1
|
Rustamzadeh A, Sadigh N, Vahabi Z, Khamseh F, Mohebi N, Ghobadi Z, Moradi F. Effects silymarin and rosuvastatin on amyloid-carriers level in dyslipidemic Alzheimer's patients: A double-blind placebo-controlled randomized clinical trial. IBRO Neurosci Rep 2024; 17:108-121. [PMID: 39139290 PMCID: PMC11321388 DOI: 10.1016/j.ibneur.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
Purpose The production/excretion rate of Amyloid-β (Aβ) is the basis of the plaque burden in alzheimer's disease (AD), which depends on both central and peripheral clearance. In this study, the effect of silymarin and rosuvastatin on serum markers and clinical outcomes in dyslipidemic AD patients was investigated. Methods Participants (n=36) were randomized to silymarin (140 mg), placebo, and rosuvastatin 10 mg orally three times a day for 6 months. Serum collection and clinical outcome tests were performed at baseline and after completion of treatment. Lipid profile markers, oxidative stress markers, Aβ1-42/Aβ1-40 ratio, and Soluble Low-density lipoprotein receptor-Related Protein-1 (sLRP1)/Soluble Receptor for Advanced Glycation End Products (sRAGE) ratio were measured. Results There was a statistically significant increase in Δ-high density lipoprotein (ΔHDL) between silymarin and placebo (P<0.000) and also between rosuvastatin and placebo (p=0.044). The level of Δ-triglycerides (ΔTG) in the silymarin group has a significant decrease compared to both the placebo and the rosuvastatin group (p<0.000 and p=0.036, respectively). The Δ-superoxide dismutase (ΔSOD) level in the silymarin group compared to placebo and rosuvastatin had a significant increase (p<0.000 and p=0.008, respectively). The ΔAβ1-42/Aβ1-40 in the silymarin group compared to both the placebo and rosuvastatin groups had a significant increase (p<0.05). There was an inverse relationship between ΔTG and ΔAβ1-42/Aβ1-40 (p=-0.493 and p=0.004). ΔAβ1-42/Aβ1-40 has a direct statistical relationship with ΔSOD marker (p=0.388 and p=0.031). Also, there was a direct correlation between the level of ΔAβ1-42/Aβ1-40 and ΔsLRP1/sRAGE (p=0.491 and p=0.005). Conclusion Our study showed the relationship between plasma lipids, especially ΔTG and ΔHDL, with ΔAβ1-42/Aβ1-40 in dyslipidemic AD patients, and modulation of these lipid factors can be used to monitor the response to treatments.
Collapse
Affiliation(s)
- Auob Rustamzadeh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nader Sadigh
- Department of Emergency Medicine, School of Medicine, Trauma and Injury Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Vahabi
- Department of Geriatric Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khamseh
- Department of Neurology, Faculty of Medicine, Islamic Azad University, Tehran, Iran
| | - Nafiseh Mohebi
- Department of Neurology, Rasool Akram Hospital, School of Medicine, Iran University of Medial Sciences, Tehran, Iran
| | - Zahra Ghobadi
- Neuroimaging and Clinical Biomarkers Research Group, Pars Darman Medical Imaging Center, Karaj, Iran
| | - Fatemeh Moradi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Ma X, Shyer M, Harris K, Wang D, Hsu YC, Farrell C, Goodwin N, Anjum S, Bukhbinder AS, Dean S, Khan T, Hunter D, Schulz PE, Jiang X, Kim Y. Deep learning to predict rapid progression of Alzheimer's disease from pooled clinical trials: A retrospective study. PLOS DIGITAL HEALTH 2024; 3:e0000479. [PMID: 38598464 PMCID: PMC11006164 DOI: 10.1371/journal.pdig.0000479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024]
Abstract
The rate of progression of Alzheimer's disease (AD) differs dramatically between patients. Identifying the most is critical because when their numbers differ between treated and control groups, it distorts the outcome, making it impossible to tell whether the treatment was beneficial. Much recent effort, then, has gone into identifying RPs. We pooled de-identified placebo-arm data of three randomized controlled trials (RCTs), EXPEDITION, EXPEDITION 2, and EXPEDITION 3, provided by Eli Lilly and Company. After processing, the data included 1603 mild-to-moderate AD patients with 80 weeks of longitudinal observations on neurocognitive health, brain volumes, and amyloid-beta (Aβ) levels. RPs were defined by changes in four neurocognitive/functional health measures. We built deep learning models using recurrent neural networks with attention mechanisms to predict RPs by week 80 based on varying observation periods from baseline (e.g., 12, 28 weeks). Feature importance scores for RP prediction were computed and temporal feature trajectories were compared between RPs and non-RPs. Our evaluation and analysis focused on models trained with 28 weeks of observation. The models achieved robust internal validation area under the receiver operating characteristic (AUROCs) ranging from 0.80 (95% CI 0.79-0.82) to 0.82 (0.81-0.83), and the area under the precision-recall curve (AUPRCs) from 0.34 (0.32-0.36) to 0.46 (0.44-0.49). External validation AUROCs ranged from 0.75 (0.70-0.81) to 0.83 (0.82-0.84) and AUPRCs from 0.27 (0.25-0.29) to 0.45 (0.43-0.48). Aβ plasma levels, regional brain volumetry, and neurocognitive health emerged as important factors for the model prediction. In addition, the trajectories were stratified between predicted RPs and non-RPs based on factors such as ventricular volumes and neurocognitive domains. Our findings will greatly aid clinical trialists in designing tests for new medications, representing a key step toward identifying effective new AD therapies.
Collapse
Affiliation(s)
- Xiaotian Ma
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Madison Shyer
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Kristofer Harris
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dulin Wang
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Yu-Chun Hsu
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Christine Farrell
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Nathan Goodwin
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Sahar Anjum
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Avram S. Bukhbinder
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Division of Pediatric Neurology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Sarah Dean
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Tanveer Khan
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - David Hunter
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Paul E. Schulz
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Xiaoqian Jiang
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Yejin Kim
- Department of Health Data Science and Artificial Intelligence, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
3
|
Tarawneh R, Pankratz VS. The search for clarity regarding "clinically meaningful outcomes" in Alzheimer disease clinical trials: CLARITY-AD and Beyond. Alzheimers Res Ther 2024; 16:37. [PMID: 38365811 PMCID: PMC10870501 DOI: 10.1186/s13195-024-01412-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
CLARITY-AD is an 18-month, double-blinded, placebo-controlled, phase 3 trial which examined the safety and efficacy of the anti-amyloid agent, lecanemab, in mild cognitive impairment and mild dementia due to Alzheimer disease (AD). Lecanemab effectively reduced mean brain amyloid burden and was associated with statistically significant favorable effects, reflected by moderately less decline in the primary and secondary clinical outcomes, at 18 months compared to placebo. However, there is controversy within the AD community regarding the clinical significance of these results and whether they translate into clinically meaningful and tangible benefits on cognition or daily functions.We here review the primary and secondary clinical outcomes of CLARITY-AD and present our interpretation of the potential clinical meaningfulness of the group-level differences in study outcomes in the context of the 18-month study duration. We propose that the validation of stage-appropriate group-level thresholds for clinical meaningfulness of AD trial outcomes in biologically confirmed cohorts will allow objective interpretation of trial results and guide clinical decision-making. Further, in accordance with FDA guidance which emphasizes patient-focused drug development, the contextualization of AD clinical trial outcomes can be facilitated by supplementary individual-level data analyses which measure the risk of disease progression or summarize intraindividual change, using prespecified thresholds of clinically meaningful change, in each of the study groups over the trial period. The concepts of "time-saved" and "time-based" slowing in disease progression can be used to communicate clinical outcomes associated with emerging disease-modifying AD therapies to various stakeholders. We also describe several factors that need to be considered when evaluating outcomes of emerging AD therapies, including disease stage, the neuropathologic complexity of AD, time-based effects of disease-modifying therapies, and the possible influence of individual factors on treatment response and/or risk for adverse events. The consideration of these factors in the design and reporting of future trials of emerging AD therapies will guide clinicians regarding their appropriateness for use in various patient populations.Finally, we emphasize that data from clinical cohorts with longer durations of treatment and follow-up, including extension studies and patient registries, is needed to evaluate the long-term safety and efficacy of lecanemab in early symptomatic AD.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology and Center for Memory and Aging, University of New Mexico, Albuquerque, NM, USA.
| | - Vernon S Pankratz
- Department of Internal Medicine, Division of Epidemiology, Biostatistics, and Preventive Medicine, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
4
|
Kapoor A, Hafeez A, Kushwaha P. Nanocarrier Mediated Intranasal Drug Delivery Systems for the Management of Parkinsonism: A Review. Curr Drug Deliv 2024; 21:709-725. [PMID: 37365787 DOI: 10.2174/1567201820666230523114259] [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: 11/09/2022] [Revised: 02/09/2023] [Accepted: 02/27/2023] [Indexed: 06/28/2023]
Abstract
The transport of drugs to the brain becomes a key concern when treating disorders of the central nervous system. Parkinsonism is one of the major concerns across the world populations, which causes difficulty in coordination and balance. However, the blood-brain barrier is a significant barrier to achieving optimal brain concentration through oral, transdermal, and intravenous routes of administration. The intranasal route with nanocarrier-based formulations has shown potential for managing Parkinsonism disorder (PD). Direct delivery to the brain through the intranasal route is possible via the olfactory and trigeminal pathways using drug-loaded nanotechnology-based drug delivery systems. The critical analysis of reported works demonstrates dose reduction, brain targeting, safety, effectiveness, and stability for drug-loaded nanocarriers. The important aspects of intranasal drug delivery, PD details, and nanocarrier-based intranasal formulations in PD management with a discussion of physicochemical characteristics, cell line studies, and animal studies are the major topics in this review. Patent reports and clinical investigations are summarized in the last sections.
Collapse
Affiliation(s)
- Archita Kapoor
- Faculty of Pharmacy, Integral University, Lucknow- 226026, India
| | - Abdul Hafeez
- Faculty of Pharmacy, Integral University, Lucknow- 226026, India Lucknow India
| | - Poonam Kushwaha
- Faculty of Pharmacy, Integral University, Lucknow- 226026, India
| |
Collapse
|
5
|
Chandler J, Done N, Desai U, Georgieva M, Gomez-Lievano A, Ye W, Zhao A, Eid D, Hilts A, Kirson N, Schilling T. Potential Implications of Slowing Disease Progression in Amyloid-Positive Early Alzheimer's Disease: Estimates from Real-World Data. J Prev Alzheimers Dis 2024; 11:310-319. [PMID: 38374737 DOI: 10.14283/jpad.2024.27] [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] [Indexed: 02/21/2024]
Abstract
BACKGROUND Emerging therapies have shown promising results for slowing the progression of Alzheimer's disease (AD). However, the potential impact of these therapies on real-world outcomes remains to be explored. OBJECTIVE To examine the impact of slowing AD progression on functional abilities and behavioral symptoms. DESIGN Retrospective observational study. SETTING Data from the National Alzheimer's Coordinating Center (NACC) Uniform Data Set (UDS) in the United States (06/2005-11/2021, primary analysis) and the Alzheimer's Disease Neuroimaging Initiative (ADNI) database (09/2005-03/2022, sensitivity analysis) were used. PARTICIPANTS Individuals with mild cognitive impairment (MCI) or mild dementia, Clinical Dementia Rating Scale Sum of Boxes (CDR-SB) score 0.5-9.0 (inclusive; first visit defined as the index date), and confirmed amyloid positivity were identified in NACC. In ADNI, individuals with at least one clinical center visit with a clinical assessment of MCI or mild dementia and confirmed amyloid positivity were identified. MEASUREMENTS Hypothetical effects of slowing disease progression as assessed by CDR-SB on functional and behavioral outcomes including the Functional Activities Questionnaire (FAQ) score, Neuropsychiatric Inventory Questionnaire (NPI-Q) score, and the probability of complete dependence over five years were evaluated using multivariable regression among NACC participants, separately for the subgroups with MCI and mild dementia at baseline, respectively. For the ADNI sensitivity analysis, the hypothetical effects of slowing disease progression were evaluated for FAQ score using multivariable regression among the MCI participants only. RESULTS Compared with natural disease progression, slowing progression by 20% over five years for NACC participants with MCI and mild dementia, respectively, would result in 1.7-point (10.8%) and 1.6-point (12.9%) less deterioration based on FAQ; 0.5-point (20.3%) and 0.5-point (19.3%) less deterioration based on NPI-Q; 4.7 percentage-point (22.2%) and 10.1 percentage-point (21.6%) lower probability of complete dependence. Among ADNI participants, delaying disease progression by 20% or 30% over 4 years would avert deterioration based on FAQ of 1.1 points (20.4%) and 1.6 points (29.6%), respectively, compared to natural disease progression. CONCLUSIONS Slowing early AD progression could result in preservation of functional and behavioral attributes and functional autonomy for longer.
Collapse
Affiliation(s)
- J Chandler
- Urvi Desai, PhD, Analysis Group, Inc., 111 Huntington Avenue, 14th Floor, Boston, MA 02199, USA, Phone: +1-617-425-8315,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Holdridge KC, Yaari R, Hoban DB, Andersen S, Sims JR. Targeting amyloid β in Alzheimer's disease: Meta-analysis of low-dose solanezumab in Alzheimer's disease with mild dementia studies. Alzheimers Dement 2023; 19:4619-4628. [PMID: 36946603 DOI: 10.1002/alz.13031] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Solanezumab is a monoclonal antibody that binds to the mid-domain of soluble amyloid β peptide. This meta-analysis evaluated the effect of low-dose solanezumab on clinical progression in three phase 3 studies. METHODS The population comprised patients aged ≥55 years with Alzheimer's disease (AD) with mild dementia, randomized to 400 mg solanezumab or placebo every 4 weeks for 80 weeks. Frequentist mixed-model repeated-measures (MMRM) and Bayesian disease progression model (DPM) longitudinal analyses were conducted. RESULTS Pooled MMRM analyses showed a statistically significant effect of solanezumab across cognitive and functional outcome measures. DPM results were generally consistent with MMRM results, ranging from 15% to 30% slowing of clinical progression. DISCUSSION These analyses suggest low-dose solanezumab slows clinical progression of AD with mild dementia. The ongoing A4 solanezumab study in participants with preclinical AD will ascertain the effect of a higher dose of solanezumab in an earlier disease stage. HIGHLIGHTS Individual EXPEDITION studies were negative but suggest low-dose solanezumab had an effect in slowing the clinical progression of Alzheimer's disease (AD) with mild dementia. At 80 weeks, mixed-model repeated-measures analyses showed numeric reductions in measures of clinical decline in solanezumab-treated arms compared with placebo across almost every outcome measure, and statistical significance in multiple outcome measures in each study. Pooled analyses suggest a high probability that low-dose solanezumab has at least some effect on slowing the clinical progression of AD with mild dementia. Across cognitive and functional outcome measures, estimates from disease progression model analyses range from 15% to 30% slowing of decline with low-dose solanezumab in AD with mild dementia.
