reported data on the volume of the total hippocampus (pooled mean sensitivity 0.73 (95% confidence interval (CI) 0.64 to 0.80); pooled mean specificity 0.71 (95% CI 0.65 to 0.77); 22 studies, 2209 participants). This evidence was of low certainty due to risk of bias and inconsistency. Seven studies reported data on the atrophy of the medial temporal lobe (mean sensitivity 0.64 (95% CI 0.53 to 0.73); mean specificity 0.65 (95% CI 0.51 to 0.76); 1077 participants) and five studies on the volume of the lateral ventricles (mean sensitivity 0.57 (95% CI 0.49 to 0.65); mean specificity 0.64 (95% CI 0.59 to 0.70); 1077 participants). This evidence was of moderate certainty due to risk of bias. Four studies with 529 participants analysed the volume of the total entorhinal cortex and four studies with 424 participants analysed the volume of the whole brain. We did not estimate pooled sensitivity and specificity for the volume of these two regions because available data were sparse and heterogeneous. We could not statistically evaluate the volumes of the lateral temporal lobe, amygdala, medial temporal gyrus, or cortical grey matter assessed in small individual studies. We found no evidence of a difference between studies in the accuracy of the total hippocampal volume with regards to duration of follow-up or age of participants, but the manual MRI technique was superior to automatic techniques in mixed (mostly indirect) comparisons. We did not assess the relative accuracy of the volumes of different brain regions measured by MRI because only indirect comparisons were available, studies were heterogeneous, and the overall accuracy of all regions was moderate. AUTHORS' CONCLUSIONS The volume of hippocampus or medial temporal lobe, the most studied brain regions, showed low sensitivity and specificity and did not qualify structural MRI as a stand-alone add-on test for an early diagnosis of dementia due to Alzheimer's disease in people with MCI. This is consistent with international guidelines, which recommend imaging to exclude non-degenerative or surgical causes of cognitive impairment and not to diagnose dementia due to Alzheimer's disease. In view of the low quality of most of the included studies, the findings of this review should be interpreted with caution. Future research should not focus on a single biomarker, but rather on combinations of biomarkers to improve an early diagnosis of Alzheimer's disease dementia.
Collapse
Affiliation(s)
- Gemma Lombardi
- University of FlorenceDepartment of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)Largo Brambilla, 3FlorenceItaly50134
| | - Giada Crescioli
- University of FlorenceDepartment of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)Largo Brambilla, 3FlorenceItaly50134
| | - Enrica Cavedo
- Pitie‐Salpetriere Hospital, Sorbonne UniversityAlzheimer Precision Medicine (APM), AP‐HP47 boulevard de l'HopitalParisFrance75013
| | - Ersilia Lucenteforte
- University of PisaDepartment of Clinical and Experimental MedicineVia Savi 10PisaItaly56126
| | - Giovanni Casazza
- Università degli Studi di MilanoDipartimento di Scienze Biomediche e Cliniche "L. Sacco"via GB Grassi 74MilanItaly20157
| | | | - Chiara Lista
- Fondazione I.R.C.C.S. Istituto Neurologico Carlo BestaNeuroepidemiology UnitVia Celoria, 11MilanoItaly20133
| | - Giorgio Costantino
- Ospedale Maggiore Policlinico, Università degli Studi di MilanoUOC Pronto Soccorso e Medicina D'Urgenza, Fondazione IRCCS Ca' GrandaMilanItaly
| | | | - Gianni Virgili
- University of FlorenceDepartment of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA)Largo Brambilla, 3FlorenceItaly50134
| | - Graziella Filippini
- Carlo Besta Foundation and Neurological InstituteScientific Director’s Officevia Celoria, 11MilanItaly20133
| | | |
Collapse
|
14
|
Broadhouse KM, Singh MF, Suo C, Gates N, Wen W, Brodaty H, Jain N, Wilson GC, Meiklejohn J, Singh N, Baune BT, Baker M, Foroughi N, Wang Y, Kochan N, Ashton K, Brown M, Li Z, Mavros Y, Sachdev PS, Valenzuela MJ. Hippocampal plasticity underpins long-term cognitive gains from resistance exercise in MCI. Neuroimage Clin 2020; 25:102182. [PMID: 31978826 PMCID: PMC6974789 DOI: 10.1016/j.nicl.2020.102182] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 01/15/2023]
Abstract
Dementia affects 47 million individuals worldwide, and assuming the status quo is projected to rise to 150 million by 2050. Prevention of age-related cognitive impairment in older persons with lifestyle interventions continues to garner evidence but whether this can combat underlying neurodegeneration is unknown. The Study of Mental Activity and Resistance Training (SMART) trial has previously reported within-training findings; the aim of this study was to investigate the long-term neurostructural and cognitive impact of resistance exercise in Mild Cognitive Impairment (MCI). For the first time we show that hippocampal subareas particularly susceptible to volume loss in Alzheimer's disease (AD) are protected by resistance exercise for up to one year after training. One hundred MCI participants were randomised to one of four training groups: (1) Combined high intensity progressive resistance and computerised cognitive training (PRT+CCT), (2) PRT+Sham CCT, (3) CCT+Sham PRT, (4) Sham physical+sham cognitive training (SHAM+SHAM). Physical, neuropsychological and MRI assessments were carried out at baseline, 6 months (directly after training) and 18 months from baseline (12 months after intervention cessation). Here we report neuro-structural and functional changes over the 18-month trial period and the association with global cognitive and executive function measures. PRT but not CCT or PRT+CCT led to global long-term cognitive improvements above SHAM intervention at 18-month follow-up. Furthermore, hippocampal subfields susceptible to atrophy in AD were protected by PRT revealing an elimination of long-term atrophy in the left subiculum, and attenuation of atrophy in left CA1 and dentate gyrus when compared to SHAM+SHAM (p = 0.023, p = 0.020 and p = 0.027). These neuroprotective effects mediated a significant portion of long-term cognitive benefits. By contrast, within-training posterior cingulate plasticity decayed after training cessation and was unrelated to long term cognitive benefits. Neither general physical activity levels nor fitness change over the 18-month period mediated hippocampal trajectory, demonstrating that enduring hippocampal subfield plasticity is not a simple reflection of post-training changes in fitness or physical activity participation. Notably, resting-state fMRI analysis revealed that both the hippocampus and posterior cingulate participate in a functional network that continued to be upregulated following intervention cessation. Multiple structural mechanisms may contribute to the long-term global cognitive benefit of resistance exercise, developing along different time courses but functionally linked. For the first time we show that 6 months of high intensity resistance exercise is capable of not only promoting better cognition in those with MCI, but also protecting AD-vulnerable hippocampal subfields from degeneration for at least 12 months post-intervention. These findings emphasise the therapeutic potential of resistance exercise; however, future work will need to establish just how long-lived these outcomes are and whether they are sufficient to delay dementia.
Collapse
Affiliation(s)
- Kathryn M Broadhouse
- Nola Thompson Centre for Advanced Imaging, Sunshine Coast Mind and Neuroscience Thompson Institute, University of the Sunshine Coast, QLD, Australia; Regenerative Neuroscience Group, Brain and Mind Centre and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.
| | - Maria Fiatarone Singh
- Physical Activity, Lifestyle, Ageing and Wellbeing Faculty Research Group, Faculty of Health Sciences and Sydney Medical School, The University of Sydney, Lidcombe, NSW, Australia; Hebrew SeniorLife and Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Chao Suo
- Regenerative Neuroscience Group, Brain and Mind Centre and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; School of Psychological Sciences and Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia
| | - Nicola Gates
- Regenerative Neuroscience Group, Brain and Mind Centre and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Wei Wen
- School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Dementia Collaborative Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Nidhi Jain
- Physical Activity, Lifestyle, Ageing and Wellbeing Faculty Research Group, Faculty of Health Sciences, The University of Sydney, Lidcombe, NSW, Australia
| | - Guy C Wilson
- Physical Activity, Lifestyle, Ageing and Wellbeing Faculty Research Group, Faculty of Health Sciences, The University of Sydney, Lidcombe, NSW, Australia
| | - Jacinda Meiklejohn
- Physical Activity, Lifestyle, Ageing and Wellbeing Faculty Research Group, Faculty of Health Sciences, The University of Sydney, Lidcombe, NSW, Australia
| | - Nalin Singh
- Physical Activity, Lifestyle, Ageing and Wellbeing Faculty Research Group, Faculty of Health Sciences, The University of Sydney, Lidcombe, NSW, Australia
| | - Bernhard T Baune
- Department of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Michael Baker
- Physical Activity, Lifestyle, Ageing and Wellbeing Faculty Research Group, Faculty of Health Sciences and Sydney Medical School, The University of Sydney, Lidcombe, NSW, Australia; School of Exercise Science, Australian Catholic University, Strathfield, NSW, Australia
| | - Nasim Foroughi
- Clinical and Rehabilitation Research Group, Faculty of Health Sciences, The University of Sydney, Lidcombe, NSW, Australia
| | - Yi Wang
- Clinical and Rehabilitation Research Group, Faculty of Health Sciences, The University of Sydney, Lidcombe, NSW, Australia; Department of Medicine and the Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Nicole Kochan
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Kevin Ashton
- Biomedical Sciences, Faculty of Health Sciences and Medicine, Bond University, QLD, Australia
| | - Matt Brown
- Institute of Health and Biomedical Innovation, Queensland University of Technology, QLD, Australia; King's College London National Institutes of Health Biomedical Research Centre, UK
| | - Zhixiu Li
- Institute of Health and Biomedical Innovation, Queensland University of Technology, QLD, Australia
| | - Yorgi Mavros
- Physical Activity, Lifestyle, Ageing and Wellbeing Faculty Research Group, Faculty of Health Sciences and Sydney Medical School, The University of Sydney, Lidcombe, NSW, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Michael J Valenzuela
- Regenerative Neuroscience Group, Brain and Mind Centre and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; School of Medical Sciences, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
15
|
Goukasian N, Porat S, Blanken A, Avila D, Zlatev D, Hurtz S, Hwang KS, Pierce J, Joshi SH, Woo E, Apostolova LG. Cognitive Correlates of Hippocampal Atrophy and Ventricular Enlargement in Adults with or without Mild Cognitive Impairment. Dement Geriatr Cogn Dis Extra 2019; 9:281-293. [PMID: 31572424 PMCID: PMC6751474 DOI: 10.1159/000490044] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 05/15/2018] [Indexed: 12/25/2022] Open
Abstract
We analyzed structural magnetic resonance imaging data from 58 cognitively normal and 101 mild cognitive impairment subjects. We used a general linear regression model to study the association between cognitive performance with hippocampal atrophy and ventricular enlargement using the radial distance method. Bilateral hippocampal atrophy was associated with baseline and longitudinal memory performance. Left hippocampal atrophy predicted longitudinal decline in visuospatial function. The multidomain ventricular analysis did not reveal any significant predictors.
