1
|
Kleven BDC, Chien LC, Cross CL, Labus B, Bernick C. Traumatic Encephalopathy Syndrome: Head Impact Exposure and Blood Biomarkers in Professional Combat Athletes. J Head Trauma Rehabil 2025:00001199-990000000-00244. [PMID: 39998558 DOI: 10.1097/htr.0000000000001048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
OBJECTIVE This study aimed to (1) determine whether there was an association between a diagnosis of traumatic encephalopathy syndrome (TES) and changes in three specific serum biomarkers, and (2) determine head impact exposure thresholds among both TES+ and TES- groups. SETTING Data were collected from Cleveland Clinic's Professional Athletes Brain Health Study (PABHS). PARTICIPANTS This study included 192 professional combat athletes, 35 years of age and older. Athletes must be actively fighting or retired with a minimum of 10 professional fights over their careers. DESIGN/INTERVENTION This was a retrospective observational study of the PABHS longitudinal cohort. MAIN MEASURES The generalized linear model with the generalized estimating equation for repeated measurements was used to compare various biomarkers between both active and retired TES- and TES+ groups. RESULTS The odds ratio for TES diagnosis was 5.44 (95% CI = 2.48, 11.94; P < .0001) among active fighters and 10.75 (95% CI = 3.52, 32.85; P < .0001) among retired fighters, indicating the odds for a TES diagnosis were over 5 times greater for active fighters with every fight completed at or beyond 30 professional fights. Retired fighters had 10 times greater odds of TES diagnosis with every fight completed at or beyond 15 professional fights. Likewise, the odds of a TES diagnosis were 2.0% (95% CI = 0.3, 3.1; P = 0.0039) greater with each pg/mL increase of glial fibrillary acidic protein (GFAP). No relationship was observed between a TES diagnosis and neurofilament light chain or P-tau231. CONCLUSION This study provides preliminary evidence that progressively elevated levels of the GFAP blood biomarker increase the odds of a TES diagnosis among retired professional fighters. Further evaluation is required to improve clarity and understanding of the relationship between progressive changes in the GFAP blood biomarker and a TES diagnosis, specifically evaluating the duration of chronicity and exposure thresholds.
Collapse
Affiliation(s)
- Brooke D Conway Kleven
- Author Affiliations: Sports Innovation Institute (Dr Kleven), Department of Brain Health, Kirk Kerkorian School of Medicine (Dr Kleven), Department of Epidemiology and Biostatistics, School of Public Health (Dr Chien, Dr Cross, and Dr Labus), University of Nevada, Las Vegas, Las Vegas, Nevada; and Cleveland Clinic Lou Ruvo Center for Brain Health (Dr Bernick), Las Vegas, Nevada
| | | | | | | | | |
Collapse
|
2
|
Biljman K, Gozes I, Lam JCK, Li VOK. An experimental framework for conjoint measures of olfaction, navigation, and motion as pre-clinical biomarkers of Alzheimer's disease. J Alzheimers Dis Rep 2024; 8:1722-1744. [PMID: 40034341 PMCID: PMC11863766 DOI: 10.1177/25424823241307617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 11/19/2024] [Indexed: 03/05/2025] Open
Abstract
Elucidating Alzheimer's disease (AD) prodromal symptoms can resolve the outstanding challenge of early diagnosis. Based on intrinsically related substrates of olfaction and spatial navigation, we propose a novel experimental framework for their conjoint study. Artificial intelligence-driven multimodal study combining self-collected olfactory and motion data with available big clinical datasets can potentially promote high-precision early clinical screenings to facilitate timely interventions targeting neurodegenerative progression.
Collapse
Affiliation(s)
- Katarina Biljman
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Illana Gozes
- Elton Laboratory for Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, The Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Jacqueline CK Lam
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Victor OK Li
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
3
|
Mohammadi H, Ariaei A, Ghobadi Z, Gorgich EAC, Rustamzadeh A. Which neuroimaging and fluid biomarkers method is better in theranostic of Alzheimer's disease? An umbrella review. IBRO Neurosci Rep 2024; 16:403-417. [PMID: 38497046 PMCID: PMC10940808 DOI: 10.1016/j.ibneur.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/24/2024] [Indexed: 03/19/2024] Open
Abstract
Biomarkers are measured to evaluate physiological and pathological processes as well as responses to a therapeutic intervention. Biomarkers can be classified as diagnostic, prognostic, predictor, clinical, and therapeutic. In Alzheimer's disease (AD), multiple biomarkers have been reported so far. Nevertheless, finding a specific biomarker in AD remains a major challenge. Three databases, including PubMed, Web of Science, and Scopus were selected with the keywords of Alzheimer's disease, neuroimaging, biomarker, and blood. The results were finalized with 49 potential CSF/blood and 35 neuroimaging biomarkers. To distinguish normal from AD patients, amyloid-beta42 (Aβ42), plasma glial fibrillary acidic protein (GFAP), and neurofilament light (NFL) as potential biomarkers in cerebrospinal fluid (CSF) as well as the serum could be detected. Nevertheless, most of the biomarkers fairly change in the CSF during AD, listed as kallikrein 6, virus-like particles (VLP-1), galectin-3 (Gal-3), and synaptotagmin-1 (Syt-1). From the neuroimaging aspect, atrophy is an accepted biomarker for the neuropathologic progression of AD. In addition, Magnetic resonance spectroscopy (MRS), diffusion weighted imaging (DWI), diffusion tensor imaging (DTI), tractography (DTT), positron emission tomography (PET), and functional magnetic resonance imaging (fMRI), can be used to detect AD. Using neuroimaging and CSF/blood biomarkers, in combination with artificial intelligence, it is possible to obtain information on prognosis and follow-up on the different stages of AD. Hence physicians could select the suitable therapy to attenuate disease symptoms and follow up on the efficiency of the prescribed drug.