Collapse
Affiliation(s)
| | - Roy Yaari
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | | - John R Sims
- Eli Lilly and Company, Indianapolis, Indiana, USA
| |
Collapse
|
7
|
Saito S, Yamashiro T, Yamauchi M, Yamamoto Y, Noguchi M, Tomita T, Kawakami D, Shikata M, Tanaka T, Ihara M. Complement 3 Is a Potential Biomarker for Cerebral Amyloid Angiopathy. J Alzheimers Dis 2022; 89:381-387. [DOI: 10.3233/jad-220494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Cerebral amyloid angiopathy is a cerebrovascular disease directly implicated in Alzheimer’s disease pathogenesis through amyloid-β deposition. Growing evidence has shown a pivotal role of chronic neuroinflammation both in cerebral amyloid angiopathy and Alzheimer’s disease. Objective: The aim of this study was to investigate whether circulating levels of the complement 3, a crucial component of the innate immune system, are increased in patients with cerebral amyloid angiopathy. Methods: Serum complement 3 levels were retrospectively measured by a sandwich enzyme-linked immunosorbent assay in a single-center cohort of patients with mild cognitive impairment. The diagnosis of cerebral amyloid angiopathy was based on the modified Boston criteria. Logistic regression analysis was performed to identify the predictive factors for cerebral amyloid angiopathy. Results: We analyzed 55 mild cognitive impairment patients (mean age [standard deviation]: 76.3 [6.8] years; 33 [60% ] men). Complement 3 levels were significantly increased in cerebral amyloid angiopathy patients (n = 16) compared with those without cerebral amyloid angiopathy (n = 39) (median [interquartile range]: 0.43 [0.34–0.65] versus 0.35 [0.25–0.45], respectively; p = 0.040). Univariate and multivariate logistic regression analysis revealed that increased complement 3 levels were significantly associated with cerebral amyloid angiopathy. After selection of the best predictive model using stepwise selection, complement 3 was preserved as a significant independent predictive factor for cerebral amyloid angiopathy (odds ratio per 0.1 unit/mL increase [95% confidence interval]: 1.407 [1.042–1.899]; p = 0.026). Conclusion: Complement activation may play a pivotal role in cerebral amyloid angiopathy. Complement 3 may be a novel diagnostic biomarker for cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Takayuki Yamashiro
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Miho Yamauchi
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yumi Yamamoto
- Department of Molecular Innovation in Epidemiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Michio Noguchi
- National Cerebral and Cardiovascular Center Biobank, Suita, Japan
| | - Tsutomu Tomita
- National Cerebral and Cardiovascular Center Biobank, Suita, Japan
| | - Daisuke Kawakami
- Analytical & Measuring Instruments Division, Shimadzu Corporation, Nakagyo-ku, Kyoto, Japan
| | - Masamitsu Shikata
- Analytical & Measuring Instruments Division, Shimadzu Corporation, Nakagyo-ku, Kyoto, Japan
| | - Tomotaka Tanaka
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
8
|
Epelbaum S, Burgos N, Canney M, Matthews D, Houot M, Santin MD, Desseaux C, Bouchoux G, Stroer S, Martin C, Habert MO, Levy M, Bah A, Martin K, Delatour B, Riche M, Dubois B, Belin L, Carpentier A. Pilot study of repeated blood-brain barrier disruption in patients with mild Alzheimer's disease with an implantable ultrasound device. Alzheimers Res Ther 2022; 14:40. [PMID: 35260178 PMCID: PMC8905724 DOI: 10.1186/s13195-022-00981-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/22/2022] [Indexed: 12/11/2022]
Abstract
Background Temporary disruption of the blood-brain barrier (BBB) using pulsed ultrasound leads to the clearance of both amyloid and tau from the brain, increased neurogenesis, and mitigation of cognitive decline in pre-clinical models of Alzheimer’s disease (AD) while also increasing BBB penetration of therapeutic antibodies. The goal of this pilot clinical trial was to investigate the safety and efficacy of this approach in patients with mild AD using an implantable ultrasound device. Methods An implantable, 1-MHz ultrasound device (SonoCloud-1) was implanted under local anesthesia in the skull (extradural) of 10 mild AD patients to target the left supra-marginal gyrus. Over 3.5 months, seven ultrasound sessions in combination with intravenous infusion of microbubbles were performed twice per month to temporarily disrupt the BBB. 18F-florbetapir and 18F-fluorodeoxyglucose positron emission tomography (PET) imaging were performed on a combined PET/MRI scanner at inclusion and at 4 and 8 months after the initiation of sonications to monitor the brain metabolism and amyloid levels along with cognitive evaluations. The evolution of cognitive and neuroimaging features was compared to that of a matched sample of control participants taken from the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Results A total of 63 BBB opening procedures were performed in nine subjects. The procedure was well-tolerated. A non-significant decrease in amyloid accumulation at 4 months of − 6.6% (SD = 7.2%) on 18F-florbetapir PET imaging in the sonicated gray matter targeted by the ultrasound transducer was observed compared to baseline in six subjects that completed treatments and who had evaluable imaging scans. No differences in the longitudinal change in the glucose metabolism were observed compared to the neighboring or contralateral regions or to the change observed in the same region in ADNI participants. No significant effect on cognition evolution was observed in comparison with the ADNI participants as expected due to the small sample size and duration of the trial. Conclusions These results demonstrate the safety of ultrasound-based BBB disruption and the potential of this technology to be used as a therapy for AD patients. Research of this technique in a larger clinical trial with a device designed to sonicate larger volumes of tissue and in combination with disease-modifying drugs may further enhance the effects observed. Trial registration ClinicalTrials.gov, NCT03119961 Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00981-1.
Collapse
Affiliation(s)
- Stéphane Epelbaum
- Sorbonne Université, Paris, France.,Institut du Cerveau - Paris Brain Institute - ICM, Paris, France.,Inserm, Paris, France.,CNRS, Paris, France.,Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Ninon Burgos
- Sorbonne Université, Paris, France.,Institut du Cerveau - Paris Brain Institute - ICM, Paris, France.,Inserm, Paris, France.,CNRS, Paris, France.,Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Aramis Project-Team, Inria-APHP Collaboration, Inria, Paris, France
| | | | | | - Marion Houot
- Sorbonne Université, Paris, France.,Institut du Cerveau - Paris Brain Institute - ICM, Paris, France.,Inserm, Paris, France.,CNRS, Paris, France.,Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Centre of Excellence of Neurodegenerative Disease (CoEN), Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Clinical Investigation Centre, Institut du Cerveau - Paris Brain Institute - ICM, Paris, France
| | - Mathieu D Santin
- Sorbonne Université, Paris, France.,Institut du Cerveau - Paris Brain Institute - ICM, Paris, France.,Inserm, Paris, France.,CNRS, Paris, France.,Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Center for NeuroImaging Research (CENIR), Institut du Cerveau - Paris Brain Institute - ICM, Paris, France
| | | | | | - Sebastian Stroer
- Department of Neuroradiology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | | | - Marie-Odile Habert
- Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Laboratoire d'Imagerie Biomédicale, Sorbonne Université, Inserm U 1146, CNRS UMR 7371, Pitié-Salpêtrière Hospital, Paris, France.,Centre Acquisition et Traitement des Images, Pitié-Salpêtrière Hospital, Paris, France
| | - Marcel Levy
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Aicha Bah
- Clinical Research Unit, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Karine Martin
- Clinical Research Unit, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Benoît Delatour
- Sorbonne Université, Paris, France.,Institut du Cerveau - Paris Brain Institute - ICM, Paris, France.,Inserm, Paris, France.,CNRS, Paris, France.,Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Maximilien Riche
- Sorbonne Université, Paris, France.,Institut du Cerveau - Paris Brain Institute - ICM, Paris, France.,Inserm, Paris, France.,CNRS, Paris, France.,Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Bruno Dubois
- Sorbonne Université, Paris, France.,Institut du Cerveau - Paris Brain Institute - ICM, Paris, France.,Inserm, Paris, France.,CNRS, Paris, France.,Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Lisa Belin
- Sorbonne Université, Paris, France.,Department of Biostatistics, Public Health and Medical Informatics, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Alexandre Carpentier
- Sorbonne Université, Paris, France. .,Department of Neurosurgery, Pitié-Salpêtrière Hospital, AP-HP, Paris, France.
| |
Collapse
|
9
|
Wessels AM, Rentz DM, Case M, Lauzon S, Sims JR. Integrated Alzheimer's Disease Rating Scale: Clinically meaningful change estimates. ALZHEIMER'S & DEMENTIA: TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2022; 8:e12312. [PMID: 35676941 PMCID: PMC9169866 DOI: 10.1002/trc2.12312] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/22/2022] [Accepted: 05/04/2022] [Indexed: 11/17/2022]
Abstract
Introduction The Integrated Alzheimer's Disease Rating Scale (iADRS) has been used to detect differences in disease progression in early Alzheimer's disease (AD). The objectives of this study were to enhance understanding of iADRS point changes within the context of clinical trials, and to establish a minimal clinically important difference (MCID) on the iADRS. Methods Data from AMARANTH and EXPEDITION3 were analyzed using various approaches, including anchor‐based, distribution‐based, regression analyses, and cumulative distribution function (CDF) plots. Three potential anchors were examined, including the Clinical Dementia Rating—Sum of Boxes, Mini‐Mental State Examination, and Functional Activities Questionnaire. Triangulation of all results was used to determine the MCID for participants with mild cognitive impairment (MCI) due to AD and AD with mild dementia. Results All three anchors met criteria for “sufficiently associated” (|r| = 0.4–0.7). Cumulatively, results from anchor‐based and distribution‐based results converged to suggest an iADRS MCID of 5 points for MCI due to AD and 9 points for AD with mild dementia. Regression analyses and CDF plots supported these values. Discussion These findings suggest the iADRS can be used in clinical trials to detect a clinically meaningful outcome of AD progression.
Collapse
Affiliation(s)
| | - Dorene M. Rentz
- Department of Neurology Massachusetts General Hospital, Harvard Medical School Boston Massachusetts USA
- Center for Alzheimer Research and Treatment Department of Neurology Brigham and Women's Hospital, Harvard Medical School Boston Massachusetts USA
| | | | | | | |
Collapse
|
10
|
Saito S, Shinmyozu K, Kawakami D, Yamauchi M, Ikeda S, Hattori Y, Yamamoto R, Hayakawa N, Ihara M. Conversion from cilostazol to OPC-13015 linked to mitigation of cognitive impairment. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12182. [PMID: 34095441 PMCID: PMC8158162 DOI: 10.1002/trc2.12182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/03/2021] [Accepted: 04/20/2021] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Cilostazol may be a novel therapeutic agent for Alzheimer's disease. Its metabolite, OPC-13015, has a stronger inhibitory effect on type 3 phosphodiesterase than cilostazol. METHODS We prospectively enrolled patients with mild cognitive impairment to whom cilostazol was newly prescribed. Patients underwent the Montreal Cognitive Assessment (MoCA) twice, at a 6-month interval. Plasma cilostazol, OPC-13015, OPC-13213, and OPC-13217 concentrations were determined using liquid chromatography-tandem mass spectrometry. RESULTS MoCA score changes from baseline to the 6-month visit were positively correlated with ratios of OPC-13015 to cilostazol and total metabolites (n = 19, P = .005). Patients with higher ratios of OPC-13015 (≥0.18, median value; n = 10) had significantly higher MoCA scores (P = .036) than patients with lower ratios (the ratio <0.18, n = 9). The absolute value of OPC-13015 concentration in blood was also higher in patients with preserved cognitive function (P = .033). DISCUSSION Blood OPC-13015 levels may be a predictive biomarker of cilostazol treatment for Alzheimer's disease.
Collapse
Affiliation(s)
- Satoshi Saito
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
- Department of Pediatric DentistryOsaka University Graduate School of DentistrySuitaOsakaJapan
| | - Kaori Shinmyozu
- Department of PharmacyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Daisuke Kawakami
- Division of Analytical & Measuring InstrumentsShimadzu CorporationNakagyo‐kuKyotoJapan
| | - Miho Yamauchi
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Shuhei Ikeda
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Yorito Hattori
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Rintaro Yamamoto
- Division of Analytical & Measuring InstrumentsShimadzu CorporationNakagyo‐kuKyotoJapan
| | - Naoki Hayakawa
- Department of PharmacyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| | - Masafumi Ihara
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaOsakaJapan
| |
Collapse
|
11
|
Arya M, Manoj Kumar MK, Sabitha M, Menon KN, Nair SC. Nanotechnology approaches for enhanced CNS delivery in treating Alzheimer's disease. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.03.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
12
|
Salloway S, Honigberg LA, Cho W, Ward M, Friesenhahn M, Brunstein F, Quartino A, Clayton D, Mortensen D, Bittner T, Ho C, Rabe C, Schauer SP, Wildsmith KR, Fuji RN, Suliman S, Reiman EM, Chen K, Paul R. Amyloid positron emission tomography and cerebrospinal fluid results from a crenezumab anti-amyloid-beta antibody double-blind, placebo-controlled, randomized phase II study in mild-to-moderate Alzheimer's disease (BLAZE). ALZHEIMERS RESEARCH & THERAPY 2018; 10:96. [PMID: 30231896 PMCID: PMC6146627 DOI: 10.1186/s13195-018-0424-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/29/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND We investigated the effect of crenezumab, a humanized anti-amyloid-beta (Aβ) immunoglobulin (Ig)G4 monoclonal antibody, on biomarkers of amyloid pathology, neurodegeneration, and disease progression in patients with mild-to-moderate Alzheimer's disease (AD). METHODS This double-blind, placebo-controlled, randomized phase II study enrolled patients with mild-to-moderate AD and a Mini-Mental State Examination (MMSE) score of 18-26. In part 1 of the study, patients were 2:1 randomized to receive low-dose subcutaneous (SC) 300 mg crenezumab every 2 weeks (q2w) or placebo for 68 weeks; in part 2, patients were 2:1 randomized to receive high-dose intravenous (IV) 15 mg/kg crenezumab every 4 weeks (q4w) or placebo for 68 weeks. The primary endpoint was change in amyloid burden from baseline to week 69 assessed by florbetapir positron emission tomography (PET) in the modified intent-to-treat population. Secondary endpoints were change from baseline to week 69 in cerebrospinal fluid (CSF) biomarkers and fluorodeoxyglucose PET, and change from baseline to week 73 in 12-point Alzheimer's Disease Assessment Scale cognitive subscale (ADAS-Cog12) and Clinical Dementia Rating Sum of Boxes (CDR-SB). Safety was assessed in patients who received at least one dose of study treatment. RESULTS From August 2011 to September 2012, 91 patients were enrolled and randomized (low-dose SC cohort: crenezumab (n = 26) or placebo (n = 13); high-dose IV cohort: crenezumab (n = 36) or placebo (n = 16)). The primary endpoint was not met using a prespecified cerebellar reference region to calculate standard uptake value ratios (SUVRs) from florbetapir PET. Exploratory analyses using subcortical white matter reference regions showed nonsignificant trends toward slower accumulation of plaque amyloid in the high-dose IV cohort. In both cohorts, a significant mean increase from baseline in CSF Aβ(1-42) levels versus placebo was observed. Nonsignificant trends toward ADAS-Cog12 and CDR-SB benefits were identified in a mild (MMSE 20-26) subset of the high-dose IV cohort. No amyloid-related imaging abnormalities due to edema/effusion were observed. CONCLUSION The primary endpoint was not met. Exploratory findings suggest potential Aβ target engagement with crenezumab and possible slower accumulation of plaque amyloid. Studies investigating the effects of higher doses of crenezumab on amyloid load and disease progression are ongoing. TRIAL REGISTRATION ClinicalTrials.gov, NCT01397578 . Registered on 18 July 2011.