Collapse
Affiliation(s)
- Naira Goukasian
- University of Vermont, Larner College of Medicine, Burlington, Vermont, USA
| | - Shai Porat
- Department of Psychology, University of Southern California, Los Angeles, California, USA
| | - Anna Blanken
- Department of Psychology, University of Southern California, Los Angeles, California, USA
| | - David Avila
- Irvine School of Medicine, University of California, Irvine, California, USA
| | - Dimitar Zlatev
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sona Hurtz
- Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Kristy S Hwang
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jonathan Pierce
- Department of Neurology, University of California, Los Angeles, California, USA
| | - Shantanu H Joshi
- Department of Neurology, University of California, Los Angeles, California, USA
| | - Ellen Woo
- Department of Neurology, University of California, Los Angeles, California, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
16
|
Barnes J, Bartlett JW, Wolk DA, van der Flier WM, Frost C. Disease Course Varies According to Age and Symptom Length in Alzheimer's Disease. J Alzheimers Dis 2019; 64:631-642. [PMID: 29914016 DOI: 10.3233/jad-170841] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Health-care professionals, patients, and families seek as much information as possible about prognosis for patients with Alzheimer's disease (AD); however, we do not yet have a robust understanding of how demographic factors predict prognosis. We evaluated associations between age at presentation, age of onset, and symptom length with cognitive decline as measured using the Mini-Mental State Examination (MMSE) and Clinical Dementia Rating sum-of-boxes (CDR-SOB) in a large dataset of AD patients. Age at presentation was associated with post-presentation decline in MMSE (p < 0.001), with younger patients showing faster decline. There was little evidence of an association with change in CDR-SOB. Symptom length, rather than age, was the strongest predictor of MMSE and CDR-SOB at presentation, with increasing symptom length associated with worse outcomes. The evidence that younger AD patients have a more aggressive disease course implies that early diagnosis is essential.
Collapse
Affiliation(s)
- Josephine Barnes
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | | | - David A Wolk
- Penn Memory Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Wiesje M van der Flier
- Alzheimer Center, Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Department of Epidemiology and Biostatistics, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Chris Frost
- Department of Medical Statistics, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
17
|
Kueper JK, Speechley M, Montero-Odasso M. The Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog): Modifications and Responsiveness in Pre-Dementia Populations. A Narrative Review. J Alzheimers Dis 2019; 63:423-444. [PMID: 29660938 PMCID: PMC5929311 DOI: 10.3233/jad-170991] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Alzheimer’s Disease Assessment Scale–Cognitive Subscale (ADAS-Cog) was developed in the 1980s to assess the level of cognitive dysfunction in Alzheimer’s disease. Advancements in the research field have shifted focus toward pre-dementia populations, and use of the ADAS-Cog has extended into these pre-dementia studies despite concerns about its ability to detect important changes at these milder stages of disease progression. If the ADAS-Cog cannot detect important changes, our understanding of pre-dementia disease progression may be compromised and trials may incorrectly conclude that a novel treatment approach is not beneficial. The purpose of this review was to assess the performance of the ADAS-Cog in pre-dementia populations, and to review all modifications that have been made to the ADAS-Cog to improve its measurement performance in dementia or pre-dementia populations. The contents of this review are based on bibliographic searches of electronic databases to locate all studies using the ADAS-Cog in pre-dementia samples or subsamples, and to locate all modified versions. Citations from relevant articles were also consulted. Overall, our results suggest the original ADAS-Cog is not an optimal outcome measure for pre-dementia studies; however, given the prominence of the ADAS-Cog, care must be taken when considering the use of alternative outcome measures. Thirty-one modified versions of the ADAS-Cog were found. Modification approaches that appear most beneficial include altering scoring methodology or adding tests of memory, executive function, and/or daily functioning. Although modifications improve the performance of the ADAS-Cog, this is at the cost of introducing heterogeneity that may limit between-study comparison.
Collapse
Affiliation(s)
- Jacqueline K Kueper
- Department of Epidemiology and Biostatistics, The University of Western Ontario, London, ON, Canada
| | - Mark Speechley
- Department of Epidemiology and Biostatistics, The University of Western Ontario, London, ON, Canada.,Schulich Interfaculty Program in Public Health, The University of Western Ontario, London, ON, Canada
| | - Manuel Montero-Odasso
- Department of Epidemiology and Biostatistics, The University of Western Ontario, London, ON, Canada.,Department of Medicine, Division of Geriatric Medicine, The University of Western Ontario, London, ON, Canada.,Gait and Brain Lab, Parkwood Institute, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
18
|
Niemantsverdriet E, Ribbens A, Bastin C, Benoit F, Bergmans B, Bier JC, Bladt R, Claes L, De Deyn PP, Deryck O, Hanseeuw B, Ivanoiu A, Lemper JC, Mormont E, Picard G, Salmon E, Segers K, Sieben A, Smeets D, Struyfs H, Thiery E, Tournoy J, Triau E, Vanbinst AM, Versijpt J, Bjerke M, Engelborghs S. A Retrospective Belgian Multi-Center MRI Biomarker Study in Alzheimer's Disease (REMEMBER). J Alzheimers Dis 2019; 63:1509-1522. [PMID: 29782314 PMCID: PMC6004934 DOI: 10.3233/jad-171140] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background: Magnetic resonance imaging (MRI) acquisition/processing techniques assess brain volumes to explore neurodegeneration in Alzheimer’s disease (AD). Objective: We examined the clinical utility of MSmetrix and investigated if automated MRI volumes could discriminate between groups covering the AD continuum and could be used as a predictor for clinical progression. Methods: The Belgian Dementia Council initiated a retrospective, multi-center study and analyzed whole brain (WB), grey matter (GM), white matter (WM), cerebrospinal fluid (CSF), cortical GM (CGM) volumes, and WM hyperintensities (WMH) using MSmetrix in the AD continuum. Baseline (n = 887) and follow-up (FU, n = 95) T1-weighted brain MRIs and time-linked neuropsychological data were available. Results: The cohort consisted of cognitively healthy controls (HC, n = 93), subjective cognitive decline (n = 102), mild cognitive impairment (MCI, n = 379), and AD dementia (n = 313). Baseline WB and GM volumes could accurately discriminate between clinical diagnostic groups and were significantly decreased with increasing cognitive impairment. MCI patients had a significantly larger change in WB, GM, and CGM volumes based on two MRIs (n = 95) compared to HC (FU>24months, p = 0.020). Linear regression models showed that baseline atrophy of WB, GM, CGM, and increased CSF volumes predicted cognitive impairment. Conclusion: WB and GM volumes extracted by MSmetrix could be used to define the clinical spectrum of AD accurately and along with CGM, they are able to predict cognitive impairment based on (decline in) MMSE scores. Therefore, MSmetrix can support clinicians in their diagnostic decisions, is able to detect clinical disease progression, and is of help to stratify populations for clinical trials.
Collapse
Affiliation(s)
- Ellis Niemantsverdriet
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | | | - Christine Bastin
- GIGA Cyclotron Research Centre in vivo Imaging, University of Liège, Liège, Belgium
| | - Florence Benoit
- Department of Geriatrics, Centre Hospitalier Universitaire (CHU) Brugmann, Brussels, Belgium
| | - Bruno Bergmans
- Department of Neurology and Center for Cognitive Disorders, AZ Sint-Jan Brugge-Oostende AV, Brugge, Belgium
| | | | - Roxanne Bladt
- Department of Radiology, Vrije Universiteit Brussel (VUB), UZ Brussel, Brussels, Belgium
| | | | - Peter Paul De Deyn
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Olivier Deryck
- Department of Neurology and Center for Cognitive Disorders, AZ Sint-Jan Brugge-Oostende AV, Brugge, Belgium
| | - Bernard Hanseeuw
- Department of Neurology, Cliniques Universitaires St Luc and Institute of Neuroscience, Université catholique de Louvain, Woluwe-Saint-Lambert (Brussels), Belgium
| | - Adrian Ivanoiu
- Department of Neurology, Cliniques Universitaires St Luc and Institute of Neuroscience, Université catholique de Louvain, Woluwe-Saint-Lambert (Brussels), Belgium
| | - Jean-Claude Lemper
- Department of Geriatrics, UZ Brussel, Brussels, Belgium.,Silva medical Scheutbos, Molenbeek-Saint-Jean (Brussels), Belgium
| | - Eric Mormont
- Department of Neurology, Centre Hospitalier Universitaire (CHU) Namur, Université catholique de Louvain, Yvoir, Belgium.,Université catholique de Louvain, Institute of Neuroscience (IoNS), Louvain-la-Neuve (Brussels), Belgium
| | - Gaëtane Picard
- Department of Neurology, Clinique Saint-Pierre, Ottignies, Belgium
| | - Eric Salmon
- GIGA Cyclotron Research Centre in vivo Imaging, University of Liège, Liège, Belgium.,Department of Neurology, Memory Clinic, Centre Hospitalier Universitaire (CHU) Liège, Liège, Belgium
| | - Kurt Segers
- Department of Neurology, Centre Hospitalier Universitaire (CHU) Brugmann, Brussels, Belgium
| | - Anne Sieben
- Department of Neurology, University Hospital Ghent, Ghent University, Ghent, Belgium
| | | | - Hanne Struyfs
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Evert Thiery
- Department of Neurology, University Hospital Ghent, Ghent University, Ghent, Belgium
| | - Jos Tournoy
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium.,Geriatric Medicine and Memory Clinic, University Hospital Leuven, Leuven, Belgium
| | | | - Anne-Marie Vanbinst
- Department of Radiology, Vrije Universiteit Brussel (VUB), UZ Brussel, Brussels, Belgium
| | - Jan Versijpt
- Department of Neurology, Vrije Universiteit Brussel (VUB), UZ Brussel, Brussels, Belgium
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| |
Collapse
|
19
|
Munir M, Ursenbach J, Reid M, Gupta Sah R, Wang M, Sitaram A, Aftab A, Tariq S, Zamboni G, Griffanti L, Smith EE, Frayne R, Sajobi TT, Coutts SB, d'Esterre CD, Barber PA. Longitudinal Brain Atrophy Rates in Transient Ischemic Attack and Minor Ischemic Stroke Patients and Cognitive Profiles. Front Neurol 2019; 10:18. [PMID: 30837927 PMCID: PMC6389669 DOI: 10.3389/fneur.2019.00018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/07/2019] [Indexed: 02/04/2023] Open
Abstract
Introduction: Patients with transient ischemic attack (TIA) and minor stroke demonstrate cognitive impairment, and a four-fold risk of late-life dementia. Aim: To study the extent to which the rates of brain volume loss in TIA patients differ from healthy controls and how they are correlated with cognitive impairment. Methods: TIA or minor stroke patients were tested with a neuropsychological battery and underwent T1 weighted volumetric magnetic resonance imaging scans at fixed intervals over a 3 years period. Linear mixed effects regression models were used to compare brain atrophy rates between groups, and to determine the relationship between atrophy rates and cognitive function in TIA and minor stroke patients. Results: Whole brain atrophy rates were calculated for the TIA and minor stroke patients; n = 38 between 24 h and 18 months, and n = 68 participants between 18 and 36 months, and were compared to healthy controls. TIA and minor stroke patients demonstrated a significantly higher whole brain atrophy rate than healthy controls over a 3 years interval (p = 0.043). Diabetes (p = 0.012) independently predicted higher atrophy rate across groups. There was a relationship between higher rates of brain atrophy and processing speed (composite P = 0.047 and digit symbol coding P = 0.02), but there was no relationship with brain atrophy rates and memory or executive composite scores or individual cognitive tests for language (Boston naming, memory recall, verbal fluency or Trails A or B score). Conclusion: TIA and minor stroke patients experience a significantly higher rate of whole brain atrophy. In this cohort of TIA and minor stroke patients changes in brain volume over time precede cognitive decline.