Collapse
Affiliation(s)
- Hossein Mohammadi
- Department of Bioimaging, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences (MUI), Isfahan, Islamic Republic of Iran
| | - Armin Ariaei
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Zahra Ghobadi
- Advanced Medical Imaging Ward, Pars Darman Medical Imaging Center, Karaj, Islamic Republic of Iran
| | - Enam Alhagh Charkhat Gorgich
- Department of Anatomy, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Islamic Republic of Iran
| | - Auob Rustamzadeh
- Cellular and Molecular Research Center, Research Institute for Non-communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
4
|
Abukuri DN. Novel Biomarkers for Alzheimer's Disease: Plasma Neurofilament Light and Cerebrospinal Fluid. Int J Alzheimers Dis 2024; 2024:6668159. [PMID: 38779175 PMCID: PMC11111307 DOI: 10.1155/2024/6668159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) represent an increasingly significant public health concern. As clinical diagnosis faces challenges, biomarkers are becoming increasingly important in research, trials, and patient assessments. While biomarkers like amyloid-β peptide, tau proteins, CSF levels (Aβ, tau, and p-tau), and neuroimaging techniques are commonly used in AD diagnosis, they are often limited and invasive in monitoring and diagnosis. For this reason, blood-based biomarkers are the optimal choice for detecting neurodegeneration in brain diseases due to their noninvasiveness, affordability, reliability, and consistency. This literature review focuses on plasma neurofilament light (NfL) and CSF NfL as blood-based biomarkers used in recent AD diagnosis. The findings revealed that the core CSF biomarkers of neurodegeneration (T-tau, P-tau, and Aβ42), CSF NFL, and plasma T-tau were strongly associated with Alzheimer's disease, and the core biomarkers were strongly associated with mild cognitive impairment due to Alzheimer's disease. Elevated levels of plasma and cerebrospinal fluid NfL were linked to decreased [18F]FDG uptake in corresponding brain areas. In participants with Aβ positivity (Aβ+), NfL correlated with reduced metabolism in regions susceptible to Alzheimer's disease. In addition, CSF NfL levels correlate with brain atrophy and predict cognitive changes, while plasma total tau does not. Plasma P-tau, especially in combination with Aβ42/Aβ40, is promising for symptomatic AD stages. Though not AD-exclusive, blood NfL holds promise for neurodegeneration detection and assessing treatment efficacy. Given the consistent levels of T-tau, P-tau, Aβ42, and NFL in CSF, their incorporation into both clinical practice and research is highly recommended.
Collapse
|
5
|
Rodriguez-Vieitez E, Kumar A, Malarte ML, Ioannou K, Rocha FM, Chiotis K. Imaging Neuroinflammation: Quantification of Astrocytosis in a Multitracer PET Approach. Methods Mol Biol 2024; 2785:195-218. [PMID: 38427196 DOI: 10.1007/978-1-0716-3774-6_13] [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: 03/02/2024]
Abstract
The recent progress in the development of in vivo biomarkers is rapidly changing how neurodegenerative diseases are conceptualized and diagnosed and how clinical trials are designed today. Alzheimer's disease (AD) - the most common neurodegenerative disorder - is characterized by a complex neuropathology involving the deposition of extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) of hyperphosphorylated tau proteins, accompanied by the activation of glial cells, i.e., astrocytes and microglia, and neuroinflammatory response, leading to neurodegeneration and cognitive dysfunction. An increasing diversity of positron emission tomography (PET) imaging radiotracers is available to selectively target the different pathophysiological processes of AD. Along with the success of Aβ PET and the more recent tau PET imaging, there is a great interest to develop PET tracers to image glial reactivity and neuroinflammation. While most research to date has focused on imaging microgliosis, there is an upsurge of interest in imaging reactive astrocytes in the AD continuum. There is increasing evidence that reactive astrocytes are morphologically and functionally heterogeneous, with different subtypes that express different markers and display various homeostatic or detrimental roles across disease stages. Therefore, multiple biomarkers are desirable to unravel the complex phenomenon of reactive astrocytosis. In the field of in vivo PET imaging in AD, the research concerning reactive astrocytes has predominantly focused on targeting monoamine oxidase B (MAO-B), most often using either 11C-deuterium-L-deprenyl (11C-DED) or 18F-SMBT-1 PET tracers. Additionally, imidazoline2 binding (I2BS) sites have been imaged using 11C-BU99008 PET. Recent studies in our group using 11C-DED PET imaging suggest that astrocytosis may be present from the early stages of disease development in AD. This chapter provides a detailed description of the practical approach used for the analysis of 11C-DED PET imaging data in a multitracer PET paradigm including 11C-Pittsburgh compound B (11C-PiB) and 18F-fluorodeoxyglucose (18F-FDG). The multitracer PET approach allows investigating the comparative regional and temporal patterns of in vivo brain astrocytosis, fibrillar Aβ deposition, glucose metabolism, and brain structural changes. It may also contribute to understanding the potential role of novel plasma biomarkers of reactive astrocytes, in particular the glial fibrillary acidic protein (GFAP), at different stages of disease progression. This chapter attempts to stimulate further research in the field, including the development of novel PET tracers that may allow visualizing different aspects of the complex astrocytic and microglial response in neurodegenerative diseases. Progress in the field will contribute to the incorporation of PET imaging of glial reactivity and neuroinflammation as biomarkers with clinical application and motivate further investigation on glial cells as therapeutic targets in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Mona-Lisa Malarte
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Ioannou
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Filipa M Rocha
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Chiotis
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
6
|
Bruno D, Gicas KM, Jauregi‐Zinkunegi A, Mueller KD, Lamar M. Delayed primacy recall performance predicts post mortem Alzheimer's disease pathology from unimpaired ante mortem cognitive baseline. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12524. [PMID: 38239330 PMCID: PMC10795090 DOI: 10.1002/dad2.12524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 01/22/2024]
Abstract
We propose a novel method to assess delayed primacy in the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) memory test. We then examine whether this measure predicts post mortem Alzheimer's disease (AD) neuropathology in individuals who were clinically unimpaired at baseline. A total of 1096 individuals were selected from the Rush Alzheimer's Disease Center database registry. All participants were clinically unimpaired at baseline, and had subsequently undergone brain autopsy. Average age at baseline was 78.8 (6.92). A Bayesian regression analysis was carried out with global pathology as an outcome; demographic, clinical, and apolipoprotein E (APOE) data as covariates; and cognitive predictors, including delayed primacy. Global AD pathology was best predicted by delayed primacy. Secondary analyses showed that delayed primacy was mostly associated with neuritic plaques, whereas total delayed recall was associated with neurofibrillary tangles. Sex differential associations were observed. We conclude that CERAD-derived delayed primacy is a useful metric for early detection and diagnosis of AD in unimpaired individuals. Highlights We propose a novel method to analyse serial position in the CERAD memory test.We analyse data from 1096 individuals who were cognitively unimpaired at baseline.Delayed primacy predicts post mortem pathology better than traditional metrics.