Collapse
Affiliation(s)
- Stephen Salloway
- Department of Neurology and Psychiatry, The Warren Alpert Medical School of Brown University, 345 Blackstone Boulevard, Providence, RI, 2906, USA.
| | | | - William Cho
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | | | - Carole Ho
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Robert Paul
- Genentech Inc., South San Francisco, CA, USA
| |
Collapse
|
13
|
Shah C, Liu J, Lv P, Sun H, Xiao Y, Liu J, Zhao Y, Zhang W, Yao L, Gong Q, Lui S. Age Related Changes in Topological Properties of Brain Functional Network and Structural Connectivity. Front Neurosci 2018; 12:318. [PMID: 29867329 PMCID: PMC5962656 DOI: 10.3389/fnins.2018.00318] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/24/2018] [Indexed: 02/05/2023] Open
Abstract
Introduction: There are still uncertainties about the true nature of age related changes in topological properties of the brain functional network and its structural connectivity during various developmental stages. In this cross- sectional study, we investigated the effects of age and its relationship with regional nodal properties of the functional brain network and white matter integrity. Method: DTI and fMRI data were acquired from 458 healthy Chinese participants ranging from age 8 to 81 years. Tractography was conducted on the DTI data using FSL. Graph Theory analyses were conducted on the functional data yielding topological properties of the functional network using SPM and GRETNA toolbox. Two multiple regressions were performed to investigate the effects of age on nodal topological properties of the functional brain network and white matter integrity. Result: For the functional studies, we observed that regional nodal characteristics such as node betweenness were decreased while node degree and node efficiency was increased in relation to increasing age. Perversely, we observed that the relationship between nodal topological properties and fasciculus structures were primarily positive for nodal betweenness but negative for nodal degree and nodal efficiency. Decrease in functional nodal betweenness was primarily located in superior frontal lobe, right occipital lobe and the global hubs. These brain regions also had both direct and indirect anatomical relationships with the 14 fiber bundles. A linear age related decreases in the Fractional anisotropy (FA) value was found in the callosum forceps minor. Conclusion: These results suggests that age related differences were more pronounced in the functional than in structural measure indicating these measures do not have direct one-to-one mapping. Our study also indicates that the fiber bundles with longer fibers exhibited a more pronounced effect on the properties of functional network.
Collapse
Affiliation(s)
- Chandan Shah
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jia Liu
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Peilin Lv
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Huaiqiang Sun
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yuan Xiao
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jieke Liu
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Youjin Zhao
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Wenjing Zhang
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Li Yao
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Qiyong Gong
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Su Lui
- Huaxi MR Research Center, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Cummings JL, Cohen S, van Dyck CH, Brody M, Curtis C, Cho W, Ward M, Friesenhahn M, Rabe C, Brunstein F, Quartino A, Honigberg LA, Fuji RN, Clayton D, Mortensen D, Ho C, Paul R. ABBY: A phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease. Neurology 2018; 90:e1889-e1897. [PMID: 29695589 PMCID: PMC5962917 DOI: 10.1212/wnl.0000000000005550] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 03/05/2018] [Indexed: 12/31/2022] Open
Abstract
Objective To evaluate the safety and efficacy of crenezumab in patients with mild to moderate Alzheimer disease (AD). Methods In this phase 2 trial, 431 patients with mild to moderate AD 50 to 80 years of age were randomized 2:1 (crenezumab:placebo). Patients received low-dose subcutaneous crenezumab 300 mg or placebo every 2 weeks (n = 184) or high-dose intravenous crenezumab 15 mg/kg or placebo every 4 weeks (n = 247) for 68 weeks. Primary outcome measures were change in Alzheimer's Disease Assessment Scale–Cognitive Subscale (ADAS-Cog12) and Clinical Dementia Rating–Sum of Boxes scores from baseline to week 73. Results The primary and secondary endpoints were not met. In an exploratory post hoc analysis, a reduction in decline on the ADAS-Cog12 was observed in the high-dose group. Separation from the placebo group on the ADAS-Cog12 was greatest in the milder subsets of AD patients and reached statistical significance in the group with Mini-Mental State Examination scores of 22 to 26. In both groups, there was a significant increase in CSF β-amyloid1-42 levels that correlated with crenezumab CSF levels. The overall rate of adverse events was balanced between groups. One case of amyloid-related imaging abnormalities indicative of vasogenic edema or effusions was reported. Conclusions Although prespecified criteria for testing treatment effects were not met, these data suggest a potential treatment effect in patients with mild AD treated with high-dose crenezumab. Together with the safety profile for crenezumab, these data support the exploration of crenezumab treatment at even higher doses in patients with early AD. Clinicaltrials.gov identifier NCT 01343966. Classification of evidence This study provides Class II evidence that, for people with AD, crenezumab does not significantly improve cognition or function at 18 months. The study is rated Class II because <80% of enrolled patients completed the study.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany.
| | - Sharon Cohen
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Christopher H van Dyck
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Mark Brody
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Craig Curtis
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - William Cho
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Michael Ward
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Michel Friesenhahn
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Christina Rabe
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Flavia Brunstein
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Angelica Quartino
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Lee A Honigberg
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Reina N Fuji
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - David Clayton
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Deborah Mortensen
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Carole Ho
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| | - Robert Paul
- From the Cleveland Clinic Lou Ruvo Center for Brain Health (J.L.C.), Las Vegas, NV; Toronto Memory Program (S.C.), ON, Canada; Alzheimer's Disease Research Unit (C.H.v.D.), Yale University School of Medicine, New Haven, CT; Brain Matters Research Inc (M.B.), Delray Beach, FL; Compass Research, LLC (C.C.), Orlando, FL; Genentech (W.C., M.W., M.F., F.B., A.Q., L.A.H., R.N.F., D.C., D.M., C.H., R.P.), South San Francisco, CA; and Roche Diagnostics (C.R.), Penzberg, Germany
| |
Collapse
|
15
|
Honig LS, Vellas B, Woodward M, Boada M, Bullock R, Borrie M, Hager K, Andreasen N, Scarpini E, Liu-Seifert H, Case M, Dean RA, Hake A, Sundell K, Poole Hoffmann V, Carlson C, Khanna R, Mintun M, DeMattos R, Selzler KJ, Siemers E. Trial of Solanezumab for Mild Dementia Due to Alzheimer's Disease. N Engl J Med 2018; 378:321-330. [PMID: 29365294 DOI: 10.1056/nejmoa1705971] [Citation(s) in RCA: 717] [Impact Index Per Article: 102.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Alzheimer's disease is characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. The humanized monoclonal antibody solanezumab was designed to increase the clearance from the brain of soluble Aβ, peptides that may lead to toxic effects in the synapses and precede the deposition of fibrillary amyloid. METHODS We conducted a double-blind, placebo-controlled, phase 3 trial involving patients with mild dementia due to Alzheimer's disease, defined as a Mini-Mental State Examination (MMSE) score of 20 to 26 (on a scale from 0 to 30, with higher scores indicating better cognition) and with amyloid deposition shown by means of florbetapir positron-emission tomography or Aβ1-42 measurements in cerebrospinal fluid. Patients were randomly assigned to receive solanezumab at a dose of 400 mg or placebo intravenously every 4 weeks for 76 weeks. The primary outcome was the change from baseline to week 80 in the score on the 14-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog14; scores range from 0 to 90, with higher scores indicating greater cognitive impairment). RESULTS A total of 2129 patients were enrolled, of whom 1057 were assigned to receive solanezumab and 1072 to receive placebo. The mean change from baseline in the ADAS-cog14 score was 6.65 in the solanezumab group and 7.44 in the placebo group, with no significant between-group difference at week 80 (difference, -0.80; 95% confidence interval, -1.73 to 0.14; P=0.10). As a result of the failure to reach significance with regard to the primary outcome in the prespecified hierarchical analysis, the secondary outcomes were considered to be descriptive and are reported without significance testing. The change from baseline in the MMSE score was -3.17 in the solanezumab group and -3.66 in the placebo group. Adverse cerebral edema or effusion lesions that were observed on magnetic resonance imaging after randomization occurred in 1 patient in the solanezumab group and in 2 in the placebo group. CONCLUSIONS Solanezumab at a dose of 400 mg administered every 4 weeks in patients with mild Alzheimer's disease did not significantly affect cognitive decline. (Funded by Eli Lilly; EXPEDITION3 ClinicalTrials.gov number, NCT01900665 .).
Collapse
Affiliation(s)
- Lawrence S Honig
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Bruno Vellas
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Michael Woodward
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Mercè Boada
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Roger Bullock
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Michael Borrie
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Klaus Hager
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Niels Andreasen
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Elio Scarpini
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Hong Liu-Seifert
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Michael Case
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Robert A Dean
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Ann Hake
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Karen Sundell
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Vicki Poole Hoffmann
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Christopher Carlson
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Rashna Khanna
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Mark Mintun
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Ronald DeMattos
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Katherine J Selzler
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| | - Eric Siemers
- From the Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York (L.S.H.); Gérontopôle, Centre Hospitalier Universitaire Toulouse, Unité Mixte de Recherche INSERM Unité 1027 Université Toulouse III-Paul Sabatier, Toulouse, France (B.V.); Austin Health Continuing Care Clinical Service Unit, Heidelberg, and the University of Melbourne, Melbourne, VIC - both in Australia (M.W.); Fundació ACE, Alzheimer Research Center and Memory Clinic, Institut Català de Neurociències Aplicades, Barcelona (M. Boada); Kingshill Research Centre, Victoria Hospital, Swindon, United Kingdom (R.B.); the Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada (M. Borrie); Clinic for Medicine of the Elderly, Diakovere Henriettenstift, Hannover, Germany (K.H.); Karolinska Institutet Alzheimer's Disease Research Center and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden (N.A.); the Department of Pathophysiology and Transplantation, Neurology Unit, Dino Ferrari Center, University of Milan, Fondazione Ca' Granda, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico, Milan (E. Scarpini); Eli Lilly (H.L.-S., M.C., A.H., K.S., V.P.H., C.C., R.K., M.M., R.D., K.J.S., E. Siemers) and the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine (R.A.D.) - both in Indianapolis; and Avid Radiopharmaceuticals, Philadelphia (M.M.)