Collapse
Affiliation(s)
- Muhammad Munir
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada
| | - Jake Ursenbach
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada
| | - Meaghan Reid
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada
| | - Rani Gupta Sah
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada.,Department of Radiology, Foothills Medical Centre, Calgary, AB, Canada.,Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Meng Wang
- Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | - Amith Sitaram
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada.,Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Arooj Aftab
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada
| | - Sana Tariq
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada
| | - Giovanna Zamboni
- Nuffield Department of Clinical Neurosciences, FMRIB Centre, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ludovica Griffanti
- Nuffield Department of Clinical Neurosciences, FMRIB Centre, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Eric E Smith
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Richard Frayne
- Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada.,Department of Radiology, Foothills Medical Centre, Calgary, AB, Canada.,Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Tolulope T Sajobi
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | - Shelagh B Coutts
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada.,Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Christopher D d'Esterre
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Seaman Family MR Center, Foothills Medical Centre, Calgary, AB, Canada
| | - Philip A Barber
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, Calgary, AB, Canada.,Department of Radiology, Foothills Medical Centre, Calgary, AB, Canada.,Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
20
|
Lee H, Nakamura K, Narayanan S, Brown RA, Arnold DL. Estimating and accounting for the effect of MRI scanner changes on longitudinal whole-brain volume change measurements. Neuroimage 2019; 184:555-565. [DOI: 10.1016/j.neuroimage.2018.09.062] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/10/2018] [Accepted: 09/21/2018] [Indexed: 01/18/2023] Open
|
21
|
Tariq S, d’Esterre CD, Sajobi TT, Smith EE, Longman RS, Frayne R, Coutts SB, Forkert ND, Barber PA. A longitudinal magnetic resonance imaging study of neurodegenerative and small vessel disease, and clinical cognitive trajectories in non demented patients with transient ischemic attack: the PREVENT study. BMC Geriatr 2018; 18:163. [PMID: 30012102 PMCID: PMC6048817 DOI: 10.1186/s12877-018-0858-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 07/09/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Late-life cognitive decline, caused by progressive neuronal loss leading to brain atrophy years before symptoms are detected, is expected to double in Canada over the next two decades. Cognitive impairment in late life is attributed to vascular and lifestyle related risk factors in mid-life in a substantial proportion of cases (50%), thereby providing an opportunity for effective prevention of cognitive decline if incipient disease is detected earlier. Patients presenting with transient ischemic attack (TIA) commonly display some degree of cognitive impairment and are at a 4-fold increased risk of dementia. In the Predementia Neuroimaging of Transient Ischemic Attack (PREVENT) study, we will address what disease processes (i.e., Alzheimer's vs. vascular disease) lead to neurodegeneration, brain atrophy, and cognitive decline, and whether imaging measurements of brain iron accumulation using quantitative susceptibility mapping predicts subsequent brain atrophy and cognitive decline. METHODS A total of 440 subjects will be recruited for this study with 220 healthy subjects and 220 TIA patients. Early Alzheimer's pathology will be determined by cerebrospinal fluid samples (including tau, a marker of neuronal injury, and amyloid β1-42) and by MR measurements of iron accumulation, a marker for Alzheimer's-related neurodegeneration. Small vessel disease will be identified by changes in white matter lesion volume. Predictors of advanced rates of cerebral and hippocampal atrophy at 1 and 3 years will include in vivo Alzheimer's disease pathology markers, and MRI measurements of brain iron accumulation and small vessel disease. Clinical and cognitive function will be assessed annually post-baseline for a period of 5-years using a clinical questionnaire and a battery of neuropsychological tests, respectively. DISCUSSION The PREVENT study expects to demonstrate that TIA patients have increased early progressive rates of cerebral brain atrophy after TIA, before cognitive decline can be clinically detected. By developing and optimizing high-level machine learning models based on clinical data, image-based (quantitative susceptibility mapping, regional brain, and white matter lesion volumes) features, and cerebrospinal fluid biomarkers, PREVENT will provide a timely opportunity to identify individuals at greatest risk of late-life cognitive decline early in the course of disease, supporting future therapeutic strategies for the promotion of healthy aging.
Collapse
Affiliation(s)
- Sana Tariq
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, 1403 - 29 Street NW, Calgary, AB Canada
- Seaman Family MR Center, Foothills Medical Centre, 1403 29th Street NW, Calgary, AB Canada
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Christopher D. d’Esterre
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, 1403 - 29 Street NW, Calgary, AB Canada
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Tolulope T. Sajobi
- Seaman Family MR Center, Foothills Medical Centre, 1403 29th Street NW, Calgary, AB Canada
- Department of Community Health Sciences & O’Brien Institute for Public Health, University of Calgary, 3280 Hospital Drive NW, Calgary, AB Canada
| | - Eric E. Smith
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, 1403 - 29 Street NW, Calgary, AB Canada
- Seaman Family MR Center, Foothills Medical Centre, 1403 29th Street NW, Calgary, AB Canada
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Richard Stewart Longman
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Richard Frayne
- Seaman Family MR Center, Foothills Medical Centre, 1403 29th Street NW, Calgary, AB Canada
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
- Department of Radiology and Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB Canada
| | - Shelagh B. Coutts
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, 1403 - 29 Street NW, Calgary, AB Canada
- Seaman Family MR Center, Foothills Medical Centre, 1403 29th Street NW, Calgary, AB Canada
- Department of Community Health Sciences & O’Brien Institute for Public Health, University of Calgary, 3280 Hospital Drive NW, Calgary, AB Canada
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Nils D. Forkert
- Seaman Family MR Center, Foothills Medical Centre, 1403 29th Street NW, Calgary, AB Canada
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
- Department of Radiology and Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB Canada
| | - Philip A. Barber
- Calgary Stroke Program, Department of Clinical Neurosciences, Foothills Medical Centre, 1403 - 29 Street NW, Calgary, AB Canada
- Seaman Family MR Center, Foothills Medical Centre, 1403 29th Street NW, Calgary, AB Canada
- Hotchkiss Brain Institute, Foothills Medical Center, Room 1A10 Health Research Innovation Center, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| |
Collapse
|
22
|
Kaneko T, Mitsui T, Kaneko K, Kadoya M. New longitudinal Visual Rating Scale Identifies Structural Alterations in People with Mild Cognitive Impairment and Those who are Cognitively Normal. INT J GERONTOL 2018. [DOI: 10.1016/j.ijge.2018.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
23
|
Kim H. Detection of severity in Alzheimer's disease (AD) using computational modeling. Bioinformation 2018; 14:259-264. [PMID: 30108425 PMCID: PMC6077821 DOI: 10.6026/97320630014259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 05/09/2018] [Accepted: 05/19/2018] [Indexed: 01/08/2023] Open
Abstract
The prevalent cause of dementia - Alzheimer's disease (AD) is characterized by an early cholinergic deficit that is in part responsible for the cognitive deficits (especially memory and attention defects). Prolonged AD leads to moderate-to-severe AD, which is one of the leading causes of death. Placebo-controlled, randomized clinical trials have shown significant effects of Acetyl cholin esterase inhibitors (ChEIs) on function, cognition, activities of daily living (ADL) and behavioral symptoms in patients. Studies have shown comparable effects for ChEIs in patients with moderate-to-severe or mild AD. Setting a fixed measurement (e.g. a Mini-Mental State Examination score, as a 'when to stop treatment limit) for the disease is not clinically rational. Detection of changed regional cerebral blood flow in mild cognitive impairment and early AD by perfusion-weighted magnetic resonance imaging has been a challenge. The utility of perfusion-weighted magnetic resonance imaging (PW-MRI) for detecting changes in regional cerebral blood flow (rCBF) in patients with mild cognitive impairment (MCI) and early AD was evaluated. We describe a computer aided prediction model to determine the severity of AD using known data in literature. We designed an automated system for the determination of AD severity. It is used to predict the clinical cases and conditions with disagreements from specialist. The model described is useful in clinical practice to validate diagnosis.
Collapse
Affiliation(s)
- Hyunjo Kim
- Department of Life Science, University of Gachon, Seungnam, Kyeonggido, Korea
- Medical Informatics Department of Ajou Medical Center, South Korea
| |
Collapse
|
24
|
Morphological Biomarker Differentiating MCI Converters from Nonconverters: Longitudinal Evidence Based on Hemispheric Asymmetry. Behav Neurol 2018; 2018:3954101. [PMID: 29755611 PMCID: PMC5884406 DOI: 10.1155/2018/3954101] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/11/2017] [Accepted: 01/03/2018] [Indexed: 11/17/2022] Open
Abstract
Identifying subjects with mild cognitive impairment (MCI) who may probably progress to Alzheimer's disease (AD) is important for better understanding the disease mechanisms and facilitating early treatments. In addition to the direct volumetric and thickness measurement based on high-resolution magnetic resonance imaging (MRI), hemispheric asymmetry could be a potential index to detect morphological variations in MCI patients with a high risk of conversion to AD. The present study collected a set of longitudinal MRI data from 53 MCI converters and nonconverters and investigated the asymmetry differences between groups. Asymmetry variation was observed in the medial temporal lobe, especially in the entorhinal cortex, between converters and nonconverters 3 years before the former developed AD. The proposed asymmetry analysis was observed to be sensitive to detect morphological changes between groups as compared to the methods of voxel-based morphometry (VBM) and thickness measurement. Hemispheric asymmetry in specific brain regions as a neuroimaging biomarker can provide helpful information for prediction of MCI conversion.