Collapse
Affiliation(s)
- Davide Bruno
- School of PsychologyLiverpool John Moores UniversityLiverpoolUK
| | | | | | - Kimberly D. Mueller
- Wisconsin Alzheimer's InstituteSchool of Medicine and Public HealthUniversity of Wisconsin – MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's Disease Research CenterSchool of Medicine and Public HealthUniversity of Wisconsin – MadisonMadisonWisconsinUSA
- Department of Communication Sciences and DisordersUniversity of Wisconsin – MadisonMadisonWisconsinUSA
| | - Melissa Lamar
- Rush Alzheimer's Disease Center and the Department of Psychiatry and Behavioral SciencesRush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
7
|
Kim SY, Park J, Choi H, Loeser M, Ryu H, Seo K. Digital Marker for Early Screening of Mild Cognitive Impairment Through Hand and Eye Movement Analysis in Virtual Reality Using Machine Learning: First Validation Study. J Med Internet Res 2023; 25:e48093. [PMID: 37862101 PMCID: PMC10625097 DOI: 10.2196/48093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/07/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND With the global rise in Alzheimer disease (AD), early screening for mild cognitive impairment (MCI), which is a preclinical stage of AD, is of paramount importance. Although biomarkers such as cerebrospinal fluid amyloid level and magnetic resonance imaging have been studied, they have limitations, such as high cost and invasiveness. Digital markers to assess cognitive impairment by analyzing behavioral data collected from digital devices in daily life can be a new alternative. In this context, we developed a "virtual kiosk test" for early screening of MCI by analyzing behavioral data collected when using a kiosk in a virtual environment. OBJECTIVE We aimed to investigate key behavioral features collected from a virtual kiosk test that could distinguish patients with MCI from healthy controls with high statistical significance. Also, we focused on developing a machine learning model capable of early screening of MCI based on these behavioral features. METHODS A total of 51 participants comprising 20 healthy controls and 31 patients with MCI were recruited by 2 neurologists from a university hospital. The participants performed a virtual kiosk test-developed by our group-where we recorded various behavioral data such as hand and eye movements. Based on these time series data, we computed the following 4 behavioral features: hand movement speed, proportion of fixation duration, time to completion, and the number of errors. To compare these behavioral features between healthy controls and patients with MCI, independent-samples 2-tailed t tests were used. Additionally, we used these behavioral features to train and validate a machine learning model for early screening of patients with MCI from healthy controls. RESULTS In the virtual kiosk test, all 4 behavioral features showed statistically significant differences between patients with MCI and healthy controls. Compared with healthy controls, patients with MCI had slower hand movement speed (t49=3.45; P=.004), lower proportion of fixation duration (t49=2.69; P=.04), longer time to completion (t49=-3.44; P=.004), and a greater number of errors (t49=-3.77; P=.001). All 4 features were then used to train a support vector machine to distinguish between healthy controls and patients with MCI. Our machine learning model achieved 93.3% accuracy, 100% sensitivity, 83.3% specificity, 90% precision, and 94.7% F1-score. CONCLUSIONS Our research preliminarily suggests that analyzing hand and eye movements in the virtual kiosk test holds potential as a digital marker for early screening of MCI. In contrast to conventional biomarkers, this digital marker in virtual reality is advantageous as it can collect ecologically valid data at an affordable cost and in a short period (5-15 minutes), making it a suitable means for early screening of MCI. We call for further studies to confirm the reliability and validity of this approach.
Collapse
Affiliation(s)
- Se Young Kim
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Jinseok Park
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hojin Choi
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Martin Loeser
- Department of Computer Science, Electrical Engineering and Mechatronics, ZHAW Zurich University of Applied Sciences, Winterthur, Switzerland
| | - Hokyoung Ryu
- Graduate School of Technology and Innovation Management, Hanyang University, Seoul, Republic of Korea
| | - Kyoungwon Seo
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
8
|
Bruno D, Gicas KM, Jauregi Zinkunegi A, Mueller KD, Lamar M. Delayed primacy recall performance predicts post mortem Alzheimer's disease pathology from unimpaired ante mortem cognitive baseline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546225. [PMID: 37425732 PMCID: PMC10327046 DOI: 10.1101/2023.06.26.546225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
INTRODUCTION We propose a novel method to assess delayed primacy in the CERAD memory test. We then examine whether this measure predicts post mortem Alzheimer's disease (AD) neuropathology in individuals who were clinically unimpaired at baseline. METHODS A total of 1096 individuals were selected from the Rush Alzheimer's Disease Center database registry. All participants were clinically unimpaired at baseline, and had subsequently undergone brain autopsy. Average age at baseline was 78.8 (6.92). A Bayesian regression analysis was carried out with global pathology as outcome; demographic, clinical and APOE data as covariates; and cognitive predictors, including delayed primacy. RESULTS Global AD pathology was best predicted by delayed primacy. Secondary analyses showed that delayed primacy was mostly associated with neuritic plaques, whereas total delayed recall was associated with neurofibrillary tangles. DISCUSSION We conclude that CERAD-derived delayed primacy is a useful metric for early detection and diagnosis of AD in unimpaired individuals.