| |
Collapse
|
16
|
Tarawneh R, D'Angelo G, Crimmins D, Herries E, Griest T, Fagan AM, Zipfel GJ, Ladenson JH, Morris JC, Holtzman DM. Diagnostic and Prognostic Utility of the Synaptic Marker Neurogranin in Alzheimer Disease. JAMA Neurol 2017; 73:561-71. [PMID: 27018940 DOI: 10.1001/jamaneurol.2016.0086] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
IMPORTANCE Synaptic loss is an early pathologic substrate of Alzheimer disease (AD). Neurogranin is a postsynaptic neuronal protein that has demonstrated utility as a cerebrospinal fluid (CSF) marker of synaptic loss in AD. OBJECTIVE To investigate the diagnostic and prognostic utility of CSF neurogranin levels in a large, well-characterized cohort of individuals with symptomatic AD and cognitively normal controls. DESIGN, SETTING, AND PARTICIPANTS A cross-sectional and longitudinal observational study of cognitive decline in patients with symptomatic AD and cognitively normal controls was performed. Participants were individuals with a clinical diagnosis of early symptomatic AD and cognitively normal controls who were enrolled in longitudinal studies of aging and dementia at the Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, from January 21, 2000, through March 21, 2011. Data analysis was performed from November 1, 2013, to March 31, 2015. MAIN OUTCOMES AND MEASURES Correlations between baseline CSF biomarker levels and future cognitive decline in patients with symptomatic AD and cognitively normal controls over time. RESULTS A total of 302 individuals (mean [SE] age, 73.1 [0.4] years) were included in this study (95 patients [52 women and 43 men] with AD and 207 controls [125 women and 82 men]). The CSF neurogranin levels differentiated patients with early symptomatic AD from controls with comparable diagnostic utility (mean [SE] area under the receiver operating characteristic curve, 0.71 [0.03]; 95% CI, 0.64-0.77) to the other CSF biomarkers. The CSF neurogranin levels correlated with brain atrophy (normalized whole-brain volumes: adjusted r = -0.38, P = .02; hippocampal volumes: adjusted r = -0.36, P = .03; entorhinal volumes: adjusted r = -0.46, P = .006; and parahippocampal volumes: adjusted r = -0.47, P = .005, n = 38) in AD and with amyloid load (r = 0.39, P = .02, n = 36) in preclinical AD. The CSF neurogranin levels predicted future cognitive impairment (adjusted hazard ratio, 1.89; 95% CI, 1.29-2.78; P = .001 as a continuous measure, and adjusted hazard ratio, 2.78; 95% CI, 1.13-5.99; P = .02 as a categorical measure using the 85th percentile cutoff value) in controls and rates of cognitive decline (Clinical Dementia Rating sum of boxes score: β estimate, 0.29; P = .001; global composite scores: β estimate, -0.11; P = .001; episodic memory scores: β estimate, -0.18; P < .001; and semantic memory scores: β estimate, -0.06; P = .04, n = 57) in patients with symptomatic AD over time, similarly to the CSF proteins VILIP-1, tau, and p-tau181. CONCLUSIONS AND RELEVANCE The CSF levels of the synaptic marker neurogranin offer diagnostic and prognostic utility for early symptomatic AD that is comparable to other CSF markers of AD. Importantly, CSF neurogranin complements the collective ability of these markers to predict future cognitive decline in cognitively normal individuals and, therefore, will be a useful addition to the current panel of AD biomarkers.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri2Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri3Charles F. and Joanne Knight Alzheimer Disease Research Center, Wash
| | - Gina D'Angelo
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, Missouri5Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri
| | - Dan Crimmins
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Elizabeth Herries
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Terry Griest
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri2Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri3Charles F. and Joanne Knight Alzheimer Disease Research Center, Wash
| | - Gregory J Zipfel
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri7Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri
| | - Jack H Ladenson
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri2Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri3Charles F. and Joanne Knight Alzheimer Disease Research Center, Wash
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri2Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri3Charles F. and Joanne Knight Alzheimer Disease Research Center, Wash
| |
Collapse
|
17
|
Webster L, Groskreutz D, Grinbergs-Saull A, Howard R, O'Brien JT, Mountain G, Banerjee S, Woods B, Perneczky R, Lafortune L, Roberts C, McCleery J, Pickett J, Bunn F, Challis D, Charlesworth G, Featherstone K, Fox C, Goodman C, Jones R, Lamb S, Moniz-Cook E, Schneider J, Shepperd S, Surr C, Thompson-Coon J, Ballard C, Brayne C, Burke O, Burns A, Clare L, Garrard P, Kehoe P, Passmore P, Holmes C, Maidment I, Murtagh F, Robinson L, Livingston G. Development of a core outcome set for disease modification trials in mild to moderate dementia: a systematic review, patient and public consultation and consensus recommendations. Health Technol Assess 2017; 21:1-192. [PMID: 28625273 PMCID: PMC5494514 DOI: 10.3310/hta21260] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND There is currently no disease-modifying treatment available to halt or delay the progression of the disease pathology in dementia. An agreed core set of the best-available and most appropriate outcomes for disease modification would facilitate the design of trials and ensure consistency across disease modification trials, as well as making results comparable and meta-analysable in future trials. OBJECTIVES To agree a set of core outcomes for disease modification trials for mild to moderate dementia with the UK dementia research community and patient and public involvement (PPI). DATA SOURCES We included disease modification trials with quantitative outcomes of efficacy from (1) references from related systematic reviews in workstream 1; (2) searches of the Cochrane Dementia and Cognitive Improvement Group study register, Cochrane Central Register of Controlled Trials, Cumulative Index to Nursing and Allied Health Literature, EMBASE, Latin American and Caribbean Health Sciences Literature and PsycINFO on 11 December 2015, and clinical trial registries [International Standard Randomised Controlled Trial Number (ISRCTN) and clinicaltrials.gov] on 22 and 29 January 2016; and (3) hand-searches of reference lists of relevant systematic reviews from database searches. REVIEW METHODS The project consisted of four workstreams. (1) We obtained related core outcome sets and work from co-applicants. (2) We systematically reviewed published and ongoing disease modification trials to identify the outcomes used in different domains. We extracted outcomes used in each trial, recording how many used each outcome and with how many participants. We divided outcomes into the domains measured and searched for validation data. (3) We consulted with PPI participants about recommended outcomes. (4) We presented all the synthesised information at a conference attended by the wider body of National Institute for Health Research (NIHR) dementia researchers to reach consensus on a core set of outcomes. RESULTS We included 149 papers from the 22,918 papers screened, referring to 125 individual trials. Eighty-one outcomes were used across trials, including 72 scales [31 cognitive, 12 activities of daily living (ADLs), 10 global, 16 neuropsychiatric and three quality of life] and nine biological techniques. We consulted with 18 people for PPI. The conference decided that only cognition and biological markers are core measures of disease modification. Cognition should be measured by the Mini Mental State Examination (MMSE) or the Alzheimer's Disease Assessment Scale - Cognitive subscale (ADAS-Cog), and brain changes through structural magnetic resonance imaging (MRI) in a subset of participants. All other domains are important but not core. We recommend using the Neuropsychiatric Inventory for neuropsychiatric symptoms: the Disability Assessment for Dementia for ADLs, the Dementia Quality of Life Measure for quality of life and the Clinical Dementia Rating scale to measure dementia globally. LIMITATIONS Most of the trials included participants with Alzheimer's disease, so recommendations may not apply to other types of dementia. We did not conduct economic analyses. The PPI consultation was limited to members of the Alzheimer's Society Research Network. CONCLUSIONS Cognitive outcomes and biological markers form the core outcome set for future disease modification trials, measured by the MMSE or ADAS-Cog, and structural MRI in a subset of participants. FUTURE WORK We envisage that the core set may be superseded in the future, particularly for other types of dementia. There is a need to develop an algorithm to compare scores on the MMSE and ADAS-Cog. STUDY REGISTRATION The project was registered with Core Outcome Measures in Effectiveness Trials [ www.comet-initiative.org/studies/details/819?result=true (accessed 7 April 2016)]. The systematic review protocol is registered as PROSPERO CRD42015027346. FUNDING The National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Lucy Webster
- Division of Psychiatry, University College London, London, UK
| | - Derek Groskreutz
- Division of Psychology and Language Sciences, University College London, London, UK
| | | | - Rob Howard
- Division of Psychiatry, University College London, London, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Gail Mountain
- School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - Sube Banerjee
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Bob Woods
- Dementia Services Development Centre Wales, Bangor University, Bangor, UK
| | - Robert Perneczky
- Faculty of Medicine, School of Public Health, Imperial College London, London, UK
| | - Louise Lafortune
- Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Charlotte Roberts
- International Consortium for Health Outcomes Measurement, London, UK
| | | | | | - Frances Bunn
- Centre for Research in Primary and Community Care, University of Hertfordshire, Hatfield, UK
| | - David Challis
- Personal Social Services Research Unit, University of Manchester, Manchester, UK
| | - Georgina Charlesworth
- Research Department of Clinical, Educational, and Health Psychology, University College London, London, UK
| | | | - Chris Fox
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Claire Goodman
- Centre for Research in Primary and Community Care, University of Hertfordshire, Hatfield, UK
| | - Roy Jones
- Research Institute for the Care of Older People, University of Bath, Bath, UK
| | - Sallie Lamb
- Oxford Clinical Trials Research Unit, University of Oxford, Oxford, UK
| | - Esme Moniz-Cook
- Faculty of Health and Social Care, University of Hull, Hull, UK
| | - Justine Schneider
- Institute of Mental Health, University of Nottingham, Nottingham, UK
| | - Sasha Shepperd
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Claire Surr
- School of Health & Community Studies, Leeds Beckett University, Leeds, UK
| | - Jo Thompson-Coon
- Collaboration for Leadership in Applied Health Research and Care South West Peninsula, University of Exeter, Exeter, UK
| | - Clive Ballard
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Carol Brayne
- Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Orlaith Burke
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Alistair Burns
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Linda Clare
- Collaboration for Leadership in Applied Health Research and Care South West Peninsula, University of Exeter, Exeter, UK
- School of Psychology, University of Exeter, Exeter, UK
- Centre for Research in Ageing and Cognitive Health, University of Exeter Medical School, Exeter, UK
| | - Peter Garrard
- Neuroscience Research Centre, St George's, University of London, UK
| | - Patrick Kehoe
- School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Peter Passmore
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Clive Holmes
- School of Medicine, University of Southampton, Southampton, UK
| | - Ian Maidment
- Aston Research Centre for Healthy Ageing, Aston University, Birmingham, UK
| | - Fliss Murtagh
- Cicely Saunders Institute, King's College London, London, UK
| | - Louise Robinson
- Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Gill Livingston
- Division of Psychiatry, University College London, London, UK
- Camden and Islington NHS Foundation Trust, London, UK
- North Thames Collaboration for Leadership in Applied Health Research and Care, London, UK
| |
Collapse
|
18
|
Day GS, Musiek ES, Roe CM, Norton J, Goate AM, Cruchaga C, Cairns NJ, Morris JC. Phenotypic Similarities Between Late-Onset Autosomal Dominant and Sporadic Alzheimer Disease: A Single-Family Case-Control Study. JAMA Neurol 2016; 73:1125-32. [PMID: 27454811 PMCID: PMC5025942 DOI: 10.1001/jamaneurol.2016.1236] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
IMPORTANCE The amyloid hypothesis posits that disrupted β-amyloid homeostasis initiates the pathological process resulting in Alzheimer disease (AD). Autosomal dominant AD (ADAD) has an early symptomatic onset and is caused by single-gene mutations that result in overproduction of β-amyloid 42. To the extent that sporadic late-onset AD (LOAD) also results from dysregulated β-amyloid 42, the clinical phenotypes of ADAD and LOAD should be similar when controlling for the effects of age. OBJECTIVE To use a family with late-onset ADAD caused by a presenilin 1 (PSEN1) gene mutation to mitigate the potential confound of age when comparing ADAD and LOAD. DESIGN, SETTING, AND PARTICIPANTS This case-control study was conducted at the Knight Alzheimer Disease Research Center at Washington University, St Louis, Missouri, and other National Institutes of Aging-funded AD centers in the United States. Ten PSEN1 A79V mutation carriers from multiple generations of a family with late-onset ADAD and 12 noncarrier family members were followed up at the Knight Alzheimer Disease Research Center (1985-2015) and 1115 individuals with neuropathologically confirmed LOAD were included from the National Alzheimer Coordinating Center database (September 2005-December 2014). Data analysis was completed in January 2016, including Knight Alzheimer Disease Research Center patient data collected up until the end of 2015. MAIN OUTCOMES AND MEASURES Planned comparison of clinical characteristics between cohorts, including age at symptom onset, associated symptoms and signs, rates of progression, and disease duration. RESULTS Of the PSEN1 A79V carriers in the family with late-onset ADAD, 4 were female (57%); among those with LOAD, 529 were female (47%). Seven mutation carriers (70%) developed AD dementia, while 3 were yet asymptomatic in their seventh and eighth decades of life. No differences were observed between mutation carriers and individuals with LOAD concerning age at symptom onset (mutation carriers: mean, 75 years [range, 63-77 years] vs those with LOAD: mean, 74 years [range, 60-101 years]; P = .29), presenting symptoms (memory loss in 7 of 7 mutation carriers [100%] vs 958 of 1063 individuals with LOAD [90.1%]; P ≥ .99) and duration (mutation carriers: mean, 9.9 years [range, 2.3-12.8 years] vs those with LOAD: 9 years [range, 1-27 years]; P = .73), and rate of progression of dementia (median annualized change in Clinical Dementia Rating-Sum of Boxes score, mutation carriers: 1.2 [range, 0.1-3.3] vs those with LOAD: 1.9 [range, -3.5 to 11.9]; P = .73). Early emergence of comorbid hallucinations and delusions were observed in 57% of individuals with ADAD (4 of 7) vs 19% of individuals with LOAD (137 of 706) (P = .03). Three of 12 noncarriers (25%) from the PSEN1 A79V family are potential phenocopies as they also developed AD dementia (median age at onset, 76.0 years). CONCLUSIONS AND RELEVANCE In this family, the amyloidogenic PSEN1 A79V mutation recapitulates the clinical attributes of LOAD. Previously reported clinical phenotypic differences between individuals with ADAD and LOAD may reflect age- or mutation-dependent effects.
Collapse
Affiliation(s)
- Gregory S Day
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Erik S Musiek
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Catherine M Roe
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Joanne Norton
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri3Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Alison M Goate
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri3Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Carlos Cruchaga
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri3Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - Nigel J Cairns
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri4Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - John C Morris
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, St Louis, Missouri2Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
19
|
Hassenstab J, Chasse R, Grabow P, Benzinger TLS, Fagan AM, Xiong C, Jasielec M, Grant E, Morris JC. Certified normal: Alzheimer's disease biomarkers and normative estimates of cognitive functioning. Neurobiol Aging 2016; 43:23-33. [PMID: 27255812 DOI: 10.1016/j.neurobiolaging.2016.03.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 12/31/2015] [Accepted: 03/13/2016] [Indexed: 12/23/2022]
Abstract
Normative samples drawn from older populations may unintentionally include individuals with preclinical Alzheimer's disease (AD) pathology, resulting in reduced means, increased variability, and overestimation of age effects on cognitive performance. A total of 264 cognitively normal (Clinical Dementia Rating = 0) older adults were classified as biomarker negative ("Robust Normal," n = 177) or biomarker positive ("Preclinical Alzheimer's Disease" [PCAD], n = 87) based on amyloid imaging, cerebrospinal fluid biomarkers, and hippocampal volumes. PCAD participants performed worse than robust normals on nearly all cognitive measures. Removing PCAD participants from the normative sample yielded higher means and less variability on episodic memory, visuospatial ability, and executive functioning measures. These results were more pronounced in participants aged 75 years and older. Notably, removing PCAD participants from the sample significantly reduced age effects across all cognitive domains. Applying norms from the robust normal sample to a separate cohort did not improve Clinical Dementia Rating classification when using standard deviation cutoff scores. Overall, removing individuals with biomarker evidence of preclinical AD improves normative sample quality and substantially reduces age effects on cognitive performance but provides no substantive benefit for diagnostic classifications.
Collapse
Affiliation(s)
- Jason Hassenstab
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychological & Brian Sciences, Washington University in St. Louis, St. Louis, MO, USA.
| | - Rachel Chasse
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Perri Grabow
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychology, University of Missouri-St. Louis, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurological Surgery, University of Missouri-St. Louis, St. Louis, MO, USA
| | - Anne M Fagan
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mateusz Jasielec
- Department of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth Grant
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Department of Physical Therapy, Washington University School of Medicine, St. Louis, MO, USA; Department of Occupational Therapy, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
20
|
Kahle-Wrobleski K, Fillit H, Kurlander J, Reed C, Belger M. Methodological challenges in assessing the impact of comorbidities on costs in Alzheimer's disease clinical trials. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2015; 16:995-1004. [PMID: 25410743 PMCID: PMC4646926 DOI: 10.1007/s10198-014-0648-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/23/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is associated with considerable costs and has a significant impact on health and social care systems. OBJECTIVE This study assessed whether baseline comorbidities present in 2,594 patients with AD participating in two semagacestat randomized placebo-controlled trials (RCTs) would significantly impact overall costs. METHODS Resource utilization was captured using the Resource Utilization in Dementia Scale-Lite. Comorbidities and concomitant medications were tabulated via patient and caregiver reports. Only baseline data were analyzed. Direct and indirect costs per month were calculated per patient. The relationship between cost and explanatory variables was explored in a regression model. RESULTS The baseline monthly cost of care in this RCT population was £1,147 ± 2,483, with informal care costs accounting for 75% of costs. Gender, age, and functional status were significant predictors of costs (p ≤ 0.0001). The cost ratio was not impacted when the number of comorbidities was added to the model (cost ratio = 0.95; 95% CI 0.91-0.99) or when combined with the number of concomitant medications (cost ratio = 0.97; 95% CI 0.95-1.00). Inconsistent findings related to the impact of individual comorbidities on costs were noted in sensitivity analyses. CONCLUSIONS The number of comorbidities, alone or when combined with concomitant medications, did not impact baseline costs of care, perhaps because RCTs often enroll less severely ill and more medically stable patients. However, higher costs were consistently associated with greater functional impairment similar to non-RCT databases. Supplemental sources (e.g., claims databases) are likely needed to better estimate the effects of disease and treatment on costs of illness captured in RCTs for AD.