Collapse
|
25
|
Fiford CM, Ridgway GR, Cash DM, Modat M, Nicholas J, Manning EN, Malone IB, Biessels GJ, Ourselin S, Carmichael OT, Cardoso MJ, Barnes J. Patterns of progressive atrophy vary with age in Alzheimer's disease patients. Neurobiol Aging 2018; 63:22-32. [PMID: 29220823 PMCID: PMC5805840 DOI: 10.1016/j.neurobiolaging.2017.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/14/2017] [Accepted: 11/06/2017] [Indexed: 01/18/2023]
Abstract
Age is not only the greatest risk factor for Alzheimer's disease (AD) but also a key modifier of disease presentation and progression. Here, we investigate how longitudinal atrophy patterns vary with age in mild cognitive impairment (MCI) and AD. Data comprised serial longitudinal 1.5-T magnetic resonance imaging scans from 153 AD, 339 MCI, and 191 control subjects. Voxel-wise maps of longitudinal volume change were obtained and aligned across subjects. Local volume change was then modeled in terms of diagnostic group and an interaction between group and age, adjusted for total intracranial volume, white-matter hyperintensity volume, and apolipoprotein E genotype. Results were significant at p < 0.05 with family-wise error correction for multiple comparisons. An age-by-group interaction revealed that younger AD patients had significantly faster atrophy rates in the bilateral precuneus, parietal, and superior temporal lobes. These results suggest younger AD patients have predominantly posterior progressive atrophy, unexplained by white-matter hyperintensity, apolipoprotein E, or total intracranial volume. Clinical trials may benefit from adapting outcome measures for patient groups with lower average ages, to capture progressive atrophy in posterior cortices.
Collapse
Affiliation(s)
- Cassidy M Fiford
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.
| | - Gerard R Ridgway
- FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Wellcome Trust Centre for Neuroimaging, 12 Queen Square, London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Marc Modat
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | | | - Emily N Manning
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Ian B Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sebastien Ourselin
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | | | - M Jorge Cardoso
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Josephine Barnes
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| |
Collapse
|
26
|
Leandrou S, Petroudi S, Kyriacou PA, Reyes-Aldasoro CC, Pattichis CS. Quantitative MRI Brain Studies in Mild Cognitive Impairment and Alzheimer's Disease: A Methodological Review. IEEE Rev Biomed Eng 2018; 11:97-111. [PMID: 29994606 DOI: 10.1109/rbme.2018.2796598] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Classifying and predicting Alzheimer's disease (AD) in individuals with memory disorders through clinical and psychometric assessment is challenging, especially in mild cognitive impairment (MCI) subjects. Quantitative structural magnetic resonance imaging acquisition methods in combination with computer-aided diagnosis are currently being used for the assessment of AD. These acquisitions methods include voxel-based morphometry, volumetric measurements in specific regions of interest (ROIs), cortical thickness measurements, shape analysis, and texture analysis. This review evaluates the aforementioned methods in the classification of cases into one of the following three groups: normal controls, MCI, and AD subjects. Furthermore, the performance of the methods is assessed on the prediction of conversion from MCI to AD. In parallel, it is also assessed which ROIs are preferred in both classification and prognosis through the different states of the disease. Structural changes in the early stages of the disease are more pronounced in the medial temporal lobe, especially in the entorhinal cortex, whereas with disease progression, both entorhinal cortex and hippocampus offer similar discriminative power. However, for the conversion from MCI subjects to AD, entorhinal cortex provides better predictive accuracies rather than other structures, such as the hippocampus.
Collapse
|
27
|
Ota K, Oishi N, Ito K, Fukuyama H. Prediction of Alzheimer's Disease in Amnestic Mild Cognitive Impairment Subtypes: Stratification Based on Imaging Biomarkers. J Alzheimers Dis 2017; 52:1385-401. [PMID: 27079727 DOI: 10.3233/jad-160145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Prediction of progression to Alzheimer's disease (AD) in amnestic mild cognitive impairment (MCI) is challenging because of its heterogeneity. OBJECTIVE To evaluate a stratification method on different cohorts and to investigate whether stratification in amnestic MCI could improve prediction accuracy. METHODS We identified 80 and 79 patients with amnestic MCI from different cohorts, respectively. They underwent baseline magnetic resonance imaging (MRI) and 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) scans. We performed hierarchical clustering with three imaging biomarkers: Brain volume on MRI, left hippocampus grey matter loss on MRI, and left inferior temporal gyrus glucose hypometabolism on FDG-PET. Regions-of-interest for biomarkers were defined by the Automated Anatomical Labeling atlas. We performed voxel-wise statistical parametric mapping to explore differences between clusters in patterns of grey matter loss and glucose hypometabolism. We compared time to progression using an interval-censored parametric model. We evaluated predictive performance using logistic regression. RESULTS Similar clusters were found in different cohorts. MCI1 had the healthiest biomarker profile of cognitive performance and imaging biomarkers. MCI2 had cognitive performance and MRI measures intermediate between those of nonconverters and converters. MCI3 showed the severest reduction in brain volume and left hippocampal atrophy. MCI4 showed remarkable glucose hypometabolism in the left inferior temporal gyrus, and also demonstrated significant decreases in most cognitive scores, including non-memory functions. MCI4 showed the highest risk for progression. The prediction of progression of MCI2 especially benefited from the stratification. CONCLUSION Stratification with imaging biomarkers in amnestic MCI can be a good approach for improving predictive performance.
Collapse
Affiliation(s)
- Kenichi Ota
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Naoya Oishi
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kengo Ito
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Hidenao Fukuyama
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | | | | |
Collapse
|
28
|
Falahati F, Ferreira D, Muehlboeck JS, Eriksdotter M, Simmons A, Wahlund LO, Westman E. Monitoring disease progression in mild cognitive impairment: Associations between atrophy patterns, cognition, APOE and amyloid. NEUROIMAGE-CLINICAL 2017; 16:418-428. [PMID: 28879083 PMCID: PMC5573795 DOI: 10.1016/j.nicl.2017.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 08/03/2017] [Accepted: 08/12/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND A disease severity index (SI) for Alzheimer's disease (AD) has been proposed that summarizes MRI-derived structural measures into a single score using multivariate data analysis. OBJECTIVES To longitudinally evaluate the use of the SI to monitor disease progression and predict future progression to AD in mild cognitive impairment (MCI). Further, to investigate the association between longitudinal change in the SI and cognitive impairment, Apolipoprotein E (APOE) genotype as well as the levels of cerebrospinal fluid amyloid-beta 1-42 (Aβ) peptide. METHODS The dataset included 195 AD, 145 MCI and 228 control subjects with annual follow-up for three years, where 70 MCI subjects progressed to AD (MCI-p). For each subject the SI was generated at baseline and follow-ups using 55 regional cortical thickness and subcortical volumes measures that extracted by the FreeSurfer longitudinal stream. RESULTS MCI-p subjects had a faster increase of the SI over time (p < 0.001). A higher SI at baseline in MCI-p was related to progression to AD at earlier follow-ups (p < 0.001) and worse cognitive impairment (p < 0.001). AD-like MCI patients with the APOE ε4 allele and abnormal Aβ levels had a faster increase of the SI, independently (p = 0.003 and p = 0.004). CONCLUSIONS Longitudinal changes in the SI reflect structural brain changes and can identify MCI patients at risk of progression to AD. Disease-related brain structural changes are influenced independently by APOE genotype and amyloid pathology. The SI has the potential to be used as a sensitive tool to predict future dementia, monitor disease progression as well as an outcome measure for clinical trials.
Collapse
Affiliation(s)
- Farshad Falahati
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - J-Sebastian Muehlboeck
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Andrew Simmons
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience; King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health, London, UK.,NIHR Biomedical Research Unit for Dementia, London, UK
| | - Lars-Olof Wahlund
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience; King's College London, London, UK
| |
Collapse
|
29
|
Nirogi R, Ajjala DR, Aleti R, Rayapati L, Pantangi HR, Boggavarapu RK, Padala NSP. Development and validation of sensitive LC-MS/MS method for the quantification of SUVN-502 and its metabolite and its application for first in human pharmacokinetic study. J Pharm Biomed Anal 2017; 145:423-430. [PMID: 28734271 DOI: 10.1016/j.jpba.2017.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 12/31/2022]
Abstract
A sensitive and rapid LC-MS/MS method was developed and validated for the quantification of SUVN-502 and M1 of SUVN-502, a 5-HT6 receptor antagonist for the treatment of dementia associated with Alzheimer's disease. Following solid-phase extraction, SUVN-502 and M1 of SUVN-502 and IS were eluted with 10mM ammonium acetate (pH 4.0) and acetonitrile using a rapid gradient program on reverse phase column. Multiple reaction monitoring mode was used to monitor the respective transitions of m/z 478.2→377.7 for SUVN-502 and m/z 464.1→377.7 for M1 of SUVN-502. The assay exhibited a linear dynamic range of 10-10000pg/mL for SUVN-502 and 20-20000pg/mL for M1 of SUVN-502 in human plasma. Acceptable precision and accuracy were obtained for concentrations over the standard curve range. The within batch accuracy and precision were within acceptable limits. All the other validation parameters were within the acceptable limits. The validated method was applied to analyze human plasma samples obtained from a human pharmacokinetic study consisting single and multiple ascending doses.
Collapse
Affiliation(s)
- Ramakrishna Nirogi
- Biopharmaceutical Research, Suven Life Sciences Ltd., Serene Chambers, Road - 5, Avenue - 7, Banjara Hills, Hyderabad 500034, India.
| | - Devender Reddy Ajjala
- Biopharmaceutical Research, Suven Life Sciences Ltd., Serene Chambers, Road - 5, Avenue - 7, Banjara Hills, Hyderabad 500034, India
| | - Raghupathi Aleti
- Biopharmaceutical Research, Suven Life Sciences Ltd., Serene Chambers, Road - 5, Avenue - 7, Banjara Hills, Hyderabad 500034, India
| | - Lakshmiprasanna Rayapati
- Biopharmaceutical Research, Suven Life Sciences Ltd., Serene Chambers, Road - 5, Avenue - 7, Banjara Hills, Hyderabad 500034, India
| | - Hanumanth Rao Pantangi
- Biopharmaceutical Research, Suven Life Sciences Ltd., Serene Chambers, Road - 5, Avenue - 7, Banjara Hills, Hyderabad 500034, India
| | - Rajesh Kumar Boggavarapu
- Biopharmaceutical Research, Suven Life Sciences Ltd., Serene Chambers, Road - 5, Avenue - 7, Banjara Hills, Hyderabad 500034, India
| | - Naga Surya Prakash Padala
- Biopharmaceutical Research, Suven Life Sciences Ltd., Serene Chambers, Road - 5, Avenue - 7, Banjara Hills, Hyderabad 500034, India
| |
Collapse
|
30
|
Orimaye SO, Wong JSM, Golden KJ, Wong CP, Soyiri IN. Predicting probable Alzheimer's disease using linguistic deficits and biomarkers. BMC Bioinformatics 2017; 18:34. [PMID: 28088191 PMCID: PMC5237556 DOI: 10.1186/s12859-016-1456-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 12/31/2016] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND The manual diagnosis of neurodegenerative disorders such as Alzheimer's disease (AD) and related Dementias has been a challenge. Currently, these disorders are diagnosed using specific clinical diagnostic criteria and neuropsychological examinations. The use of several Machine Learning algorithms to build automated diagnostic models using low-level linguistic features resulting from verbal utterances could aid diagnosis of patients with probable AD from a large population. For this purpose, we developed different Machine Learning models on the DementiaBank language transcript clinical dataset, consisting of 99 patients with probable AD and 99 healthy controls. RESULTS Our models learned several syntactic, lexical, and n-gram linguistic biomarkers to distinguish the probable AD group from the healthy group. In contrast to the healthy group, we found that the probable AD patients had significantly less usage of syntactic components and significantly higher usage of lexical components in their language. Also, we observed a significant difference in the use of n-grams as the healthy group were able to identify and make sense of more objects in their n-grams than the probable AD group. As such, our best diagnostic model significantly distinguished the probable AD group from the healthy elderly group with a better Area Under the Receiving Operating Characteristics Curve (AUC) using the Support Vector Machines (SVM). CONCLUSIONS Experimental and statistical evaluations suggest that using ML algorithms for learning linguistic biomarkers from the verbal utterances of elderly individuals could help the clinical diagnosis of probable AD. We emphasise that the best ML model for predicting the disease group combines significant syntactic, lexical and top n-gram features. However, there is a need to train the diagnostic models on larger datasets, which could lead to a better AUC and clinical diagnosis of probable AD.