Collapse
Affiliation(s)
- Davide Bruno
- School of Psychology, Liverpool John Moores University, UK
| | | | | | - Kimberly D. Mueller
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, USA
- Department of Communication Sciences and Disorders, University of Wisconsin – Madison, Madison, WI, USA
| | - Melissa Lamar
- Rush Alzheimer’s Disease Center and the Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
9
|
Luebke M, Parulekar M, Thomas FP. Fluid biomarkers for the diagnosis of neurodegenerative diseases. Biomark Neuropsychiatry 2023. [DOI: 10.1016/j.bionps.2023.100062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
|
10
|
Schäfer S, Mallick E, Schwed L, König A, Zhao J, Linz N, Bodin TH, Skoog J, Possemis N, ter Huurne D, Zettergren A, Kern S, Sacuiu S, Ramakers I, Skoog I, Tröger J. Screening for Mild Cognitive Impairment Using a Machine Learning Classifier and the Remote Speech Biomarker for Cognition: Evidence from Two Clinically Relevant Cohorts. J Alzheimers Dis 2023; 91:1165-1171. [PMID: 36565116 PMCID: PMC9912722 DOI: 10.3233/jad-220762] [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] [Indexed: 12/24/2022]
Abstract
BACKGROUND Modern prodromal Alzheimer's disease (AD) clinical trials might extend outreach to a general population, causing high screen-out rates and thereby increasing study time and costs. Thus, screening tools that cost-effectively detect mild cognitive impairment (MCI) at scale are needed. OBJECTIVE Develop a screening algorithm that can differentiate between healthy and MCI participants in different clinically relevant populations. METHODS Two screening algorithms based on the remote ki:e speech biomarker for cognition (ki:e SB-C) were designed on a Dutch memory clinic cohort (N = 121) and a Swedish birth cohort (N = 404). MCI classification was each evaluated on the training cohort as well as on the unrelated validation cohort. RESULTS The algorithms achieved a performance of AUC 0.73 and AUC 0.77 in the respective training cohorts and AUC 0.81 in the unseen validation cohorts. CONCLUSION The results indicate that a ki:e SB-C based algorithm robustly detects MCI across different cohorts and languages, which has the potential to make current trials more efficient and improve future primary health care.
Collapse
Affiliation(s)
- Simona Schäfer
- ki:elements, Saarbrücken, Germany,Correspondence to: Simona Schäfer, ki elements GmbH, Am Holzbrunnen 1a, 66121 Saarbrücken, Germany. Tel.: +49681 372009200; E-mail:
| | | | | | - Alexandra König
- ki:elements, Saarbrücken, Germany,Institut National de Recherche en Informatique et en Automatique (INRIA), Stars Team, Sophia Antipolis, Valbonne, France
| | | | | | - Timothy Hadarsson Bodin
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Skoog
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nina Possemis
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Daphne ter Huurne
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Anna Zettergren
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Silke Kern
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Simona Sacuiu
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Inez Ramakers
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Ingmar Skoog
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
11
|
Summers KL, Roseman G, Schilling KM, Dolgova NV, Pushie MJ, Sokaras D, Kroll T, Harris HH, Millhauser GL, Pickering IJ, George GN. Alzheimer's Drug PBT2 Interacts with the Amyloid β 1-42 Peptide Differently than Other 8-Hydroxyquinoline Chelating Drugs. Inorg Chem 2022; 61:14626-14640. [PMID: 36073854 PMCID: PMC9957665 DOI: 10.1021/acs.inorgchem.2c01694] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although Alzheimer's disease (AD) was first described over a century ago, it remains the leading cause of age-related dementia. Innumerable changes have been linked to the pathology of AD; however, there remains much discord regarding which might be the initial cause of the disease. The "amyloid cascade hypothesis" proposes that the amyloid β (Aβ) peptide is central to disease pathology, which is supported by elevated Aβ levels in the brain before the development of symptoms and correlations of amyloid burden with cognitive impairment. The "metals hypothesis" proposes a role for metal ions such as iron, copper, and zinc in the pathology of AD, which is supported by the accumulation of these metals within amyloid plaques in the brain. Metals have been shown to induce aggregation of Aβ, and metal ion chelators have been shown to reverse this reaction in vitro. 8-Hydroxyquinoline-based chelators showed early promise as anti-Alzheimer's drugs. Both 5-chloro-7-iodo-8-hydroxyquinoline (CQ) and 5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline (PBT2) underwent unsuccessful clinical trials for the treatment of AD. To gain insight into the mechanism of action of 8HQs, we have investigated the potential interaction of CQ, PBT2, and 5,7-dibromo-8-hydroxyquinoline (B2Q) with Cu(II)-bound Aβ(1-42) using X-ray absorption spectroscopy (XAS), high energy resolution fluorescence detected (HERFD) XAS, and electron paramagnetic resonance (EPR). By XAS, we found CQ and B2Q sequestered ∼83% of the Cu(II) from Aβ(1-42), whereas PBT2 sequestered only ∼59% of the Cu(II) from Aβ(1-42), suggesting that CQ and B2Q have a higher relative Cu(II) affinity than PBT2. From our EPR, it became clear that PBT2 sequestered Cu(II) from a heterogeneous mixture of Cu(II)Aβ(1-42) species in solution, leaving a single Cu(II)Aβ(1-42) species. It follows that the Cu(II) site in this Cu(II)Aβ(1-42) species is inaccessible to PBT2 and may be less solvent-exposed than in other Cu(II)Aβ(1-42) species. We found no evidence to suggest that these 8HQs form ternary complexes with Cu(II)Aβ(1-42).