Collapse
Affiliation(s)
| | - Howard Fillit
- Alzheimer's Drug Discovery Foundation and Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Mark Belger
- Eli Lilly and Company Limited, Windlesham, UK
| |
Collapse
|
21
|
Pravatà E, Tavernier J, Parker R, Vavro H, Mintzer JE, Spampinato MV. The neural correlates of anomia in the conversion from mild cognitive impairment to Alzheimer's disease. Neuroradiology 2015; 58:59-67. [PMID: 26400852 DOI: 10.1007/s00234-015-1596-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/14/2015] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Language impairment is frequently observed in patients with Alzheimer's disease (AD): in this study, we investigated the extent and distribution of brain atrophy in subjects with conversion from mild cognitive impairment (MCI) to AD with and without naming difficulties. METHODS This study was approved by the institutional review board and was HIPAA compliant. All subjects or their legal representatives gave informed consent for participation. Ninety-one subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) with (N = 51) and without (N = 40) naming impairment as per the Boston Naming Test (BNT), underwent brain magnetic resonance (MR) imaging 12 months before, at AD diagnosis, and 12 months after. Structural MR images were processed using voxel-based morphometry. Cross-sectional comparisons and mixed ANOVA models for assessing regional gray matter (GM) volume differences were performed. RESULTS As from 12 months prior to AD diagnosis, patients with naming difficulties showed distinct areas of greater GM loss in the left fusiform gyrus (Brodmann area 20) than patients without naming difficulties. Differences in the GM atrophy extended to the left hemisphere in the subsequent 12 months. CONCLUSION This study provided evidence of distinct patterns and dynamics of brain atrophy in AD patients with naming difficulties when compared to those with intact language, as early as 12 months prior to AD diagnosis and in the subsequent 12 months.
Collapse
Affiliation(s)
- Emanuele Pravatà
- Department of Radiology and Radiological Sciences, Medical University of South Carolina, Charleston, SC, USA. .,Department of Neuroradiology, Neurocenter of Southern Switzerland, Ospedale Regionale di Lugano, Via Tesserete 46, 6900, Lugano, Switzerland.
| | - Joshua Tavernier
- Department of Radiology and Radiological Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Ryan Parker
- Department of Anesthesiology, Vanderbilt University, Nashville, TN, USA
| | | | - Jacobo E Mintzer
- Clinical Biotechnology Research Institute, Roper Hospital, Charleston, SC, USA
| | - Maria Vittoria Spampinato
- Department of Radiology and Radiological Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
22
|
Tarawneh R, Head D, Allison S, Buckles V, Fagan AM, Ladenson JH, Morris JC, Holtzman DM. Cerebrospinal Fluid Markers of Neurodegeneration and Rates of Brain Atrophy in Early Alzheimer Disease. JAMA Neurol 2015; 72:656-65. [PMID: 25867677 DOI: 10.1001/jamaneurol.2015.0202] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
IMPORTANCE Measures of neuronal loss are likely good surrogates for clinical and radiological disease progression in Alzheimer disease (AD). Cerebrospinal fluid (CSF) markers of neuronal injury or neurodegeneration may offer usefulness in predicting disease progression and guiding outcome assessments and prognostic decisions in clinical trials of disease-modifying therapies. Visinin-like protein 1 (VILIP-1) has demonstrated potential usefulness as a marker of neuronal injury in AD. OBJECTIVE To investigate the usefulness of CSF VILIP-1, tau, p-tau181, and Aβ42 levels in predicting rates of whole-brain and regional atrophy in early AD and cognitively normal control subjects over time. DESIGN, SETTING, AND PARTICIPANTS Longitudinal observational study of brain atrophy in participants with early AD and cognitively normal controls. Study participants had baseline CSF biomarker measurements and longitudinal magnetic resonance imaging assessments for a mean follow-up period of 2 to 3 years. Mixed linear models assessed the ability of standardized baseline CSF biomarker measures to predict rates of whole-brain and regional atrophy over the follow-up period. The setting was The Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine in St Louis. Participants (mean age, 72.6 years) were individuals with a clinical diagnosis of very mild AD (n = 23) and cognitively normal controls (n = 64) who were enrolled in longitudinal studies of healthy aging and dementia. The study dates were 2000 to 2010. MAIN OUTCOMES AND MEASURES Correlations between baseline CSF biomarker measures and rates of whole-brain or regional atrophy in the AD and control cohorts over the follow-up period. RESULTS Baseline CSF VILIP-1, tau, and p-tau181 levels (but not Aβ42 levels) predicted rates of whole-brain and regional atrophy in AD over the follow-up period. Baseline CSF VILIP-1 levels predicted whole-brain (P = .006), hippocampal (P = .01), and entorhinal (P = .001) atrophy rates at least as well as tau and p-tau181 in early AD. Cognitively normal controls whose CSF VILIP-1, tau, or p-tau181 levels were in the upper tercile had higher rates of whole-brain (P = .02, P = .003, and P = .02, respectively), hippocampal (P = .001, P = .01, and P = .02, respectively), and entorhinal (P = .007, P = .01, and P = .01, respectively) atrophy compared with those whose levels were in the lower 2 terciles. CONCLUSIONS AND RELEVANCE Cerebrospinal fluid VILIP-1 levels predict rates of whole-brain and regional atrophy similarly to tau and p-tau181 and may provide a useful CSF biomarker surrogate for neurodegeneration in early symptomatic and preclinical AD.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri2Hope Center for Neurological Disorders, Washington University School of Medicine in St Louis, St Louis, Missouri3The Charles F. and Joanne Knight Alzheimer's
| | - Denise Head
- The Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine in St Louis, St Louis, Missouri5Department of Radiology, Washington University School of Medicine in St Louis, St Louis, Missouri6Department of
| | - Samantha Allison
- Department of Psychology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Virginia Buckles
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri3The Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri2Hope Center for Neurological Disorders, Washington University School of Medicine in St Louis, St Louis, Missouri3The Charles F. and Joanne Knight Alzheimer's
| | - Jack H Ladenson
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - John C Morris
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri3The Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine in St Louis, St Louis, Missouri7Department of
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri2Hope Center for Neurological Disorders, Washington University School of Medicine in St Louis, St Louis, Missouri3The Charles F. and Joanne Knight Alzheimer's
| |
Collapse
|
23
|
Manero RM, Casals-Coll M, Sánchez-Benavides G, Rodríguez-de los Reyes ON, Aguilar M, Badenes D, Molinuevo JL, Robles A, Barquero MS, Antúnez C, Martínez-Parra C, Frank-García A, Fernández M, Blesa R, Peña-Casanova J. Diagnostic validity of the Alzheimer's disease functional assessment and change scale in mild cognitive impairment and mild to moderate Alzheimer's disease. Dement Geriatr Cogn Disord 2015; 37:366-75. [PMID: 24556708 DOI: 10.1159/000350800] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The Alzheimer's Disease Functional Assessment and Change Scale (ADFACS) is a functional assessment instrument widely used in clinical research. AIMS To test the diagnostic and concurrent validity of the Spanish version of this scale and to describe the functional deficit pattern for mild cognitive impairment (MCI) and Alzheimer's disease (AD) dementia. METHODS The ADFACS, the Interview for Deterioration in Daily Living Activities in Dementia (IDDD), and the Mini Mental State Examination (MMSE) were administered to 146 control subjects (CS) and 165 patients (67 MCI and 98 AD). Nonparametric tests were used to compare the diagnostic groups. Cronbach's α and correlations with the MMSE and the IDDD were calculated. Sensitivity, specificity and predictive values were studied. RESULTS The ADFACS had a high internal consistency (α = 0.95). Three cutoff points of 1, 4, and 17 were provided to separate CS and MCI patients, MCI and mild AD patients, and mild AD and moderate AD patients, respectively. The ADFACS strongly correlated with functional (IDDD, 0.927) and cognitive (MMSE, 0.747) measures. A similar pattern of dysfunction, but in different grades, was found for the MCI and AD groups. CONCLUSION The ADFACS is a reliable, valid, and sensitive instrument to assess functional abilities; it is useful in dementia assessment for elderly populations.
Collapse
Affiliation(s)
- R M Manero
- Service of Neurology and Section of Behavioral Neurology and Dementias, Hospital del Mar, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
The effect of APOE ε4 allele on cholinesterase inhibitors in patients with Alzheimer disease: evaluation of the feasibility of resting state functional connectivity magnetic resonance imaging. Alzheimer Dis Assoc Disord 2015; 28:122-7. [PMID: 24830360 DOI: 10.1097/wad.0b013e318299d096] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This work is to determine whether apolipoprotein E (APOE) genotype modulates the effect of cholinesterase inhibitor (ChEI) treatment on resting state functional connectivity magnetic resonance imaging (rs-fcMRI) in patients with Alzheimer disease (AD). We retrospectively studied very mild and mild AD participants who were treated (N=25) or untreated (N=19) with ChEIs with respect to rs-fcMRI measure of 5 resting state networks (RSNs): default mode, dorsal attention (DAN), control (CON), salience (SAL), and sensory motor. For each network, a composite score was computed as the mean of Pearson correlations between pairwise time courses extracted from areas comprising this network. The composite scores were analyzed as a function of ChEI treatment and APOE ε4 allele. Across all participants, significant interactions between ChEI treatment and APOE ε4 allele were observed for all 5 RSNs. Within APOE ε4 carriers, significantly greater composite scores were observed in the DAN, CON, and SAL for treated compared with untreated participants. Within APOE ε4 noncarriers, treated and untreated participants did not have significantly different composite scores for all RSNs. These data suggest that APOE genotype affects the response to ChEI using rs-fcMRI. Rs-fcMRI may be useful for assessing the therapeutic effect of medications in AD clinical trials.
Collapse
|
25
|
Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, Joffe S, Kieburtz K, Raman R, Sun X, Aisen PS, Siemers E, Liu-Seifert H, Mohs R. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer's disease. N Engl J Med 2014; 370:311-21. [PMID: 24450890 DOI: 10.1056/nejmoa1312889] [Citation(s) in RCA: 1149] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Alzheimer's disease is characterized by amyloid-beta plaques, neurofibrillary tangles, gliosis, and neuronal loss. Solanezumab, a humanized monoclonal antibody, preferentially binds soluble forms of amyloid and in preclinical studies promoted its clearance from the brain. METHODS In two phase 3, double-blind trials (EXPEDITION 1 and EXPEDITION 2), we randomly assigned 1012 and 1040 patients, respectively, with mild-to-moderate Alzheimer's disease to receive placebo or solanezumab (administered intravenously at a dose of 400 mg) every 4 weeks for 18 months. The primary outcomes were the changes from baseline to week 80 in scores on the 11-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog11; range, 0 to 70, with higher scores indicating greater cognitive impairment) and the Alzheimer's Disease Cooperative Study-Activities of Daily Living scale (ADCS-ADL; range, 0 to 78, with lower scores indicating worse functioning). After analysis of data from EXPEDITION 1, the primary outcome for EXPEDITION 2 was revised to the change in scores on the 14-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog14; range, 0 to 90, with higher scores indicating greater impairment), in patients with mild Alzheimer's disease. RESULTS Neither study showed significant improvement in the primary outcomes. The modeled difference between groups (solanezumab group minus placebo group) in the change from baseline was -0.8 points for the ADAS-cog11 score (95% confidence interval [CI], -2.1 to 0.5; P=0.24) and -0.4 points for the ADCS-ADL score (95% CI, -2.3 to 1.4; P=0.64) in EXPEDITION 1 and -1.3 points (95% CI, -2.5 to 0.3; P=0.06) and 1.6 points (95% CI, -0.2 to 3.3; P=0.08), respectively, in EXPEDITION 2. Between-group differences in the changes in the ADAS-cog14 score were -1.7 points in patients with mild Alzheimer's disease (95% CI, -3.5 to 0.1; P=0.06) and -1.5 in patients with moderate Alzheimer's disease (95% CI, -4.1 to 1.1; P=0.26). In the combined safety data set, the incidence of amyloid-related imaging abnormalities with edema or hemorrhage was 0.9% with solanezumab and 0.4% with placebo for edema (P=0.27) and 4.9% and 5.6%, respectively, for hemorrhage (P=0.49). CONCLUSIONS Solanezumab, a humanized monoclonal antibody that binds amyloid, failed to improve cognition or functional ability. (Funded by Eli Lilly; EXPEDITION 1 and 2 ClinicalTrials.gov numbers, NCT00905372 and NCT00904683.).