Collapse
Affiliation(s)
- Sylvester O. Orimaye
- Intelligent Health Research Group, School of Information Technology, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Malaysia
| | - Jojo S-M. Wong
- Intelligent Health Research Group, School of Information Technology, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Malaysia
| | - Karen J. Golden
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Malaysia
| | - Chee P. Wong
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Malaysia
| | - Ireneous N. Soyiri
- Centre for Medical Informatics, Usher Institute for Population Health Sciences & Informatics, The University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG UK
| |
Collapse
|
31
|
Ertekin T, Acer N, Köseoğlu E, Zararsız G, Sönmez A, Gümüş K, Kurtoğlu E. Total intracranial and lateral ventricle volumes measurement in Alzheimer's disease: A methodological study. J Clin Neurosci 2016; 34:133-139. [PMID: 27475320 DOI: 10.1016/j.jocn.2016.05.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/07/2016] [Accepted: 05/21/2016] [Indexed: 11/17/2022]
Abstract
Measuring of brain and its compartments' sizes from magnetic resonance (MR) images is an effective way to assess disease progression in neurodegenerative disorders, particularly Alzheimer's disease (AD). The objective of this study was to compare total intracranial volume (TIV) and lateral ventricle volume (LVV) in patients with Alzheimer's disease with those in elderly control subjects, and to compare an automated method (automatic lateral ventricle delineation [ALVIN]) and a manual method (ImageJ). MRI of the brain was performed on 20 patients with Alzheimer's disease and 18 control subjects. The TIV was calculated by a manual method and the LVV was calculated by using two methods: an automated and manual method. We found a significant increase in LVVs in Alzheimer's disease patients compared to control subjects, but no difference in TIV between the two groups. A perfect agreement, with 0.989 (0.973-0.996) intraclass correlation coefficient (ICC) and 0.978 (0.946-0.991) concordance correlation coefficient (CCC), was observed between the manual and automatic lateral ventricle measurements in Alzheimer patients. The results revealed that LVV measure has predictive performance in AD. We demonstrated that ALVIN and ImageJ are both effective in determining lateral ventricular volume, providing an objective tool for quantitative assessment of AD.
Collapse
Affiliation(s)
- Tolga Ertekin
- Department of Anatomy, Medical Faculty, Erciyes University, Köşk, Talas Blv, Kayseri 38039, Turkey.
| | - Niyazi Acer
- Department of Anatomy, Medical Faculty, Erciyes University, Köşk, Talas Blv, Kayseri 38039, Turkey
| | - Emel Köseoğlu
- Department of Neurology, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Gökmen Zararsız
- Department of Biostatistics, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Ali Sönmez
- Department of Neurology, Elbistan State Hospital, Kahramanmaraş, Turkey
| | - Kazım Gümüş
- Department of Biophysics, Medical Faculty, Erciyes University, Kayseri, Turkey
| | - Erdal Kurtoğlu
- Department of Anatomy, Medical Faculty, Erciyes University, Köşk, Talas Blv, Kayseri 38039, Turkey
| |
Collapse
|
32
|
Cover KS, van Schijndel RA, Versteeg A, Leung KK, Mulder ER, Jong RA, Visser PJ, Redolfi A, Revillard J, Grenier B, Manset D, Damangir S, Bosco P, Vrenken H, van Dijk BW, Frisoni GB, Barkhof F. Reproducibility of hippocampal atrophy rates measured with manual, FreeSurfer, AdaBoost, FSL/FIRST and the MAPS-HBSI methods in Alzheimer's disease. Psychiatry Res Neuroimaging 2016; 252:26-35. [PMID: 27179313 DOI: 10.1016/j.pscychresns.2016.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 02/16/2016] [Accepted: 04/08/2016] [Indexed: 11/23/2022]
Abstract
The purpose of this study is to assess the reproducibility of hippocampal atrophy rate measurements of commonly used fully-automated algorithms in Alzheimer disease (AD). The reproducibility of hippocampal atrophy rate for FSL/FIRST, AdaBoost, FreeSurfer, MAPS independently and MAPS combined with the boundary shift integral (MAPS-HBSI) were calculated. Back-to-back (BTB) 3D T1-weighted MPRAGE MRI from the Alzheimer's Disease Neuroimaging Initiative (ADNI1) study at baseline and year one were used. Analysis on 3 groups of subjects was performed - 562 subjects at 1.5T, a 75 subject group that also had manual segmentation and 111 subjects at 3T. A simple and novel statistical test based on the binomial distribution was used that handled outlying data points robustly. Median hippocampal atrophy rates were -1.1%/year for healthy controls, -3.0%/year for mildly cognitively impaired and -5.1%/year for AD subjects. The best reproducibility was observed for MAPS-HBSI (1.3%), while the other methods tested had reproducibilities at least 50% higher at 1.5T and 3T which was statistically significant. For a clinical trial, MAPS-HBSI should require less than half the subjects of the other methods tested. All methods had good accuracy versus manual segmentation. The MAPS-HBSI method has substantially better reproducibility than the other methods considered.
Collapse
Affiliation(s)
- Keith S Cover
- VU University Medical Center, Amsterdam, Netherlands.
| | | | | | | | - Emma R Mulder
- VU University Medical Center, Amsterdam, Netherlands
| | - Remko A Jong
- VU University Medical Center, Amsterdam, Netherlands
| | | | | | | | | | | | | | - Paolo Bosco
- IRCCS San Giovanni di Dio Fatebenefratelli, Italy
| | - Hugo Vrenken
- VU University Medical Center, Amsterdam, Netherlands
| | | | - Giovanni B Frisoni
- IRCCS San Giovanni di Dio Fatebenefratelli, Italy; University Hospitals and University of Geneva, Switzerland
| | | |
Collapse
|
33
|
Alberdi A, Aztiria A, Basarab A. On the early diagnosis of Alzheimer's Disease from multimodal signals: A survey. Artif Intell Med 2016; 71:1-29. [PMID: 27506128 DOI: 10.1016/j.artmed.2016.06.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/23/2016] [Accepted: 06/07/2016] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The number of Alzheimer's Disease (AD) patients is increasing with increased life expectancy and 115.4 million people are expected to be affected in 2050. Unfortunately, AD is commonly diagnosed too late, when irreversible damages have been caused in the patient. OBJECTIVE An automatic, continuous and unobtrusive early AD detection method would be required to improve patients' life quality and avoid big healthcare costs. Thus, the objective of this survey is to review the multimodal signals that could be used in the development of such a system, emphasizing on the accuracy that they have shown up to date for AD detection. Some useful tools and specific issues towards this goal will also have to be reviewed. METHODS An extensive literature review was performed following a specific search strategy, inclusion criteria, data extraction and quality assessment in the Inspec, Compendex and PubMed databases. RESULTS This work reviews the extensive list of psychological, physiological, behavioural and cognitive measurements that could be used for AD detection. The most promising measurements seem to be magnetic resonance imaging (MRI) for AD vs control (CTL) discrimination with an 98.95% accuracy, while electroencephalogram (EEG) shows the best results for mild cognitive impairment (MCI) vs CTL (97.88%) and MCI vs AD distinction (94.05%). Available physiological and behavioural AD datasets are listed, as well as medical imaging analysis steps and neuroimaging processing toolboxes. Some issues such as "label noise" and multi-site data are discussed. CONCLUSIONS The development of an unobtrusive and transparent AD detection system should be based on a multimodal system in order to take full advantage of all kinds of symptoms, detect even the smallest changes and combine them, so as to detect AD as early as possible. Such a multimodal system might probably be based on physiological monitoring of MRI or EEG, as well as behavioural measurements like the ones proposed along the article. The mentioned AD datasets and image processing toolboxes are available for their use towards this goal. Issues like "label noise" and multi-site neuroimaging incompatibilities may also have to be overcome, but methods for this purpose are already available.
Collapse
Affiliation(s)
- Ane Alberdi
- Mondragon University, Electronics and Computing Department, Goiru Kalea, 2, Arrasate 20500, Spain.
| | - Asier Aztiria
- Mondragon University, Electronics and Computing Department, Goiru Kalea, 2, Arrasate 20500, Spain.
| | - Adrian Basarab
- Université de Toulouse, Institut de Recherche en Informatique de Toulouse, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5505, Université Paul Sabatier, 118 Route de Narbonne, 31062 Toulouse, France.
| |
Collapse
|
34
|
Andrews KA, Frost C, Modat M, Cardoso MJ, Rowe CC, Villemagne V, Fox NC, Ourselin S, Schott JM, Rowe CC, Villemagne V, Fox NC, Ourselin S, Schott JM. Acceleration of hippocampal atrophy rates in asymptomatic amyloidosis. Neurobiol Aging 2016; 39:99-107. [PMID: 26923406 DOI: 10.1016/j.neurobiolaging.2015.10.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 09/09/2015] [Accepted: 10/14/2015] [Indexed: 11/24/2022]
|
35
|
Cerebral atrophy in mild cognitive impairment: A systematic review with meta-analysis. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2015; 1:487-504. [PMID: 27239527 PMCID: PMC4879488 DOI: 10.1016/j.dadm.2015.11.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Although mild cognitive impairment (MCI) diagnosis is mainly based on cognitive assessment, reliable estimates of structural changes in specific brain regions, that could be contrasted against normal brain aging and inform diagnosis, are lacking. This study aimed to systematically review the literature reporting on MCI-related brain changes. METHODS The MEDLINE database was searched for studies investigating longitudinal structural changes in MCI. Studies with compatible data were included in the meta-analyses. A qualitative review was conducted for studies excluded from meta-analyses. RESULTS The analyses revealed a 2.2-fold higher volume loss in the hippocampus, 1.8-fold in the whole brain, and 1.5-fold in the entorhinal cortex in MCI participants. DISCUSSION Although the medial temporal lobe is likely to be more vulnerable to MCI pathology, atrophy in this brain area represents a relatively small proportion of whole brain loss, suggesting that future investigations are needed to identify the source of unaccounted volume loss in MCI.