Collapse
Affiliation(s)
- Kelly L. Summers
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham Roseman
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Kevin M. Schilling
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Natalia V. Dolgova
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - M. Jake Pushie
- Department of Surgery, University of Saskatchewan, 103 Hospital Dr, Saskatoon, Saskatchewan S7N 0W8, Canada
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Hugh H. Harris
- Department of Chemistry, University of Adelaide, South Australia 5005, Australia
| | - Glenn L. Millhauser
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Ingrid J. Pickering
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham N. George
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| |
Collapse
|
12
|
Pemberton HG, Collij LE, Heeman F, Bollack A, Shekari M, Salvadó G, Alves IL, Garcia DV, Battle M, Buckley C, Stephens AW, Bullich S, Garibotto V, Barkhof F, Gispert JD, Farrar G. Quantification of amyloid PET for future clinical use: a state-of-the-art review. Eur J Nucl Med Mol Imaging 2022; 49:3508-3528. [PMID: 35389071 PMCID: PMC9308604 DOI: 10.1007/s00259-022-05784-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Amyloid-β (Aβ) pathology is one of the earliest detectable brain changes in Alzheimer's disease (AD) pathogenesis. The overall load and spatial distribution of brain Aβ can be determined in vivo using positron emission tomography (PET), for which three fluorine-18 labelled radiotracers have been approved for clinical use. In clinical practice, trained readers will categorise scans as either Aβ positive or negative, based on visual inspection. Diagnostic decisions are often based on these reads and patient selection for clinical trials is increasingly guided by amyloid status. However, tracer deposition in the grey matter as a function of amyloid load is an inherently continuous process, which is not sufficiently appreciated through binary cut-offs alone. State-of-the-art methods for amyloid PET quantification can generate tracer-independent measures of Aβ burden. Recent research has shown the ability of these quantitative measures to highlight pathological changes at the earliest stages of the AD continuum and generate more sensitive thresholds, as well as improving diagnostic confidence around established binary cut-offs. With the recent FDA approval of aducanumab and more candidate drugs on the horizon, early identification of amyloid burden using quantitative measures is critical for enrolling appropriate subjects to help establish the optimal window for therapeutic intervention and secondary prevention. In addition, quantitative amyloid measurements are used for treatment response monitoring in clinical trials. In clinical settings, large multi-centre studies have shown that amyloid PET results change both diagnosis and patient management and that quantification can accurately predict rates of cognitive decline. Whether these changes in management reflect an improvement in clinical outcomes is yet to be determined and further validation work is required to establish the utility of quantification for supporting treatment endpoint decisions. In this state-of-the-art review, several tools and measures available for amyloid PET quantification are summarised and discussed. Use of these methods is growing both clinically and in the research domain. Concurrently, there is a duty of care to the wider dementia community to increase visibility and understanding of these methods.
Collapse
Affiliation(s)
- Hugh G Pemberton
- GE Healthcare, Amersham, UK.
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK.
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Lyduine E Collij
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Fiona Heeman
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ariane Bollack
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Isadora Lopes Alves
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Brain Research Center, Amsterdam, The Netherlands
| | - David Vallez Garcia
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mark Battle
- GE Healthcare, Amersham, UK
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | | | | | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
- NIMTLab, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Frederik Barkhof
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK
- UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | | |
Collapse
|
13
|
Proteomic Discovery and Validation of Novel Fluid Biomarkers for Improved Patient Selection and Prediction of Clinical Outcomes in Alzheimer’s Disease Patient Cohorts. Proteomes 2022; 10:proteomes10030026. [PMID: 35997438 PMCID: PMC9397030 DOI: 10.3390/proteomes10030026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/13/2022] [Accepted: 07/23/2022] [Indexed: 01/25/2023] Open
Abstract
Alzheimer’s disease (AD) is an irreversible neurodegenerative disease characterized by progressive cognitive decline. The two cardinal neuropathological hallmarks of AD include the buildup of cerebral β amyloid (Aβ) plaques and neurofibrillary tangles of hyperphosphorylated tau. The current disease-modifying treatments are still not effective enough to lower the rate of cognitive decline. There is an urgent need to identify early detection and disease progression biomarkers that can facilitate AD drug development. The current established readouts based on the expression levels of amyloid beta, tau, and phospho-tau have shown many discrepancies in patient samples when linked to disease progression. There is an urgent need to identify diagnostic and disease progression biomarkers from blood, cerebrospinal fluid (CSF), or other biofluids that can facilitate the early detection of the disease and provide pharmacodynamic readouts for new drugs being tested in clinical trials. Advances in proteomic approaches using state-of-the-art mass spectrometry are now being increasingly applied to study AD disease mechanisms and identify drug targets and novel disease biomarkers. In this report, we describe the application of quantitative proteomic approaches for understanding AD pathophysiology, summarize the current knowledge gained from proteomic investigations of AD, and discuss the development and validation of new predictive and diagnostic disease biomarkers.
Collapse
|
14
|
Varesi A, Carrara A, Pires VG, Floris V, Pierella E, Savioli G, Prasad S, Esposito C, Ricevuti G, Chirumbolo S, Pascale A. Blood-Based Biomarkers for Alzheimer's Disease Diagnosis and Progression: An Overview. Cells 2022; 11:1367. [PMID: 35456047 PMCID: PMC9044750 DOI: 10.3390/cells11081367] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disease characterized by amyloid-β (Aβ) plaque deposition and neurofibrillary tangle accumulation in the brain. Although several studies have been conducted to unravel the complex and interconnected pathophysiology of AD, clinical trial failure rates have been high, and no disease-modifying therapies are presently available. Fluid biomarker discovery for AD is a rapidly expanding field of research aimed at anticipating disease diagnosis and following disease progression over time. Currently, Aβ1-42, phosphorylated tau, and total tau levels in the cerebrospinal fluid are the best-studied fluid biomarkers for AD, but the need for novel, cheap, less-invasive, easily detectable, and more-accessible markers has recently led to the search for new blood-based molecules. However, despite considerable research activity, a comprehensive and up-to-date overview of the main blood-based biomarker candidates is still lacking. In this narrative review, we discuss the role of proteins, lipids, metabolites, oxidative-stress-related molecules, and cytokines as possible disease biomarkers. Furthermore, we highlight the potential of the emerging miRNAs and long non-coding RNAs (lncRNAs) as diagnostic tools, and we briefly present the role of vitamins and gut-microbiome-related molecules as novel candidates for AD detection and monitoring, thus offering new insights into the diagnosis and progression of this devastating disease.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Almo Collegio Borromeo, 27100 Pavia, Italy
| | - Adelaide Carrara
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Vitor Gomes Pires
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA;
| | - Valentina Floris
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (A.C.); (V.F.)