Collapse
Affiliation(s)
- Rachelle S Doody
- From the Alzheimer's Disease and Memory Disorders Center, Department of Neurology, Baylor College of Medicine, Houston (R.S.D.); Alzheimer's Disease Cooperative Study, Department of Family and Preventive Medicine (R.G.T., R.R., X.S.), and the Department of Neurosciences (R.G.T., R.R., P.S.A., R.M.), University of California at San Diego, San Diego; Indiana Alzheimer Disease Center, Indiana University (M.F.), and Eli Lilly (E.S., H.L.-S., R.M.) - both in Indianapolis; the Department of Neuropathology, School of Medicine, and the Department of Neuropathology and Neuroscience, School of Pharmacological Science, University of Tokyo, Tokyo (T.I.); Gerontopole, Unité Mixte de Recherche 1027, Centre Hospitalier Universitaire Toulouse, Toulouse, France (B.V.); the Department of Medical Ethics and Health Policy, University of Pennsylvania, Philadelphia (S.J.); and the Center for Human Experimental Therapeutics, University of Rochester Medical Center, Rochester, NY (K.K.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
BACKGROUND New research criteria for preclinical Alzheimer's disease have been proposed, which include stages for cognitively normal individuals with abnormal amyloid markers (stage 1), abnormal amyloid and neuronal injury markers (stage 2), or abnormal amyloid and neuronal injury markers and subtle cognitive changes (stage 3). We aimed to investigate the prevalence and long-term outcome of preclinical Alzheimer's disease according to these criteria. METHODS Participants were cognitively normal (clinical dementia rating [CDR]=0) community-dwelling volunteers aged at least 65 years who were enrolled between 1998 and 2011 at the Washington University School of Medicine (MO, USA). CSF amyloid-β1-42 and tau concentrations and a memory composite score were used to classify participants as normal (both markers normal), preclinical Alzheimer's disease stage 1-3, or suspected non-Alzheimer pathophysiology (SNAP, abnormal injury marker without abnormal amyloid marker). The primary outcome was the proportion of participants in each preclinical AD stage. Secondary outcomes included progression to CDR at least 0·5, symptomatic Alzheimer's disease (score of at least 0·5 for memory and at least one other domain and cognitive impairments deemed to be due to Alzheimer's disease), and mortality. We undertook survival analyses using subdistribution and standard Cox hazards models and linear mixed models. FINDINGS Of 311 participants, 129 (41%) were classed as normal, 47 (15%) as stage 1, 36 (12%) as stage 2, 13 (4%) as stage 3, 72 (23%) as SNAP, and 14 (5%) remained unclassified. The 5-year progression rate to CDR at least 0·5, symptomatic Alzheimer's disease was 2% for participants classed as normal, 11% for stage 1, 26% for stage 2, 56% for stage 3, and 5% for SNAP. Compared with individuals classed as normal, participants with preclinical Alzheimer's disease had an increased risk of death after adjusting for covariates (hazard ratio 6·2, 95% CI 1·1-35·0; p=0·040). INTERPRETATION Preclinical Alzheimer's disease is common in cognitively normal elderly people and is associated with future cognitive decline and mortality. Thus, preclinical Alzheimer's disease could be an important target for therapeutic intervention. FUNDING National Institute of Aging of the National Institutes of Health (P01-AG003991, P50-AG05681, P01-AG02676), Internationale Stichting Alzheimer Onderzoek, the Center for Translational Molecular Medicine project LeARN, the EU/EFPIA Innovative Medicines Initiative Joint Undertaking, and the Charles and Joanne Knight Alzheimer Research Initiative.
Collapse
|
27
|
Doody RS, Raman R, Farlow M, Iwatsubo T, Vellas B, Joffe S, Kieburtz K, He F, Sun X, Thomas RG, Aisen PS, Siemers E, Sethuraman G, Mohs R. A phase 3 trial of semagacestat for treatment of Alzheimer's disease. N Engl J Med 2013; 369:341-50. [PMID: 23883379 DOI: 10.1056/nejmoa1210951] [Citation(s) in RCA: 860] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Alzheimer's disease is characterized by the presence of cortical amyloid-beta (Aβ) protein plaques, which result from the sequential action of β-secretase and γ-secretase on amyloid precursor protein. Semagacestat is a small-molecule γ-secretase inhibitor that was developed as a potential treatment for Alzheimer's disease. METHODS We conducted a double-blind, placebo-controlled trial in which 1537 patients with probable Alzheimer's disease underwent randomization to receive 100 mg of semagacestat, 140 mg of semagacestat, or placebo daily. Changes in cognition from baseline to week 76 were assessed with the use of the cognitive subscale of the Alzheimer's Disease Assessment Scale for cognition (ADAS-cog), on which scores range from 0 to 70 and higher scores indicate greater cognitive impairment, and changes in functioning were assessed with the Alzheimer's Disease Cooperative Study-Activities of Daily Living (ADCS-ADL) scale, on which scores range from 0 to 78 and higher scores indicate better functioning. A mixed-model repeated-measures analysis was used. RESULTS The trial was terminated before completion on the basis of a recommendation by the data and safety monitoring board. At termination, there were 189 patients in the group receiving placebo, 153 patients in the group receiving 100 mg of semagacestat, and 121 patients in the group receiving 140 mg of semagacestat. The ADAS-cog scores worsened in all three groups (mean change, 6.4 points in the placebo group, 7.5 points in the group receiving 100 mg of the study drug, and 7.8 points in the group receiving 140 mg; P=0.15 and P=0.07, respectively, for the comparison with placebo). The ADCS-ADL scores also worsened in all groups (mean change at week 76, -9.0 points in the placebo group, -10.5 points in the 100-mg group, and -12.6 points in the 140-mg group; P=0.14 and P<0.001, respectively, for the comparison with placebo). Patients treated with semagacestat lost more weight and had more skin cancers and infections, treatment discontinuations due to adverse events, and serious adverse events (P<0.001 for all comparisons with placebo). Laboratory abnormalities included reduced levels of lymphocytes, T cells, immunoglobulins, albumin, total protein, and uric acid and elevated levels of eosinophils, monocytes, and cholesterol; the urine pH was also elevated. CONCLUSIONS As compared with placebo, semagacestat did not improve cognitive status, and patients receiving the higher dose had significant worsening of functional ability. Semagacestat was associated with more adverse events, including skin cancers and infections. (Funded by Eli Lilly; ClinicalTrials.gov number, NCT00594568.)
Collapse
Affiliation(s)
- Rachelle S Doody
- Alzheimer's Disease and Memory Disorders Center, Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Recognition of positive vocalizations is impaired in behavioral-variant frontotemporal dementia. J Int Neuropsychol Soc 2013; 19:483-7. [PMID: 23369869 DOI: 10.1017/s1355617712001592] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Recognition of negative emotions is impaired in behavioral-variant frontotemporal dementia (bvFTD). Less is known about the identification of positive emotions. One limitation likely arises from the stimulus sets used in previous studies. The widely used Ekman 60 Faces Test, for example, consists of four negative emotions (anger, fear, disgust and sadness) but only one positive emotion (happiness). Here, patients with bvFTD (n = 9), AD (n = 9), and controls (n = 15) recognized a range of experimentally-validated positive and negative non-verbal vocalizations (e.g., cheers for triumph; retching for disgust) that have recently become available. The bvFTD group was impaired in the recognition of both positive and negative vocalizations. In contrast, performance in the AD cohort was comparable to that of controls. Findings in the bvFTD group point to a global emotion recognition deficit in this syndrome. These results are consistent with a growing body of research showing that deficits also extend to positive emotions.
Collapse
|
29
|
The experience of caregiving: differences between behavioral variant of frontotemporal dementia and Alzheimer disease. Am J Geriatr Psychiatry 2012; 20:724-8. [PMID: 21941168 PMCID: PMC4005886 DOI: 10.1097/jgp.0b013e318233154d] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To examine caregiver strain, depression, perceived sense of control, and distress from patient neuropsychiatric symptoms in family caregivers of persons with Alzheimer disease (AD) and behavioral variant of frontotemporal dementia (bvFTD) and determine whether group differences exist. METHODS Family caregivers were recruited from the Memory and Aging Center in San Francisco, California. Analyses of cross-sectional data on 53 family caregivers (AD = 31, bvFTD = 22) were performed. The Mann-Whitney U test was used to contrast groups. RESULTS There were statistically significant differences between the AD and bvFTD caregivers in strain, distress, and perceived control but not in depression. On average, bvFTD caregivers experienced greater strain and distress, more depressive symptoms, and lower perceived control. CONCLUSIONS Findings support that experiences of AD and bvFTD caregivers may differ. Further study is needed to identify possible explanatory factors for these group differences.
Collapse
|
30
|
Williams MM, Storandt M, Roe CM, Morris JC. Progression of Alzheimer's disease as measured by Clinical Dementia Rating Sum of Boxes scores. Alzheimers Dement 2012; 9:S39-44. [PMID: 22858530 DOI: 10.1016/j.jalz.2012.01.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 12/23/2011] [Accepted: 01/10/2012] [Indexed: 11/25/2022]
Abstract
BACKGROUND This study examined rates of dementia progression as ascertained by the Clinical Dementia Rating Sum of Boxes (CDR-SB) for symptomatic Alzheimer's disease (sAD), and assessed participant characteristics as predictors of CDR-SB progression. METHODS Participants (n = 792) were enrolled in longitudinal studies at an Alzheimer's Disease Research Center, received a diagnosis of sAD with a global CDR of 0.5 (n = 466) or 1 (n = 326), and had at least one follow-up assessment. Progression in CDR-SB over time as a function of baseline global CDR was examined. RESULTS A longitudinal increase (P < .0001) in CDR-SB was observed. The annual rate of change in CDR-SB scores was 1.43 (standard error [SE] = 0.05) in the CDR 0.5 sample and 1.91 (SE = 0.07) in the CDR 1 sample. For participants followed from the beginning of the CDR stage, time to progression to a higher global CDR was longer for individuals who were CDR 0.5 (3.75 years; 95% confidence interval [CI]: 3.18-4.33) than those who were CDR 1 at baseline (2.98 years; 95% CI: 2.75-3.22). In the total CDR 0.5 sample, the significant predictors of progression to the next global CDR stage (P < .01) were age at first sAD diagnosis and apolipoprotein E4 genotype. CONCLUSIONS The study findings are relevant to sAD clinical trial design and accurate, reliable ascertainment of the effect of disease-modifying treatments.
Collapse
Affiliation(s)
- Monique M Williams
- Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA.
| | | | | | | |
Collapse
|
31
|
Tarawneh R, Lee JM, Ladenson JH, Morris JC, Holtzman DM. CSF VILIP-1 predicts rates of cognitive decline in early Alzheimer disease. Neurology 2012; 78:709-19. [PMID: 22357717 DOI: 10.1212/wnl.0b013e318248e568] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Measures of neuronal damage/dysfunction are likely good surrogates for disease progression in Alzheimer disease (AD). CSF markers of neuronal injury may offer utility in predicting disease progression and guiding prognostic and outcome assessments in therapeutic trials. Visinin-like protein-1 (VILIP-1) has demonstrated potential utility as a marker of neuronal injury. We here investigate the utility of VILIP-1 and VILIP-1/Aβ42 in predicting rates of cognitive decline in early AD. METHODS Individuals with a clinical diagnosis of very mild or mild AD (n = 60) and baseline CSF measures of VILIP-1, tau, p-tau181, and Aβ42 were followed longitudinally for an average of 2.6 years. Annual assessments included the Clinical Dementia Rating (CDR), CDR-sum of boxes (CDR-SB), and global composite scores. Mixed linear models assessed the ability of CSF biomarker measures to predict rates of cognitive decline over time. RESULTS Baseline CSF VILIP-1 and VILIP-1/Aβ42 levels predicted rates of future decline in CDR-SB and global composite scores over the follow-up period. Individuals with CSF VILIP-1 ≥560 pg/mL (corresponding to the upper tercile) progressed much more rapidly in CDR-SB (1.61 boxes/year; p = 0.0077) and global scores (-0.53 points/year; p = 0.0002) than individuals with lower values (0.85 boxes/year and -0.15 points/year, respectively) over the follow-up period. CSF tau, p-tau181, tau/Aβ42, and p-tau181/Aβ42 also predicted more rapid cognitive decline in CDR-SB and global scores over time. CONCLUSION These findings suggest that CSF VILIP-1 and VILIP-1/Aβ42 predict rates of global cognitive decline similarly to tau and tau/Aβ42, and may be useful CSF surrogates for neurodegeneration in early AD.
Collapse
Affiliation(s)
- R Tarawneh
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | |
Collapse
|
32
|
|
33
|
Yoshida H, Terada S, Honda H, Kishimoto Y, Takeda N, Oshima E, Hirayama K, Yokota O, Uchitomi Y. Validation of the revised Addenbrooke's Cognitive Examination (ACE-R) for detecting mild cognitive impairment and dementia in a Japanese population. Int Psychogeriatr 2012; 24:28-37. [PMID: 21843399 DOI: 10.1017/s1041610211001190] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Early detection of dementia will be important for implementation of disease-modifying treatments in the near future. We aimed to investigate the diagnostic validity and reliability of the Japanese version of the revised Addenbrooke's Cognitive Examination (ACE-R J) for identifying mild cognitive impairment (MCI) and dementia. METHODS We translated and adapted the original ACE-R for use with a Japanese population. Standard tests for evaluating cognitive decline and dementing disorders were applied. A total of 242 subjects (controls = 73, MCI = 39, dementia = 130) participated in this study. RESULTS The optimal cut-off scores of ACE-R J for detecting MCI and dementia were 88/89 (sensitivity 0.87, specificity 0.92) and 82/83 (sensitivity 0.99, specificity 0.99) respectively. ACE-R J was superior to the Mini-Mental State Examination in the detection of MCI (area under the curve (AUC): 0.952 vs. 0.868), while the accuracy of the two instruments did not differ significantly in identifying dementia (AUC: 0.999 vs. 0.993). The inter-rater reliability (ICC = 0.999), test-retest reliability (ICC = 0.883), and internal consistency (Cronbach's α = 0.903) of ACE-R J were excellent. CONCLUSION ACE-R J proved to be an accurate cognitive instrument for detecting MCI and mild dementia. Further neuropsychological evaluation is required for the differential diagnosis of dementia subtypes.