Collapse
|
36
|
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Cedarbaum J, Donohue MC, Green RC, Harvey D, Jack CR, Jagust W, Morris JC, Petersen RC, Saykin AJ, Shaw L, Thompson PM, Toga AW, Trojanowski JQ. Impact of the Alzheimer's Disease Neuroimaging Initiative, 2004 to 2014. Alzheimers Dement 2015; 11:865-84. [PMID: 26194320 PMCID: PMC4659407 DOI: 10.1016/j.jalz.2015.04.005] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/04/2015] [Accepted: 04/23/2015] [Indexed: 01/18/2023]
Abstract
INTRODUCTION The Alzheimer's Disease Neuroimaging Initiative (ADNI) was established in 2004 to facilitate the development of effective treatments for Alzheimer's disease (AD) by validating biomarkers for AD clinical trials. METHODS We searched for ADNI publications using established methods. RESULTS ADNI has (1) developed standardized biomarkers for use in clinical trial subject selection and as surrogate outcome measures; (2) standardized protocols for use across multiple centers; (3) initiated worldwide ADNI; (4) inspired initiatives investigating traumatic brain injury and post-traumatic stress disorder in military populations, and depression, respectively, as an AD risk factor; (5) acted as a data-sharing model; (6) generated data used in over 600 publications, leading to the identification of novel AD risk alleles, and an understanding of the relationship between biomarkers and AD progression; and (7) inspired other public-private partnerships developing biomarkers for Parkinson's disease and multiple sclerosis. DISCUSSION ADNI has made myriad impacts in its first decade. A competitive renewal of the project in 2015 would see the use of newly developed tau imaging ligands, and the continued development of recruitment strategies and outcome measures for clinical trials.
Collapse
Affiliation(s)
- Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Department of Radiology, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Dallas P Veitch
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Paul S Aisen
- Department of Neurosciences, University of California- San Diego, La Jolla, CA, USA
| | - Laurel A Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, Davis, CA, USA
| | - Nigel J Cairns
- Department of Neurology, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jesse Cedarbaum
- Neurology Early Clinical Development, Biogen Idec, Cambridge, MA, USA
| | - Michael C Donohue
- Division of Biostatistics and Bioinformatics, Department of Family Medicine and Public Health, University of California, San Diego, San Diego, CA, USA
| | - Robert C Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, Davis, CA, USA
| | | | - William Jagust
- Helen Wills Neuroscience Institute and the School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Andrew J Saykin
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leslie Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Marina Del Rey, CA, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute of Neuroimaging and Informatics, Keck School of Medicine of University of Southern California Los Angeles, CA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Alzheimer's Disease Core Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Udall Parkinson's Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
37
|
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Cedarbaum J, Green RC, Harvey D, Jack CR, Jagust W, Luthman J, Morris JC, Petersen RC, Saykin AJ, Shaw L, Shen L, Schwarz A, Toga AW, Trojanowski JQ. 2014 Update of the Alzheimer's Disease Neuroimaging Initiative: A review of papers published since its inception. Alzheimers Dement 2015; 11:e1-120. [PMID: 26073027 PMCID: PMC5469297 DOI: 10.1016/j.jalz.2014.11.001] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/18/2013] [Indexed: 01/18/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) is an ongoing, longitudinal, multicenter study designed to develop clinical, imaging, genetic, and biochemical biomarkers for the early detection and tracking of Alzheimer's disease (AD). The initial study, ADNI-1, enrolled 400 subjects with early mild cognitive impairment (MCI), 200 with early AD, and 200 cognitively normal elderly controls. ADNI-1 was extended by a 2-year Grand Opportunities grant in 2009 and by a competitive renewal, ADNI-2, which enrolled an additional 550 participants and will run until 2015. This article reviews all papers published since the inception of the initiative and summarizes the results to the end of 2013. The major accomplishments of ADNI have been as follows: (1) the development of standardized methods for clinical tests, magnetic resonance imaging (MRI), positron emission tomography (PET), and cerebrospinal fluid (CSF) biomarkers in a multicenter setting; (2) elucidation of the patterns and rates of change of imaging and CSF biomarker measurements in control subjects, MCI patients, and AD patients. CSF biomarkers are largely consistent with disease trajectories predicted by β-amyloid cascade (Hardy, J Alzheimer's Dis 2006;9(Suppl 3):151-3) and tau-mediated neurodegeneration hypotheses for AD, whereas brain atrophy and hypometabolism levels show predicted patterns but exhibit differing rates of change depending on region and disease severity; (3) the assessment of alternative methods of diagnostic categorization. Currently, the best classifiers select and combine optimum features from multiple modalities, including MRI, [(18)F]-fluorodeoxyglucose-PET, amyloid PET, CSF biomarkers, and clinical tests; (4) the development of blood biomarkers for AD as potentially noninvasive and low-cost alternatives to CSF biomarkers for AD diagnosis and the assessment of α-syn as an additional biomarker; (5) the development of methods for the early detection of AD. CSF biomarkers, β-amyloid 42 and tau, as well as amyloid PET may reflect the earliest steps in AD pathology in mildly symptomatic or even nonsymptomatic subjects and are leading candidates for the detection of AD in its preclinical stages; (6) the improvement of clinical trial efficiency through the identification of subjects most likely to undergo imminent future clinical decline and the use of more sensitive outcome measures to reduce sample sizes. Multimodal methods incorporating APOE status and longitudinal MRI proved most highly predictive of future decline. Refinements of clinical tests used as outcome measures such as clinical dementia rating-sum of boxes further reduced sample sizes; (7) the pioneering of genome-wide association studies that leverage quantitative imaging and biomarker phenotypes, including longitudinal data, to confirm recently identified loci, CR1, CLU, and PICALM and to identify novel AD risk loci; (8) worldwide impact through the establishment of ADNI-like programs in Japan, Australia, Argentina, Taiwan, China, Korea, Europe, and Italy; (9) understanding the biology and pathobiology of normal aging, MCI, and AD through integration of ADNI biomarker and clinical data to stimulate research that will resolve controversies about competing hypotheses on the etiopathogenesis of AD, thereby advancing efforts to find disease-modifying drugs for AD; and (10) the establishment of infrastructure to allow sharing of all raw and processed data without embargo to interested scientific investigators throughout the world.
Collapse
Affiliation(s)
- Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Department of Radiology, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA.
| | - Dallas P Veitch
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Paul S Aisen
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Laurel A Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jesse Cedarbaum
- Neurology Early Clinical Development, Biogen Idec, Cambridge, MA, USA
| | - Robert C Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | | | - William Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Johan Luthman
- Neuroscience Clinical Development, Neuroscience & General Medicine Product Creation Unit, Eisai Inc., Philadelphia, PA, USA
| | - John C Morris
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | | | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leslie Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adam Schwarz
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute of Neuroimaging and Informatics, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - John Q Trojanowski
- Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Alzheimer's Disease Core Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Udall Parkinson's Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Lin L, Fu Z, Xu X, Wu S. Mouse brain magnetic resonance microscopy: Applications in Alzheimer disease. Microsc Res Tech 2015; 78:416-24. [PMID: 25810274 DOI: 10.1002/jemt.22489] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/23/2015] [Indexed: 01/26/2023]
Abstract
Over the past two decades, various Alzheimer's disease (AD) trangenetic mice models harboring genes with mutation known to cause familial AD have been created. Today, high-resolution magnetic resonance microscopy (MRM) technology is being widely used in the study of AD mouse models. It has greatly facilitated and advanced our knowledge of AD. In this review, most of the attention is paid to fundamental of MRM, the construction of standard mouse MRM brain template and atlas, the detection of amyloid plaques, following up on brain atrophy and the future applications of MRM in transgenic AD mice. It is believed that future testing of potential drugs in mouse models with MRM will greatly improve the predictability of drug effect in preclinical trials.
Collapse
Affiliation(s)
- Lan Lin
- Biomedical Engineering Department, College of Life Science and Bioengineering, Beijing University of Technology, Beijing, 100124, China
| | | | | | | |
Collapse
|
39
|
Cover KS, van Schijndel RA, Popescu V, van Dijk BW, Redolfi A, Knol DL, Frisoni GB, Barkhof F, Vrenken H. The SIENA/FSL whole brain atrophy algorithm is no more reproducible at 3T than 1.5 T for Alzheimer's disease. Psychiatry Res 2014; 224:14-21. [PMID: 25089020 DOI: 10.1016/j.pscychresns.2014.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 07/03/2014] [Accepted: 07/04/2014] [Indexed: 11/28/2022]
Abstract
The back-to-back (BTB) acquisition of MP-RAGE MRI scans of the Alzheimer׳s Disease Neuroimaging Initiative (ADNI1) provides an excellent data set with which to check the reproducibility of brain atrophy measures. As part of ADNI1, 131 subjects received BTB MP-RAGEs at multiple time points and two field strengths of 3T and 1.5 T. As a result, high quality data from 200 subject-visit-pairs was available to compare the reproducibility of brain atrophies measured with FSL/SIENA over 12 to 18 month intervals at both 3T and 1.5 T. Although several publications have reported on the differing performance of brain atrophy measures at 3T and 1.5 T, no formal comparison of reproducibility has been published to date. Another goal was to check whether tuning SIENA options, including -B, -S, -R and the fractional intensity threshold (f) had a significant impact on the reproducibility. The BTB reproducibility for SIENA was quantified by the 50th percentile of the absolute value of the difference in the percentage brain volume change (PBVC) for the BTB MP-RAGES. At both 3T and 1.5 T the SIENA option combination of "-B f=0.2", which is different from the default values of f=0.5, yielded the best reproducibility as measured by the 50th percentile yielding 0.28 (0.23-0.39)% and 0.26 (0.20-0.32)%. These results demonstrated that in general 3T had no advantage over 1.5 T for the whole brain atrophy measure - at least for SIENA. While 3T MRI is superior to 1.5 T for many types of measurements, and thus worth the additional cost, brain atrophy measurement does not seem to be one of them.