| | - Elisa Pierella
- School of Medicine, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Gabriele Savioli
- Emergency Department, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Sakshi Prasad
- Faculty of Medicine, National Pirogov Memorial Medical University, 21018 Vinnytsya, Ukraine;
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri, University of Pavia, 27100 Pavia, Italy;
| | - Giovanni Ricevuti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy;
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
15
|
Papp KV, Buckley RF, Jacobs HIL, Schultz AP, Properzi MJ, Vannini P, Hanseeuw BJ, Rentz DM, Johnson KA, Sperling RA. Association of Emerging β-Amyloid and Tau Pathology With Early Cognitive Changes in Clinically Normal Older Adults. Neurology 2022; 98:e1512-e1524. [PMID: 35338074 PMCID: PMC9012271 DOI: 10.1212/wnl.0000000000200137] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 01/14/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Alzheimer's disease (AD) clinical trials are moving earlier in the disease process, based on emerging signs of beta-amyloid (Aβ) and tau pathology. If early treatment is the right time for intervention, it is critical to find the right test to optimize cognitive outcome measures for clinical trials. We sought to identify cognitive measures associated with the earliest detectable signs of emerging Aβ and tau pathology. METHODS 112 clinically normal adults with longitudinal PIB-PET, FTP-PET and cognitive data for 7+ years were included from the Harvard Aging Brain Study (HABS). Analyses assessed those initially classified as PIB- (<Centiloid (CL) 20), then expanded to include PIB+ individuals up to CL40, the approximate threshold beyond which neocortical tau proliferation begins. Separate linear mixed effects models assessed the effects of emerging global Aβ (PIB slope) and tau (baseline FTP level and FTP slope) in the entorhinal (ERC) and inferior temporal (IT) cortices on multiple cognitive tasks and the Preclinical Alzheimer's Cognitive Composite (PACC) over time. RESULTS Steeper PIB slopes were associated with declining processing speed (DSST, Trails A) in those <CL20 and expanded to include learning/memory retrieval (FCSRT-FR, SRT-tr, LM-immed) in the <CL40 group. FTP had limited effects under CL20, with only rising right IT FTP slope related to declining FCSRT-FR and SRT-tr learning/memory retrieval (FCSRT-FR, SRT-tr). Expanding to include those initially <CL40, rising FTP level and/or slope were related to declines across all tasks, and PIB slope effects on memory retrieval but not DSST were reduced. A composite measure of processing speed and memory retrieval tasks provided the strongest prediction of decline under CL40, while PACC remained optimal at high levels of Aβ (>CL40). DISCUSSION Early, Aβ-mediated cognitive slowing was detected for processing speed measures, while early memory retrieval declines were associated with emerging Aβ and tau pathology. Composites of these measures may help determine whether anti-Aβ or anti-tau therapies administered at the first signs of pathology might preserve cognitive function. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that in clinically normal older adults, emerging PET-detected Alzheimer's disease pathology is associated with declining processing speeds and memory retrieval.
Collapse
Affiliation(s)
- Kathryn V Papp
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;3
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;3.,Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Australia
| | - Heidi I L Jacobs
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrizia Vannini
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;3
| | - Bernard J Hanseeuw
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;3
| | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;3.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA .,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA;3
| |
Collapse
|
16
|
Homann J, Osburg T, Ohlei O, Dobricic V, Deecke L, Bos I, Vandenberghe R, Gabel S, Scheltens P, Teunissen CE, Engelborghs S, Frisoni G, Blin O, Richardson JC, Bordet R, Lleó A, Alcolea D, Popp J, Clark C, Peyratout G, Martinez-Lage P, Tainta M, Dobson RJB, Legido-Quigley C, Sleegers K, Van Broeckhoven C, Wittig M, Franke A, Lill CM, Blennow K, Zetterberg H, Lovestone S, Streffer J, ten Kate M, Vos SJB, Barkhof F, Visser PJ, Bertram L. Genome-Wide Association Study of Alzheimer's Disease Brain Imaging Biomarkers and Neuropsychological Phenotypes in the European Medical Information Framework for Alzheimer's Disease Multimodal Biomarker Discovery Dataset. Front Aging Neurosci 2022; 14:840651. [PMID: 35386118 PMCID: PMC8979334 DOI: 10.3389/fnagi.2022.840651] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/15/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent neurodegenerative disease with an increasing prevalence in industrialized, aging populations. AD susceptibility has an established genetic basis which has been the focus of a large number of genome-wide association studies (GWAS) published over the last decade. Most of these GWAS used dichotomized clinical diagnostic status, i.e., case vs. control classification, as outcome phenotypes, without the use of biomarkers. An alternative and potentially more powerful study design is afforded by using quantitative AD-related phenotypes as GWAS outcome traits, an analysis paradigm that we followed in this work. Specifically, we utilized genotype and phenotype data from n = 931 individuals collected under the auspices of the European Medical Information Framework for Alzheimer's Disease Multimodal Biomarker Discovery (EMIF-AD MBD) study to perform a total of 19 separate GWAS analyses. As outcomes we used five magnetic resonance imaging (MRI) traits and seven cognitive performance traits. For the latter, longitudinal data from at least two timepoints were available in addition to cross-sectional assessments at baseline. Our GWAS analyses revealed several genome-wide significant associations for the neuropsychological performance measures, in particular those assayed longitudinally. Among the most noteworthy signals were associations in or near EHBP1 (EH domain binding protein 1; on chromosome 2p15) and CEP112 (centrosomal protein 112; 17q24.1) with delayed recall as well as SMOC2 (SPARC related modular calcium binding 2; 6p27) with immediate recall in a memory performance test. On the X chromosome, which is often excluded in other GWAS, we identified a genome-wide significant signal near IL1RAPL1 (interleukin 1 receptor accessory protein like 1; Xp21.3). While polygenic score (PGS) analyses showed the expected strong associations with SNPs highlighted in relevant previous GWAS on hippocampal volume and cognitive function, they did not show noteworthy associations with recent AD risk GWAS findings. In summary, our study highlights the power of using quantitative endophenotypes as outcome traits in AD-related GWAS analyses and nominates several new loci not previously implicated in cognitive decline.