Collapse
Affiliation(s)
- Hidenori Yoshida
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Tarawneh R, D'Angelo G, Macy E, Xiong C, Carter D, Cairns NJ, Fagan AM, Head D, Mintun MA, Ladenson JH, Lee JM, Morris JC, Holtzman DM. Visinin-like protein-1: diagnostic and prognostic biomarker in Alzheimer disease. Ann Neurol 2011; 70:274-85. [PMID: 21823155 DOI: 10.1002/ana.22448] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE There is a growing need to identify cerebrospinal fluid (CSF) markers that can detect Alzheimer's disease (AD) pathology in cognitively normal individuals because it is in this population that disease-modifying therapies may have the greatest chance of success. While AD pathology is estimated to begin ~10-15 years prior to the onset of cognitive decline, substantial neuronal loss is present by the time the earliest signs of cognitive impairment appear. Visinin-like protein-1 (VILIP-1) has demonstrated potential utility as a marker of neuronal injury. Here we investigate CSF VILIP-1 and VILIP-1/amyloid-β42 (Aβ42) ratio as diagnostic and prognostic markers in early AD. METHODS We assessed CSF levels of VILIP-1, tau, phosphorylated-tau181 (p-tau181), and Aβ42 in cognitively normal controls (CNC) (n = 211), individuals with early symptomatic AD (n = 98), and individuals with other dementias (n = 19). Structural magnetic resonance imaging (n = 192) and amyloid imaging with Pittsburgh Compound-B (n = 156) were obtained in subsets of this cohort. Among the CNC cohort, 164 individuals had follow-up annual cognitive assessments for 2-3 years. RESULTS CSF VILIP-1 levels differentiated individuals with AD from CNC and individuals with other dementias. CSF VILIP-1 levels correlated with CSF tau, p-tau181, and brain volumes in AD. VILIP-1 and VILIP-1/Aβ42 predicted future cognitive impairment in CNC over the follow-up period. Importantly, CSF VILIP-1/Aβ42 predicted future cognitive impairment at least as well as tau/Aβ42 and p-tau181/Aβ42. INTERPRETATION These findings suggest that CSF VILIP-1 and VILIP-1/Aβ42 offer diagnostic utility for early AD, and can predict future cognitive impairment in cognitively normal individuals similarly to tau and tau/Aβ42, respectively.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Suitability of the Clinical Dementia Rating‐Sum of Boxes as a single primary endpoint for Alzheimer's disease trials. Alzheimers Dement 2011; 7:602-610.e2. [DOI: 10.1016/j.jalz.2011.01.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Revised: 10/29/2010] [Accepted: 01/28/2011] [Indexed: 11/19/2022]
|
36
|
Spampinato MV, Rumboldt Z, Hosker RJ, Mintzer JE. Apolipoprotein E and gray matter volume loss in patients with mild cognitive impairment and Alzheimer disease. Radiology 2010; 258:843-52. [PMID: 21163916 DOI: 10.1148/radiol.10100307] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To examine the influence of apoliprotein E ε4 allele (APOE4) carrier status on disease progression by evaluating the rate of regional gray matter (GM) volume loss and disease severity in patients with newly diagnosed Alzheimer disease (AD) and stable amnestic mild cognitive impairment (MCI). MATERIALS AND METHODS This study was approved by the institutional review board and was HIPAA compliant. All subjects or their legal representatives gave informed consent for participation. Ninety-five subjects (63 male; average age, 77.1 years; age range, 58-91 years; 51 APOE4 carriers; 44 noncarriers) with either documented MCI to AD conversion or stable amnestic MCI underwent three yearly magnetic resonance imaging examinations. Voxel-based morphometry for image postprocessing and Clinical Dementia Rating (CDR) scale for cognitive assessment were used. RESULTS In APOE4 carriers, GM volume loss affected the hippocampi, temporal and parietal lobes, right caudate nucleus, and insulae in patients with MCI to AD conversion and the insular and temporal lobes in patients in whom MCI was stable. In subjects who were not APOE4 carriers, there was no significant GM volume change. There were no differences in CDR scores between APOE4 carriers and noncarriers. CONCLUSION APOE4 carriers with cognitive decline undergo faster GM atrophy than do noncarriers. The involvement of APOE4 in the progression of hippocampal atrophy, neocortical atrophy, or both has potential important implications for diagnosis and therapeutic approaches in patients with AD and should be considered in clinical trials. The present results and the results of prior studies indicate that the rate of hippocampal and neocortical atrophy is greater in association with APOE4 in nondemented elderly subjects, subjects with MCI, and those with AD.
Collapse
Affiliation(s)
- Maria Vittoria Spampinato
- Department of Radiology, Medical University of South Carolina, 96 Jonathan Lucas St, MSC 323, Charleston, SC, USA.
| | | | | | | | | |
Collapse
|
37
|
Autoregulation of cerebral blood flow to changes in arterial pressure in mild Alzheimer's disease. J Cereb Blood Flow Metab 2010; 30:1883-9. [PMID: 20736966 PMCID: PMC2972357 DOI: 10.1038/jcbfm.2010.135] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Studies in transgenic mice overexpressing amyloid precursor protein (APP) demonstrate impaired autoregulation of cerebral blood flow (CBF) to changes in arterial pressure and suggest that cerebrovascular dysfunction may be critically important in the development of pathological Alzheimer's disease (AD). Given the relevance of such a finding for guiding hypertension treatment in the elderly, we assessed autoregulation in individuals with AD. Twenty persons aged 75±6 years with very mild or mild symptomatic AD (Clinical Dementia Rating 0.5 or 1.0) underwent (15)O-positron emission tomography (PET) CBF measurements before and after mean arterial pressure (MAP) was lowered from 107±13 to 92±9 mm Hg with intravenous nicardipine; (11)C-PIB-PET imaging and magnetic resonance imaging (MRI) were also obtained. There were no significant differences in mean CBF before and after MAP reduction in the bilateral hemispheres (-0.9±5.2 mL per 100 g per minute, P=0.4, 95% confidence interval (CI)=-3.4 to 1.5), cortical borderzones (-1.9±5.0 mL per 100 g per minute, P=0.10, 95% CI=-4.3 to 0.4), regions of T2W-MRI-defined leukoaraiosis (-0.3±4.4 mL per 100 g per minute, P=0.85, 95% CI=-3.3 to 3.9), or regions of peak (11)C-PIB uptake (-2.5±7.7 mL per 100 g per minute, P=0.30, 95% CI=-7.7 to 2.7). The absence of significant change in CBF with a 10 to 15 mm Hg reduction in MAP within the normal autoregulatory range demonstrates that there is neither a generalized nor local defect of autoregulation in AD.
Collapse
|
38
|
Jelic V, Darreh-Shori T. Donepezil: A Review of Pharmacological Characteristics and Role in the Management of Alzheimer Disease. ACTA ACUST UNITED AC 2010. [DOI: 10.4137/cmt.s5410] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Donepezil is a potent, selective, noncompetitive, and rapidly reversible inhibitor of acetylcholinesterase (AChEI) licensed for the treatment of Alzheimer disease (AD); and is the first and only AChEI licensed for the treatment of severe AD. Its efficacy as monotherapy, or in combination with the NMDA-agonist, memantine, has been documented in several randomised double-blind, placebo-controlled, short-term clinical trials, as well as long-term extension trials and observational studies. Donepezil is a well tolerated drug that is generally safe as demonstrated even in patients with multiple co-morbidities receiving polypharmacy. It has been shown that donepezil improves cognition and global function in patients with mild-to-moderate AD; and long-term efficacy is maintained for up to 50 weeks. There is a dose-response relationship, with higher doses more likely to produce symptomatic benefit. Furthermore, donepezil-treated patients may improve cognitively and show global clinical improvement in all disease stages, including severe AD. Less consistent results in all disease stages were obtained on measures of function and behavior, and observations of mood. No effect on transition to AD has been found in long-term, randomized clinical trials in mild cognitive impairment (MCI). Cost-effectiveness of the treatment has been questioned by one long-term open-label societal study of 2-years duration. This study reported modest improvement of cognition but no statistically significant benefits during donepezil treatment as compared to placebo, in terms of rates of institutionalization and progression toward greater disability. However, there is a need for further research on clinically meaningful outcomes and treatment benefits favored by patients and caregivers, which are traditionally not defined as outcomes in clinical trials. Likewise, we need to know how to select responders, what is an optimal AChE inhibition particularly during the long-term treatment, in which patients the dosage should be increased for a sustained benefit, what is the optimal duration of treatment and when is meaningful to stop the treatment. After almost two decades of donepezil use in everyday clinical practice these issues are still unresolved.
Collapse
Affiliation(s)
- Vesna Jelic
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society (NVS); Division of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Taher Darreh-Shori
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society (NVS); Division of Alzheimer Neurobiology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
39
|
Abstract
BACKGROUND Since 1979, our clinicians have used an autobiographical memory task testing for events that occurred over the most recent week and most recent month in their semistructured interview when assessing for dementia. OBJECTIVE To examine correlations between scores on the autobiographical memory task and on 2 other commonly used brief memory tasks with results of a clinical assessment for dementia. DESIGN Correlation study. SETTING Academic research. PARTICIPANTS Participants were enrolled in Washington University Alzheimer Disease Research Center studies, were 60 years or older, and participated in assessments between May 29, 2002, and August 15, 2005 (N = 425). MAIN OUTCOME MEASURES Nonparametric Spearman rank correlations, adjusted for age and education status, between the Clinical Dementia Rating sum of boxes (CDR-SB) and scores on the autobiographical memory task and on 2 clinical brief memory tasks obtained from the Mini-Mental State Examination and the Short Blessed Test. RESULTS Scores on the autobiographical memory task and on each of the other 2 memory tasks correlated significantly with the CDR-SB (P < .001). Scores on the autobiographical memory task had a significantly higher correlation with results of the CDR-SB than the other 2 memory tasks (P < .001). CONCLUSION Clinicians may find the autobiographical memory task an important indicator of memory function and the autobiographical query a useful tool when assessing for dementia.
Collapse
Affiliation(s)
- Denise Maue Dreyfus
- Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63108, USA
| | | | | |
Collapse
|
40
|
Wilkinson D, Schindler R, Schwam E, Waldemar G, Jones RW, Gauthier S, Lopez OL, Cummings J, Xu Y, Feldman HH. Effectiveness of donepezil in reducing clinical worsening in patients with mild-to-moderate alzheimer's disease. Dement Geriatr Cogn Disord 2009; 28:244-51. [PMID: 19786776 PMCID: PMC3202931 DOI: 10.1159/000241877] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Therapeutic endpoints based on reduced clinical worsening represent clinically relevant and realistic goals for patients suffering from progressive neurodegenerative disorders such as Alzheimer's disease (AD). METHODS Data from 906 patients (388 receiving placebo; 518 receiving donepezil) with mild-to-moderate AD [Mini-Mental State Examination (MMSE) score 10-27] were pooled from 3 randomized, double-blind placebo-controlled studies. Clinical worsening was defined as decline in (1) cognition (MMSE), (2) cognition and global ratings (Clinician's Interview-Based Impression of Change plus Caregiver Input/Gottfries-Bråne-Steen scale) or (3) cognition, global ratings and function (various functional measures). RESULTS At week 24, lower percentages of donepezil-treated patients than placebo patients met the criteria for clinical worsening, regardless of the definition. The odds of declining were significantly reduced for donepezil-treated versus placebo patients (p < 0.0001; all definitions). Among patients meeting criteria for clinical worsening, mean declines in MMSE scores were greater for placebo than donepezil-treated patients. CONCLUSION In this population, donepezil treatment was associated with reduced odds of clinical worsening of AD symptoms. Moreover, patients worsening on donepezil were likely to experience less cognitive decline than expected if left untreated. This suggests that AD patients showing clinical worsening on donepezil may still derive benefits compared with placebo/untreated patients.
Collapse
Affiliation(s)
- David Wilkinson
- Memory Assessment and Research Centre, Moorgreen Hospital, Southampton, UK.
| | | | - Elias Schwam
- Pfizer Global Pharmaceuticals, Pfizer Inc., New York, N.Y., USA
| | - Gunhild Waldemar
- Memory Disorders Research Group, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Roy W. Jones
- Research Institute for the Care of Older People, Royal United Hospital, Bath, UK
| | - Serge Gauthier
- McGill Center for Studies in Aging, Douglas Mental Health University Institute, Douglas Hospital, Verdun, Que., Canada
| | - Oscar L. Lopez
- Alzheimer's Disease Research Center, Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, Pa., USA
| | - Jeffrey Cummings
- Mary S. Easton Center for Alzheimer's Disease Research, UCLA, Los Angeles, Calif., USA
| | - Yikang Xu
- Pfizer Global Pharmaceuticals, Pfizer Inc., New York, N.Y., USA
| | - Howard H. Feldman
- Division of Neurology, Clinic for Alzheimer's Disease and Related Disorders, University of British Columbia, Vancouver, B.C., Canada
| |
Collapse
|
41
|
Scales as outcome measures for Alzheimer's disease. Alzheimers Dement 2009; 5:324-39. [PMID: 19560103 DOI: 10.1016/j.jalz.2009.05.667] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 05/18/2009] [Indexed: 11/24/2022]
Abstract
The assessment of patient outcomes in clinical trials of new therapeutics for Alzheimer's disease (AD) continues to evolve. In addition to assessing drugs for symptomatic relief, an increasing number of trials are focusing on potential disease-modifying agents. Moreover, participants with AD are being studied earlier in their course of disease. As a result, the limitations of current outcome measures have become more apparent, as has the need for better instruments. In recognition of the need to review and possibly revise current assessment measures, the Alzheimer's Association, in cooperation with industry leaders and academic investigators, convened a Research Roundtable meeting devoted to scales as outcome measures for AD clinical trials. The meeting included a discussion of methodological issues in the use of scales in AD clinical trials, including cross-cultural issues. Specific topics related to the use of cognitive, functional, global, and neuropsychiatric scales were also presented. Speakers also addressed academic and industry initiatives for pooling data from untreated and placebo-treated patients in clinical trials. A number of regulatory topics were also discussed with agency representatives. Panel discussions highlighted areas of controversy, in an effort to gain consensus on various topics.
Collapse
|
42
|
Snider BJ, Fagan AM, Roe C, Shah AR, Grant EA, Xiong C, Morris JC, Holtzman DM. Cerebrospinal fluid biomarkers and rate of cognitive decline in very mild dementia of the Alzheimer type. ACTA ACUST UNITED AC 2009; 66:638-45. [PMID: 19433664 DOI: 10.1001/archneurol.2009.55] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) levels of Abeta peptide 1-42 (Abeta 42), tau, and phosphorylated tau (ptau) are potential biomarkers of Alzheimer disease. OBJECTIVE To determine whether Abeta 42, tau, and ptau predict the rate of cognitive change in individuals with very mild dementia of the Alzheimer type (DAT). DESIGN Retrospective analysis of CSF biomarkers and clinical data. SETTING An academic Alzheimer disease research center. PARTICIPANTS Research volunteers in a longitudinal study of aging and cognition. Participants (n = 49) had a clinical diagnosis of very mild DAT with a Clinical Dementia Rating (CDR) of 0.5 at the time of lumbar puncture. All the participants had at least 1 follow-up assessment (mean [SD] follow-up, 3.5 [1.8] years). MAIN OUTCOME MEASURES Baseline CSF levels of Abeta 42, Abeta 40, tau, and ptau at threonine 181 (ptau181) and the rate of dementia progression as measured using the CDR sum of boxes (CDR-SB) score and psychometric performance. RESULTS The rate of dementia progression was significantly more rapid in individuals with lower baseline CSF Abeta 42 levels, higher tau or ptau181 levels, or high tau: Abeta 42 ratios. For example, the annual change in the CDR-SB score was 1.1 for the lowest 2 tertiles of Abeta 42 values and 0.3 for the highest tertile of Abeta 42 values. CONCLUSIONS In individuals with very mild DAT, lower CSF Abeta 42 levels, high tau or ptau181 levels, or high tau:Abeta 42 ratios quantitatively predict more rapid progression of cognitive deficits and dementia. Biomarkers of CSF may be useful prognostically and to identify individuals who are more likely to progress for participation in therapeutic clinical trials.