Collapse
Affiliation(s)
- Keith S Cover
- Department of Physics and Medical Technology, VU University medical center, Amsterdam, The Netherlands.
| | | | - Veronica Popescu
- Department of Radiology, VU University medical center, Amsterdam, The Netherlands
| | - Bob W van Dijk
- Department of Physics and Medical Technology, VU University medical center, Amsterdam, The Netherlands
| | - Alberto Redolfi
- Laboratory of Epidemiology & Neuroimaging, IRCCS San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy
| | - Dirk L Knol
- Department of Epidemiology and Biostatistics, VU University medical center, Amsterdam, The Netherlands
| | - Giovanni B Frisoni
- Laboratory of Epidemiology & Neuroimaging, IRCCS San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy
| | - Frederik Barkhof
- Department of Radiology, VU University medical center, Amsterdam, The Netherlands; MS Center Amsterdam and Alzheimer Center, VU University medical center, Amsterdam, The Netherlands
| | - Hugo Vrenken
- Department of Physics and Medical Technology, VU University medical center, Amsterdam, The Netherlands; Department of Radiology, VU University medical center, Amsterdam, The Netherlands; MS Center Amsterdam and Alzheimer Center, VU University medical center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
40
|
Retico A, Bosco P, Cerello P, Fiorina E, Chincarini A, Fantacci ME. Predictive Models Based on Support Vector Machines: Whole-Brain versus Regional Analysis of Structural MRI in the Alzheimer's Disease. J Neuroimaging 2014; 25:552-63. [PMID: 25291354 DOI: 10.1111/jon.12163] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 03/09/2014] [Accepted: 05/25/2014] [Indexed: 01/31/2023] Open
Abstract
Decision-making systems trained on structural magnetic resonance imaging data of subjects affected by the Alzheimer's disease (AD) and healthy controls (CTRL) are becoming widespread prognostic tools for subjects with mild cognitive impairment (MCI). This study compares the performances of three classification methods based on support vector machines (SVMs), using as initial sets of brain voxels (ie, features): (1) the segmented grey matter (GM); (2) regions of interest (ROIs) by voxel-wise t-test filtering; (3) parceled ROIs, according to prior knowledge. The recursive feature elimination (RFE) is applied in all cases to investigate whether feature reduction improves the classification accuracy. We analyzed more than 600 AD Neuroimaging Initiative (ADNI) subjects, training the SVMs on the AD/CTRL dataset, and evaluating them on a trial MCI dataset. The classification performance, evaluated as the area under the receiver operating characteristic curve (AUC), reaches AUC = (88.9 ± .5)% in 20-fold cross-validation on the AD/CTRL dataset, when the GM is classified as a whole. The highest discrimination accuracy between MCI converters and nonconverters is achieved when the SVM-RFE is applied to the whole GM: with AUC reaching (70.7 ± .9)%, it outperforms both ROI-based approaches in predicting the AD conversion.
Collapse
Affiliation(s)
| | - Paolo Bosco
- Dipartimento di Fisica, Università degli Studi di Genova, Genova, Italy.,Istituto Nazionale di Fisica Nucleare, Sezione di Genova, Genova, Italy
| | | | - Elisa Fiorina
- Istituto Nazionale di Fisica Nucleare, Sezione di Torino, Torino, Italy.,Dipartimento di Fisica, Università di Torino, Torino, Italy
| | - Andrea Chincarini
- Istituto Nazionale di Fisica Nucleare, Sezione di Genova, Genova, Italy
| | - Maria Evelina Fantacci
- Istituto Nazionale di Fisica Nucleare, Sezione di Pisa, Pisa, Italy.,Dipartimento di Fisica, Università di Pisa, Pisa, Italy
| |
Collapse
|
41
|
Bertens D, Knol DL, Scheltens P, Visser PJ. Temporal evolution of biomarkers and cognitive markers in the asymptomatic, MCI, and dementia stage of Alzheimer's disease. Alzheimers Dement 2014; 11:511-22. [PMID: 25150730 DOI: 10.1016/j.jalz.2014.05.1754] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 05/10/2014] [Accepted: 05/30/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND We investigated the pattern of disease progression in the asymptomatic, mild cognitive impairment (MCI), and dementia stage of Alzheimer's disease (AD). METHODS We selected 284 subjects with AD pathology, defined as abnormal levels of amyloid beta 1-42 (Aβ1-42) in cerebrospinal fluid (CSF). Disease outcome measures included six biomarkers and five cognitive markers. We compared differences in baseline measures and decline over 4 years between the AD stages and tested whether these changes differed from subjects, without AD pathology (N = 132). RESULTS CSF Aβ1-42 reached the maximum abnormality level in the asymptomatic stage and tau in the MCI stage. The imaging and cognitive markers started to decline in the asymptomatic stage, and decline accelerated with advancing clinical stage. CONCLUSION This study provides further evidence for a temporal evolution of AD biomarkers. Our findings may be helpful to determine stage specific outcome measures for clinical trials.
Collapse
Affiliation(s)
- Daniela Bertens
- Department of Neurology/Alzheimer Centre, VU Medical Centre, Amsterdam, The Netherlands.
| | - Dirk L Knol
- Department of Epidemiology and Biostatistics, VU Medical Centre, Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Neurology/Alzheimer Centre, VU Medical Centre, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Department of Neurology/Alzheimer Centre, VU Medical Centre, Amsterdam, The Netherlands; Department of Psychiatry and Neuropsychology, Maastricht University, School for Mental Health and Neuroscience (MHeNS), Alzheimer Centre Limburg, University Medical Centre, Maastricht, The Netherlands
| | | |
Collapse
|
42
|
Chaalal A, Poirier R, Blum D, Gillet B, Le Blanc P, Basquin M, Buée L, Laroche S, Enderlin V. PTU-induced hypothyroidism in rats leads to several early neuropathological signs of Alzheimer's disease in the hippocampus and spatial memory impairments. Hippocampus 2014; 24:1381-93. [DOI: 10.1002/hipo.22319] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Amina Chaalal
- Centre de Neurosciences Paris-Sud; CNRS; UMR 8195 F-91405 Orsay France
- Université Paris-Sud; UMR 8195 F-91405 Orsay France
| | - Roseline Poirier
- Centre de Neurosciences Paris-Sud; CNRS; UMR 8195 F-91405 Orsay France
- Université Paris-Sud; UMR 8195 F-91405 Orsay France
| | - David Blum
- Université Lille-Nord de France; UDSL; F-59000 Lille France
- Inserm U837, Centre de recherche Jean-Pierre Aubert; IMPRT; F-59000 Lille France
- CHRU-Lille; F-59000 Lille France
| | - Brigitte Gillet
- Université Paris-Sud; UMR 8195 F-91405 Orsay France
- Imagerie par Résonance Magnétique Médicale et MultiModalité; CNRS-UMR8081 F-91405 Orsay France
| | - Pascale Le Blanc
- Centre de Neurosciences Paris-Sud; CNRS; UMR 8195 F-91405 Orsay France
- Université Paris-Sud; UMR 8195 F-91405 Orsay France
| | - Marie Basquin
- Université Lille-Nord de France; UDSL; F-59000 Lille France
- Inserm U837, Centre de recherche Jean-Pierre Aubert; IMPRT; F-59000 Lille France
| | - Luc Buée
- Université Lille-Nord de France; UDSL; F-59000 Lille France
- Inserm U837, Centre de recherche Jean-Pierre Aubert; IMPRT; F-59000 Lille France
- CHRU-Lille; F-59000 Lille France
| | - Serge Laroche
- Centre de Neurosciences Paris-Sud; CNRS; UMR 8195 F-91405 Orsay France
- Université Paris-Sud; UMR 8195 F-91405 Orsay France
| | - Valérie Enderlin
- Centre de Neurosciences Paris-Sud; CNRS; UMR 8195 F-91405 Orsay France
- Université Paris-Sud; UMR 8195 F-91405 Orsay France
| |
Collapse
|
43
|
Liao W, Long X, Jiang C, Diao Y, Liu X, Zheng H, Zhang L. Discerning mild cognitive impairment and Alzheimer Disease from normal aging: morphologic characterization based on univariate and multivariate models. Acad Radiol 2014; 21:597-604. [PMID: 24433704 DOI: 10.1016/j.acra.2013.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 02/02/2023]
Abstract
RATIONALE AND OBJECTIVES Differentiating mild cognitive impairment (MCI) and Alzheimer Disease (AD) from healthy aging remains challenging. This study aimed to explore the cerebral structural alterations of subjects with MCI or AD as compared to healthy elderly based on the individual and collective effects of cerebral morphologic indices using univariate and multivariate analyses. MATERIALS AND METHODS T1-weighted images (T1WIs) were retrieved from Alzheimer Disease Neuroimaging Initiative database for 116 subjects who were categorized into groups of healthy aging, MCI, and AD. Analysis of covariance (ANCOVA) and multivariate analysis of covariance (MANCOVA) were performed to explore the intergroup morphologic alterations indexed by surface area, curvature index, cortical thickness, and subjacent white matter volume with age and sex controlled as covariates, in 34 parcellated gyri regions of interest (ROIs) for both cerebral hemispheres based on the T1WI. Statistical parameters were mapped on the anatomic images to facilitate visual inspection. RESULTS Global rather than region-specific structural alterations were revealed in groups of MCI and AD relative to healthy elderly using MANCOVA. ANCOVA revealed that the cortical thickness decreased more prominently in entorhinal, temporal, and cingulate cortices and was positively correlated with patients' cognitive performance in AD group but not in MCI. The temporal lobe features marked atrophy of white matter during the disease dynamics. Significant intercorrelations were observed among the morphologic indices with univariate analysis for given ROIs. CONCLUSIONS Significant global structural alterations were identified in MCI and AD based on MANCOVA model with improved sensitivity. The intercorrelation among the morphologic indices may dampen the use of individual morphological parameter in featuring cerebral structural alterations. Decrease in cortical thickness is not reflective of the cognitive performance at the early stage of AD.
Collapse
Affiliation(s)
- Weiqi Liao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, Guangdong Province 518055, China
| | - Xiaojing Long
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, Guangdong Province 518055, China
| | - Chunxiang Jiang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, Guangdong Province 518055, China
| | - Yanjun Diao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, Guangdong Province 518055, China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, Guangdong Province 518055, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, Guangdong Province 518055, China
| | - Lijuan Zhang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Ave., Shenzhen, Guangdong Province 518055, China.
| |
Collapse
|
44
|
Braskie MN, Thompson PM. A focus on structural brain imaging in the Alzheimer's disease neuroimaging initiative. Biol Psychiatry 2014; 75:527-33. [PMID: 24367935 PMCID: PMC4019004 DOI: 10.1016/j.biopsych.2013.11.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 11/05/2013] [Accepted: 11/06/2013] [Indexed: 01/18/2023]
Abstract
In recent years, numerous laboratories and consortia have used neuroimaging to evaluate the risk for and progression of Alzheimer's disease (AD). The Alzheimer's Disease Neuroimaging Initiative is a longitudinal, multicenter study that is evaluating a range of biomarkers for use in diagnosis of AD, prediction of patient outcomes, and clinical trials. These biomarkers include brain metrics derived from magnetic resonance imaging (MRI) and positron emission tomography scans as well as metrics derived from blood and cerebrospinal fluid. We focus on Alzheimer's Disease Neuroimaging Initiative studies published between 2011 and March 2013 for which structural MRI was a major outcome measure. Our main goal was to review key articles offering insights into progression of AD and the relationships of structural MRI measures to cognition and to other biomarkers in AD. In Supplement 1, we also discuss genetic and environmental risk factors for AD and exciting new analysis tools for the efficient evaluation of large-scale structural MRI data sets such as the Alzheimer's Disease Neuroimaging Initiative data.