Collapse
Affiliation(s)
- Jan Homann
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck, Germany
| | - Tim Osburg
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck, Germany
| | - Olena Ohlei
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck, Germany
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck, Germany
| | - Laura Deecke
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck, Germany
| | - Isabelle Bos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, Netherlands
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Neurology Service, University Hospital Leuven, Leuven, Belgium
| | - Silvy Gabel
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Center for Neurosciences, Universitair Ziekenhuis Brussel and Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Giovanni Frisoni
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Olivier Blin
- Institut Neurosciences Timone, AIX Marseille University, Marseille, France
| | - Jill C. Richardson
- Neurosciences Therapeutic Area, GlaxoSmithKline R&D, Stevenage, United Kingdom
| | - Regis Bordet
- Lille Neuroscience and Cognition, University of Lille, Inserm, CHU Lille, Lille, France
| | - Alberto Lleó
- Memory Unit, Neurology Department, Hospital de Sant Pau, Barcelona and Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Memory Unit, Neurology Department, Hospital de Sant Pau, Barcelona and Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Julius Popp
- Department of Geriatric Psychiatry, University Hospital of Psychiatry Zurich, Zurich, Switzerland
- Old Age Psychiatry, Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Christopher Clark
- Department of Geriatric Psychiatry, University Hospital of Psychiatry Zurich, Zurich, Switzerland
| | - Gwendoline Peyratout
- Old Age Psychiatry, Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Pablo Martinez-Lage
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, Donostia-San Sebastian, Spain
| | - Mikel Tainta
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, Donostia-San Sebastian, Spain
| | - Richard J. B. Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, United Kingdom
- Health Data Research UK London, University College London, London, United Kingdom
- Institute of Health Informatics, University College London, London, United Kingdom
- NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Cristina Legido-Quigley
- Steno Diabetes Center, Copenhagen, Denmark
- King’s College London, Institute of Pharmaceutical Sciences, London, United Kingdom
| | - Kristel Sleegers
- Complex Genetics of Alzheimer’s Disease Group, Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Michael Wittig
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Christina M. Lill
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck, Germany
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at University College London, London, United Kingdom
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Johannes Streffer
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Janssen R&D, LLC. Beerse, Belgium
| | - Mara ten Kate
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Stephanie J. B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, Netherlands
- Institutes of Neurology and Healthcare Engineering, University College London, London, United Kingdom
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, Netherlands
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Lübeck, Germany
- Department of Psychology, University of Oslo, Oslo, Norway
| |
Collapse
|
17
|
Nirmalraj PN, Schneider T, Felbecker A. Spatial organization of protein aggregates on red blood cells as physical biomarkers of Alzheimer's disease pathology. SCIENCE ADVANCES 2021; 7:eabj2137. [PMID: 34559561 PMCID: PMC8462905 DOI: 10.1126/sciadv.abj2137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/04/2021] [Indexed: 06/13/2023]
Abstract
Quantifying physical differences of protein aggregates implicated in Alzheimer’s disease (AD), in blood, could provide crucial information on disease stages. Here, red blood cells (RBCs) from 50 patients with neurocognitive complaints and 16 healthy individuals were profiled using an atomic force microscope (AFM). AFM measurements revealed patient age– and stage of neurocognitive disorder–dependent differences in size, shape, morphology, assembly, and prevalence of protein aggregates on RBCs, referred to as physical biomarkers. Crystals composed of fibrils were exclusively detected on RBCs for AD patients aged above 80 years. Fibril prevalence was negatively correlated with the cerebrospinal fluid (CSF) β-amyloid (Aβ) 42/40 ratio and was observed to be higher in the Aβ-positive patient category. Using a cutoff of ≥40% fibril prevalence, the CSF Aβ status was classified with 88% accuracy (sensitivity 100%, specificity 73%). The merits and challenges in integrating physical biomarkers in AD diagnosis are discussed.
Collapse
Affiliation(s)
- Peter Niraj Nirmalraj
- Transport at Nanoscale Interfaces Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf CH-8600, Switzerland
| | - Thomas Schneider
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen CH-9007, Switzerland
| | - Ansgar Felbecker
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen CH-9007, Switzerland
| |
Collapse
|
18
|
Pan L, Ou YN, Tan L, Tan L, Yu JT. Cerebrospinal fluid heart fatty acid‐binding protein as a predictive biomarker of neurodegeneration in Alzheimer’s disease. BRAIN SCIENCE ADVANCES 2021. [DOI: 10.26599/bsa.2021.9050003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Objective This study aims to investigate whether the heart fatty acid‐binding protein (HFABP) in the cerebrospinal fluid (CSF) was a potential predictive biomarker for Alzheimer’s disease (AD). Methods We evaluated the associations of CSF HFABP levels with core biomarkers, cognition, and brain structure in a sample population ( n = 302) from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database. Multiple linear regression and mixed‐effects models were employed in the analyses. AD progression was assessed using the Kaplan–Meier survival analysis. Results CSF HFABP was higher in patients with mild cognitive impairment and AD than the normal controls ( p < 0.001) and was particularly higher in those with amyloid‐β (Aβ) pathologic features. CSF HFABP was associated with higher baseline CSF t‐tau ( p < 0.001), CSF p‐tau ( p < 0.001), and CSF t‐tau/Aβ42 and CSF p‐tau/Aβ42 ( p < 0.01). Moreover, CSF HFABP was found to play predictive roles in hippocampal atrophy ( p < 0.01), cognitive decline ( p < 0.05), and the risk of AD ( p < 0.001). Conclusion Our findings suggest that CSF HFABP can be a predictive biomarker of AD.