Collapse
Affiliation(s)
- Barbara J Snider
- Department of Neurology, Washington University School of Medicine, Campus Box 8111, 660 S Euclid, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Informant-based dementia screening in a population-based sample of African Americans. Alzheimer Dis Assoc Disord 2009; 23:117-23. [PMID: 19484913 DOI: 10.1097/wad.0b013e318190a709] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND An informant-based screening tool for dementia may be useful in population-based studies of minority populations. OBJECTIVE Investigate the feasibility of screening for very mild dementia in a community sample of African Americans using an informant-based screening tool (AD8). DESIGN Cohort study. PARTICIPANTS One hundred forty-seven persons from the African American Health (AAH) project were screened for dementia; 61 of 93 who were invited had follow-up clinical assessments for dementia diagnosis. MEASUREMENTS The AD8, Mini-Mental State Examination, Short Blessed Test, Brief Instrument for Dementia Detection, and a neuropsychologic battery were administered at visit 1. The Clinical Dementia Rating (CDR) was administered at visit 2 by clinicians blinded to visit 1 results; the presence of dementia was determined by a CDR greater than 0. RESULTS Four hundred sixty-five individuals from the AAH cohort were sent a letter describing the study and, among this group, 252 individuals were contacted by phone to request participation in this study. Six percent (14/252) of the participants contacted by phone were unable to identify an informant (required for the AD8). One hundred fifty individuals agreed by phone to participate of which 2% (n=3) did not have an informant available at the time of participation. The AD8 alone was effective at discriminating between CDR 0 and CDR 0.5 (area under the curve=0.847; P<0.001; 95% confidence interval, 0.73-0.96). CONCLUSIONS A brief informant-based instrument, the AD8, has high sensitivity and specificity for distinguishing CDR 0 from CDR 0.5 in the community. Informant availability may not be a barrier for using the AD8 in an African American community sample; however, further study in larger samples with a higher response rate, different community settings (eg, community clinics), and among older age groups (eg, age 75+) is warranted to confirm this.
Collapse
|
44
|
Wilkins CH, Roe CM, Morris JC. A brief clinical tool to assess physical function: the mini-physical performance test. Arch Gerontol Geriatr 2009; 50:96-100. [PMID: 19282039 DOI: 10.1016/j.archger.2009.02.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Revised: 01/25/2009] [Accepted: 02/04/2009] [Indexed: 11/29/2022]
Abstract
The aim was to develop a brief physical performance assessment tool that can be reliably used to detect physical impairment in older adults with and without mild dementia. Scores on the 9-item physical performance test (PPT) from non-demented participants were used to develop and validate the 4-item mini-PPT. The validated mini-PPT was then used to predict total PPT score and functional physical status in participants with mild dementia. Receiver operating curve (ROC) analyses were used to generate a cutoff score that classifies participants as functional vs. not functional. The setting was in the Alzheimer's Disease Research Center (Washington University). A total of 1199 participants met inclusion criteria: 574 non-demented participants, 436 with very mild dementia, measured by the clinical dementia rating (CDR)=0.5 and 189 with mild dementia (CDR=1). The mean age of the sample was 76.4 years, mean educational attainment was 14 years, 58% were women, and 11% were African American. A 4-item scale, the mini-PPT, was developed (based on the results of multiple regression analyses and clinical meaningfulness) that highly correlated with total PPT score (r=0.917, p<0.0001) in the non-demented sample. The correlation of the mini-PPT with total PPT was 0.90 among those with very mild, and 0.91 among those with mild dementia. Using the ROCs, a cutoff score of 12 correctly classified at least 85% of non-demented and demented persons. The 4-item mini-PPT is highly correlated with the 9-item PPT in non-demented and mildly demented persons. This brief tool may be useful in detecting early physical impairment in the clinical setting.
Collapse
Affiliation(s)
- Consuelo H Wilkins
- Department of Medicine, Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO 63108, USA.
| | | | | |
Collapse
|
45
|
Roe CM, Mintun MA, D'Angelo G, Xiong C, Grant EA, Morris JC. Alzheimer disease and cognitive reserve: variation of education effect with carbon 11-labeled Pittsburgh Compound B uptake. ACTA ACUST UNITED AC 2008; 65:1467-71. [PMID: 19001165 DOI: 10.1001/archneur.65.11.1467] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
OBJECTIVE To evaluate the cognitive reserve hypothesis by examining whether individuals of greater educational attainment have better cognitive function than individuals with less education in the presence of elevated fibrillar brain amyloid levels. DESIGN, SETTING, AND PARTICIPANTS Uptake of carbon 11-labeled Pittsburgh Compound B ([(11)C]PiB) was measured for participants assessed between August 15, 2003, and January 8, 2008, at the Washington University Alzheimer's Disease Research Center and diagnosed either as nondemented (n = 161) or with dementia of the Alzheimer type (n = 37). Multiple regression was used to determine whether [(11)C]PiB uptake interacted with level of educational attainment to predict cognitive function. MAIN OUTCOME MEASURES Scores on the Clinical Dementia Rating sum of boxes, Mini-Mental State Examination, and Short Blessed Test and individual measures from a psychometric battery. RESULTS Uptake of [(11)C]PiB interacted with years of education in predicting scores on the Clinical Dementia Rating sum of boxes (P = .003), the Mini-Mental State Examination (P < .001), the Short Blessed Test (P = .03), and a measure of verbal abstract reasoning and conceptualization (P = .02) such that performance on these measures increased with increasing education for participants with elevated PiB uptake. Education was unrelated to global cognitive functioning scores among those with lower PiB uptake. CONCLUSION The results support the hypothesis that cognitive reserve influences the association between Alzheimer disease pathological burden and cognition.
Collapse
Affiliation(s)
- Catherine M Roe
- Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Winblad B, Gauthier S, Scinto L, Feldman H, Wilcock GK, Truyen L, Mayorga AJ, Wang D, Brashear HR, Nye JS. Safety and efficacy of galantamine in subjects with mild cognitive impairment. Neurology 2008; 70:2024-35. [PMID: 18322263 DOI: 10.1212/01.wnl.0000303815.69777.26] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the safety of galantamine in subjects with mild cognitive impairment (MCI), the ability of galantamine to benefit cognition and global functioning in subjects with MCI, and the ability of galantamine to delay conversion to dementia. METHODS In two studies, 2,048 subjects, 990 in Study 1 and 1,058 in Study 2, with a Clinical Dementia Rating (CDR) = 0.5, CDR memory score > or =0.5, without dementia were randomized to double-blind galantamine (16-24 mg/day) or placebo for 24 months. Primary efficacy endpoint at month 24 was number (%) of subjects who converted from MCI to dementia (CDR > or = 1.0). RESULTS There were no differences between galantamine and placebo in 24-month conversion rates (Study 1: 22.9% [galantamine] vs 22.6% [placebo], p = 0.146; Study 2: 25.4% [galantamine] vs 31.2% [placebo], p = 0.619). Mean CDR-sum of boxes declined less with galantamine than placebo at 12 and 24 months in Study 1 (p = 0.024 [12 months] and p = 0.028 [24 months]), but not in Study 2 (p = 0.662 [12 months] and p = 0.056 [24 months]). Digit Symbol Substitution Test scores improved with galantamine in Study 1 at 12 months and in Study 2 at 24 months (Study 1: p = 0.009 [month 12] and p = 0.079 [Month 24]; Study 2: p = 0.154 [month 12] and p = 0.020 [month 24]). The most frequently reported adverse event was nausea (galantamine, 29%; placebo, 10%). Serious AEs occurred in 19% of each group. Mortality of the cohort after retrospectively determining the status of subjects (98.3%) at 24 months was 1.4% (galantamine) and 0.3% (placebo); RR (95% CI), 1.70 (1.00, 2.90). CONCLUSIONS Galantamine failed to significantly influence conversion to dementia. Galantamine was generally well tolerated. Whereas recorded mortality was greater in the galantamine group than in the placebo group in the original per-protocol assessment, a post hoc analysis of the cohort was consistent with no increased risk.
Collapse
Affiliation(s)
- B Winblad
- Karolinska Institutet, Alzheimer's Disease Research Center, Division of Geriatric Medicine, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Improving linear modeling of cognitive decline in patients with mild cognitive impairment: comparison of two methods. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2007. [PMID: 17982900 DOI: 10.1007/978-3-211-73574-9_30] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
BACKGROUND High variability of estimates of cognitive decline in patients with Alzheimer's disease (AD) derived from unbalanced longitudinal designs may result as much from the applied statistical model as from true biological variability. OBJECTIVE To compare the accuracy of two statistical models, serial subtraction score (SSA) and mixed-effects regression analysis (MEM), to estimate rates of cognitive decline in patients with amnestic mild cognitive impairment (MCI), a group at risk for AD. METHODS We recorded serial mini mental state examination (MMSE) scores from 78 MCI patients. Additionally, we derived simulated trajectories of cognitive decline with unequally spaced observation intervals. Rates of change were assessed from clinical and simulated data using SSA and MEM models. RESULTS MEM reduced variability of rates of change significantly compared to SSA. In a polynomial model, overall length of observation time explained a significant amount of variance of SSA, but not of MEM estimates. For simulated data, MEM was significantly more accurate in predicting true rates of change compared to SSA (p < 0.001). CONCLUSION MEM yields more accurate estimates of cognitive decline from unbalanced longitudinal data. Simulation studies may be useful to select the appropriate statistical model for a given set of clinical data.
Collapse
|
48
|
Burns A, Gauthier S, Perdomo C. Efficacy and safety of donepezil over 3 years: an open-label, multicentre study in patients with Alzheimer's disease. Int J Geriatr Psychiatry 2007; 22:806-12. [PMID: 17199235 DOI: 10.1002/gps.1746] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE This 132-week, open-label extension study assessed the long-term efficacy and safety of donepezil in 579 patients with mild to moderate Alzheimer's disease (AD) who had previously participated in a 24-week double-blind study of 5 or 10 mg/day donepezil vs placebo. METHOD Patients enrolled in the present study had a 6-week single-blind placebo washout period followed by treatment with donepezil 5 mg/day for 6 weeks with an optional increase in dosage to 10 mg/day between weeks 6 and 32. RESULTS After 6 weeks of open-label treatment with donepezil 5 mg/day, mean Alzheimer's Disease Assessment Scale -- cognitive subscale scores (ADAS-cog) improved by approximately two points, while after 12 weeks of open-label treatment (with a majority of patients receiving 10 mg/day), the mean ADAS-cog score was 1 point better than the score at the end of the placebo washout period. Scores then declined gradually over the remainder of the study. Mean changes in Clinical Dementia Rating-Sum of Boxes scores showed slight improvement over the first 12 weeks of open-label treatment and then slowly declined for the remainder of the study period. Donepezil was well tolerated over the entire 162-week study period. Overall, 85% of patients experienced at least one adverse event (AE). The most common included diarrhoea (12%), nausea (11%), infection (11%) and accidental injury (10%). Some patients discontinued the study due to AEs (15%). CONCLUSIONS These results support the conclusion that donepezil is safe and effective for the long-term treatment of patients with mild to moderate AD.
Collapse
Affiliation(s)
- A Burns
- University of Manchster, Manchester, UK.
| | | | | |
Collapse
|
49
|
Wilkins CH, Sheline YI, Roe CM, Birge SJ, Morris JC. Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry 2006; 14:1032-40. [PMID: 17138809 DOI: 10.1097/01.jgp.0000240986.74642.7c] [Citation(s) in RCA: 288] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Vitamin D deficiency is common in older adults and has been implicated in psychiatric and neurologic disorders. This study examined the relationship among vitamin D status, cognitive performance, mood, and physical performance in older adults. METHODS A cross-sectional group of 80 participants, 40 with mild Alzheimer disease (AD) and 40 nondemented persons, were selected from a longitudinal study of memory and aging. Cognitive function was assessed using the Short Blessed Test (SBT), Mini-Mental State Exam (MMSE), Clinical Dementia Rating (CDR; a higher Sum of Boxes score indicates greater dementia severity), and a factor score from a neuropsychometric battery; mood was assessed using clinician's diagnosis and the depression symptoms inventory. The Physical Performance Test (PPT) was used to measure functional status. Serum 25-hydroxyvitamin D levels were measured for all participants. RESULTS The mean vitamin D level in the total sample was 18.58 ng/mL (standard deviation: 7.59); 58% of the participants had abnormally low vitamin D levels defined as less than 20 ng/mL. After adjusting for age, race, gender, and season of vitamin D determination, vitamin D deficiency was associated with presence of an active mood disorder (odds ratio: 11.69, 95% confidence interval: 2.04-66.86; Wald chi(2) = 7.66, df = 2, p = 0.022). Using the same covariates in a linear regression model, vitamin D deficiency was associated with worse performance on the SBT (F = 5.22, df = [2, 77], p = 0.044) and higher CDR Sum of Box scores (F = 3.20, df = [2, 77], p = 0.047) in the vitamin D-deficient group. There was no difference in performance on the MMSE, PPT, or factor scores between the vitamin D groups. CONCLUSIONS In a cross-section of older adults, vitamin D deficiency was associated with low mood and with impairment on two of four measures of cognitive performance.
Collapse
Affiliation(s)
- Consuelo H Wilkins
- Department of Medicine, Division of Geriatrics and Nutritional Science, Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA.
| | | | | | | | | |
Collapse
|
50
|
Lynch CA, Walsh C, Blanco A, Moran M, Coen RF, Walsh JB, Lawlor BA. The clinical dementia rating sum of box score in mild dementia. Dement Geriatr Cogn Disord 2006; 21:40-3. [PMID: 16254429 DOI: 10.1159/000089218] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/08/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Making an early diagnosis of dementia is becoming increasingly important, but is difficult in practice. The Clinical Dementia Rating (CDR) scale is a widely used dementia staging instrument, yielding a global score and a summated score (sum of box score). This study examines the utility of the CDR sum of box score, rather than the CDR global score, in making a diagnosis of early dementia. OBJECTIVE To determine whether the CDR sum of box score is predictive of an ICD-10 diagnosis of dementia in cases with mild cognitive deficits. METHODS Clinical data recorded on our Memory Clinic database were examined for all patients seen over a 6-year period. Data were extracted from 276 first visits in which patients had scored 0.5 using the CDR global score. We examined the relationship between CDR sum of box score and consensus diagnosis of dementia using logistic regression. RESULTS We found that increased CDR sum of box score was significantly associated with a higher probability of being assigned an ICD-10 diagnosis of dementia (p < 0.001). The odds ratio for the coefficient of CDR sum of box was 2.3 (95% CI 1.7-3.1), indicating that the likelihood of being diagnosed as having dementia increased by a factor of 2.3 for every point increase on the CDR sum of box score. CONCLUSION These findings indicate that the CDR sum of box score provides additional information to the CDR global score in mild cases. The CDR sum of box score is a helpful indicator in making/excluding a diagnosis of dementia in people with mild cognitive deficits.
Collapse
Affiliation(s)
- C A Lynch
- Mercer's Institute for Research on Ageing, St James's Hospital, Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|