Collapse
Affiliation(s)
- Meredith N Braskie
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, California; Department of Neurology, University of Southern California, Los Angeles, California
| | - Paul M Thompson
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, California; Department of Neurology, University of Southern California, Los Angeles, California; Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, California; Department of Radiology, University of Southern California, Los Angeles, California; Department of Pediatrics, University of Southern California, Los Angeles, California; Department of Ophthalmology, University of Southern California, Los Angeles, California; Keck School of Medicine, and Viterbi School of Engineering, University of Southern California, Los Angeles, California.
| |
Collapse
|
45
|
Seghier ML, Ramsden S, Lim L, Leff AP, Price CJ. Gradual lesion expansion and brain shrinkage years after stroke. Stroke 2014; 45:877-9. [PMID: 24425126 DOI: 10.1161/strokeaha.113.003587] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Lesioned brains of patients with stroke may change through the course of recovery; however, little is known about their evolution in the chronic phase. Here, we aimed to quantify the extent of lesion volume change and brain atrophy in the chronic poststroke brain using magnetic resonance imaging. METHODS Optimized T1-weighted scans were collected more than once (time between visits=2 months to 6 years) in 56 patients (age=36-90 years; time poststroke=3 months to 20 years). Volumetric changes attributable to lesion growth and atrophy were quantified with automated procedures. We looked at how volumetric changes related to time between visits, using nonparametric statistics, after controlling for age, time poststroke, and brain and lesion size at the earlier time. RESULTS Lesions expanded more in patients who had longer time-intervals between their imaging sessions (partial rank correlation ρ=0.56; P<0.001). The median rate of lesion growth was 1.59 cm(3) per year. Across patients, the whole-brain atrophy rate was 0.95% per year, with accelerated atrophy in the ipsilesional hemisphere. CONCLUSIONS We show gradual lesion expansion many years after stroke, beyond that expected by normal aging and after controlling for other variables. Future studies need to understand how structural reorganization enables long-term recovery even when the brain is shrinking.
Collapse
Affiliation(s)
- Mohamed L Seghier
- From the Wellcome Trust Centre for Neuroimaging, Institute of Neurology, UCL, London, UK
| | | | | | | | | |
Collapse
|
46
|
Ventricular enlargement and its clinical correlates in the imaging cohort from the ADCS MCI donepezil/vitamin E study. Alzheimer Dis Assoc Disord 2013; 27:174-81. [PMID: 23694947 DOI: 10.1097/wad.0b013e3182677b3d] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We analyzed the baseline and 3-year T1-weighted magnetic resonance imaging data of 110 amnestic mild cognitive impairment (MCI) participants with minimal hippocampal atrophy at baseline from the Alzheimer's Disease Cooperative Study group MCI Donepezil/Vitamin E trial. Forty-six subjects converted to Alzheimer disease (AD) (MCIc), whereas 64 remained stable (MCInc). We used the radial distance technique to examine the differences in lateral ventricle shape and size between MCIc and MCInc and the associations between ventricular enlargement and cognitive decline. MCIc group had significantly larger frontal and right body/occipital horns relative to MCInc at baseline and significantly larger bilateral frontal, body/occipital, and left temporal horns at follow-up. Global cognitive decline measured with AD Assessment scale cognitive subscale and Mini-Mental State Examination and decline in activities of daily living (ADL) were associated with posterior lateral ventricle enlargement. Decline in AD Assessment scale cognitive subscale and ADL were associated with left temporal and decline in Mini-Mental State Examination with right temporal horn enlargement. After correction for baseline hippocampal volume, decline in ADL showed a significant association with right frontal horn enlargement. Executive decline was associated with right frontal and left temporal horn enlargement.
Collapse
|
47
|
The value of hippocampal and temporal horn volumes and rates of change in predicting future conversion to AD. Alzheimer Dis Assoc Disord 2013; 27:168-73. [PMID: 22760170 PMCID: PMC4154837 DOI: 10.1097/wad.0b013e318260a79a] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hippocampal pathology occurs early in Alzheimer disease (AD), and atrophy, measured by volumes and volume changes, may predict which subjects will develop AD. Measures of the temporal horn (TH), which is situated adjacent to the hippocampus, may also indicate early changes in AD. Previous studies suggest that these metrics can predict conversion from amnestic mild cognitive impairment (MCI) to AD with conversion and volume change measured concurrently. However, the ability of these metrics to predict future conversion has not been investigated. We compared the abilities of hippocampal, TH, and global measures to predict future conversion from MCI to AD. TH, hippocampi, whole brain, and ventricles were measured using baseline and 12-month scans. Boundary shift integral was used to measure the rate of change. We investigated the prediction of conversion between 12 and 24 months in subjects classified as MCI from baseline to 12 months. All measures were predictive of future conversion. Local and global rates of change were similarly predictive of conversion. There was evidence that the TH expansion rate is more predictive than the hippocampal atrophy rate (P=0.023) and that the TH expansion rate is more predictive than the TH volume (P=0.036). Prodromal atrophy rates may be useful predictors of future conversion to sporadic AD from amnestic MCI.
Collapse
|
48
|
Understanding cognitive deficits in Alzheimer's disease based on neuroimaging findings. Trends Cogn Sci 2013; 17:510-6. [PMID: 24029445 DOI: 10.1016/j.tics.2013.08.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 08/07/2013] [Indexed: 01/21/2023]
Abstract
Brain amyloid can be measured using positron emission tomography (PET). There are mixed reports regarding whether amyloid measures are correlated with measures of cognition (in particular memory), depending on the cohorts and cognitive domains assessed. In Alzheimer's disease (AD) patients and those at heightened risk for AD, cognitive performance may be related to the level and extent of classical AD pathology (amyloid plaques and neurofibrillary angles), but it is also influenced by neurodegeneration, neurocognitive reserve, and vascular health. We discuss what recent neuroimaging research has discovered about cognitive deficits in AD and offer suggestions for future research.
Collapse
|
49
|
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Green RC, Harvey D, Jack CR, Jagust W, Liu E, Morris JC, Petersen RC, Saykin AJ, Schmidt ME, Shaw L, Shen L, Siuciak JA, Soares H, Toga AW, Trojanowski JQ. The Alzheimer's Disease Neuroimaging Initiative: a review of papers published since its inception. Alzheimers Dement 2013; 9:e111-94. [PMID: 23932184 DOI: 10.1016/j.jalz.2013.05.1769] [Citation(s) in RCA: 319] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/18/2013] [Indexed: 01/19/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) is an ongoing, longitudinal, multicenter study designed to develop clinical, imaging, genetic, and biochemical biomarkers for the early detection and tracking of Alzheimer's disease (AD). The study aimed to enroll 400 subjects with early mild cognitive impairment (MCI), 200 subjects with early AD, and 200 normal control subjects; $67 million funding was provided by both the public and private sectors, including the National Institute on Aging, 13 pharmaceutical companies, and 2 foundations that provided support through the Foundation for the National Institutes of Health. This article reviews all papers published since the inception of the initiative and summarizes the results as of February 2011. The major accomplishments of ADNI have been as follows: (1) the development of standardized methods for clinical tests, magnetic resonance imaging (MRI), positron emission tomography (PET), and cerebrospinal fluid (CSF) biomarkers in a multicenter setting; (2) elucidation of the patterns and rates of change of imaging and CSF biomarker measurements in control subjects, MCI patients, and AD patients. CSF biomarkers are consistent with disease trajectories predicted by β-amyloid cascade (Hardy, J Alzheimers Dis 2006;9(Suppl 3):151-3) and tau-mediated neurodegeneration hypotheses for AD, whereas brain atrophy and hypometabolism levels show predicted patterns but exhibit differing rates of change depending on region and disease severity; (3) the assessment of alternative methods of diagnostic categorization. Currently, the best classifiers combine optimum features from multiple modalities, including MRI, [(18)F]-fluorodeoxyglucose-PET, CSF biomarkers, and clinical tests; (4) the development of methods for the early detection of AD. CSF biomarkers, β-amyloid 42 and tau, as well as amyloid PET may reflect the earliest steps in AD pathology in mildly symptomatic or even nonsymptomatic subjects, and are leading candidates for the detection of AD in its preclinical stages; (5) the improvement of clinical trial efficiency through the identification of subjects most likely to undergo imminent future clinical decline and the use of more sensitive outcome measures to reduce sample sizes. Baseline cognitive and/or MRI measures generally predicted future decline better than other modalities, whereas MRI measures of change were shown to be the most efficient outcome measures; (6) the confirmation of the AD risk loci CLU, CR1, and PICALM and the identification of novel candidate risk loci; (7) worldwide impact through the establishment of ADNI-like programs in Europe, Asia, and Australia; (8) understanding the biology and pathobiology of normal aging, MCI, and AD through integration of ADNI biomarker data with clinical data from ADNI to stimulate research that will resolve controversies about competing hypotheses on the etiopathogenesis of AD, thereby advancing efforts to find disease-modifying drugs for AD; and (9) the establishment of infrastructure to allow sharing of all raw and processed data without embargo to interested scientific investigators throughout the world. The ADNI study was extended by a 2-year Grand Opportunities grant in 2009 and a renewal of ADNI (ADNI-2) in October 2010 through to 2016, with enrollment of an additional 550 participants.
Collapse
Affiliation(s)
- Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) is a large multi-center study designed to develop optimized methods for acquiring longitudinal neuroimaging, cognitive, and biomarker measures of AD progression in a large cohort of patients with Alzheimer's disease (AD), patients with Mild Cognitive Impairment, and healthy controls. Detailed neuropsychological testing was conducted on all participants. We examined the factor structure of the ADNI Neuropsychological Battery across older adults with differing levels of clinical AD severity based on the Clinical Dementia Rating Scale (CDR). Confirmatory factor analysis (CFA) of 23 variables from 10 neuropsychological tests resulted in five factors (memory, language, visuospatial functioning, attention, and executive function/processing speed) that were invariant across levels of cognitive impairment. Thus, these five factors can be used as indicators of cognitive function in older adults who are participants in ADNI.
Collapse
|