Collapse
Affiliation(s)
- Lu Pan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Lin Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian 116044, Liaoning, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| |
Collapse
|
19
|
Cullen NC, Leuzy A, Janelidze S, Palmqvist S, Svenningsson AL, Stomrud E, Dage JL, Mattsson-Carlgren N, Hansson O. Plasma biomarkers of Alzheimer's disease improve prediction of cognitive decline in cognitively unimpaired elderly populations. Nat Commun 2021; 12:3555. [PMID: 34117234 PMCID: PMC8196018 DOI: 10.1038/s41467-021-23746-0] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Plasma biomarkers of amyloid, tau, and neurodegeneration (ATN) need to be characterized in cognitively unimpaired (CU) elderly individuals. We therefore tested if plasma measurements of amyloid-β (Aβ)42/40, phospho-tau217 (P-tau217), and neurofilament light (NfL) together predict clinical deterioration in 435 CU individuals followed for an average of 4.8 ± 1.7 years in the BioFINDER study. A combination of all three plasma biomarkers and basic demographics best predicted change in cognition (Pre-Alzheimer's Clinical Composite; R2 = 0.14, 95% CI [0.12-0.17]; P < 0.0001) and subsequent AD dementia (AUC = 0.82, 95% CI [0.77-0.91], P < 0.0001). In a simulated clinical trial, a screening algorithm combining all three plasma biomarkers would reduce the required sample size by 70% (95% CI [54-81]; P < 0.001) with cognition as trial endpoint, and by 63% (95% CI [53-70], P < 0.001) with subsequent AD dementia as trial endpoint. Plasma ATN biomarkers show usefulness in cognitively unimpaired populations and could make large clinical trials more feasible and cost-effective.
Collapse
Affiliation(s)
| | - Antoine Leuzy
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | | | - Sebastian Palmqvist
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Anna L Svenningsson
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | | | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Department of Neurology, Skåne University Hospital, Lund, Sweden.
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden.
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
20
|
Schwarz AJ. The Use, Standardization, and Interpretation of Brain Imaging Data in Clinical Trials of Neurodegenerative Disorders. Neurotherapeutics 2021; 18:686-708. [PMID: 33846962 PMCID: PMC8423963 DOI: 10.1007/s13311-021-01027-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Imaging biomarkers play a wide-ranging role in clinical trials for neurological disorders. This includes selecting the appropriate trial participants, establishing target engagement and mechanism-related pharmacodynamic effect, monitoring safety, and providing evidence of disease modification. In the early stages of clinical drug development, evidence of target engagement and/or downstream pharmacodynamic effect-especially with a clear relationship to dose-can provide confidence that the therapeutic candidate should be advanced to larger and more expensive trials, and can inform the selection of the dose(s) to be further tested, i.e., to "de-risk" the drug development program. In these later-phase trials, evidence that the therapeutic candidate is altering disease-related biomarkers can provide important evidence that the clinical benefit of the compound (if observed) is grounded in meaningful biological changes. The interpretation of disease-related imaging markers, and comparability across different trials and imaging tools, is greatly improved when standardized outcome measures are defined. This standardization should not impinge on scientific advances in the imaging tools per se but provides a common language in which the results generated by these tools are expressed. PET markers of pathological protein aggregates and structural imaging of brain atrophy are common disease-related elements across many neurological disorders. However, PET tracers for pathologies beyond amyloid β and tau are needed, and the interpretability of structural imaging can be enhanced by some simple considerations to guard against the possible confound of pseudo-atrophy. Learnings from much-studied conditions such as Alzheimer's disease and multiple sclerosis will be beneficial as the field embraces rarer diseases.
Collapse
Affiliation(s)
- Adam J Schwarz
- Takeda Pharmaceuticals Ltd., 40 Landsdowne Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
21
|
Hansson O, Batrla R, Brix B, Carrillo MC, Corradini V, Edelmayer RM, Esquivel RN, Hall C, Lawson J, Bastard NL, Molinuevo JL, Nisenbaum LK, Rutz S, Salamone SJ, Teunissen CE, Traynham C, Umek RM, Vanderstichele H, Vandijck M, Wahl S, Weber CJ, Zetterberg H, Blennow K. The Alzheimer's Association international guidelines for handling of cerebrospinal fluid for routine clinical measurements of amyloid β and tau. Alzheimers Dement 2021; 17:1575-1582. [PMID: 33788410 DOI: 10.1002/alz.12316] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/29/2021] [Indexed: 01/01/2023]
Abstract
The core cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers amyloid beta (Aβ42 and Aβ40), total tau, and phosphorylated tau, have been extensively clinically validated, with very high diagnostic performance for AD, including the early phases of the disease. However, between-center differences in pre-analytical procedures may contribute to variability in measurements across laboratories. To resolve this issue, a workgroup was led by the Alzheimer's Association with experts from both academia and industry. The aim of the group was to develop a simplified and standardized pre-analytical protocol for CSF collection and handling before analysis for routine clinical use, and ultimately to ensure high diagnostic performance and minimize patient misclassification rates. Widespread application of the protocol would help minimize variability in measurements, which would facilitate the implementation of unified cut-off levels across laboratories, and foster the use of CSF biomarkers in AD diagnostics for the benefit of the patients.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | | | | | | | | | | | | | - John Lawson
- Fujirebio Diagnostics Inc, Malvern, Pennsylvania, USA
| | | | - José Luis Molinuevo
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation Barcelona, Barcelona, Spain.,AD and Other Cognitive Disorders Unit Hospital Clinic, Barcelona, Spain
| | | | | | | | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | - Simone Wahl
- Saladax Biomedical, Inc. Bethlehem, Bethlehem, Pennsylvania, USA
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
22
|
Yang J, Jia L, Li Y, Qiu Q, Quan M, Jia J. Fluid Biomarkers in Clinical Trials for Alzheimer's Disease: Current and Future Application. J Alzheimers Dis 2021; 81:19-32. [PMID: 33749646 DOI: 10.3233/jad-201068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Alzheimer's disease (AD) research is entering a unique moment in which enormous information about the molecular basis of this disease is being translated into therapeutics. However, almost all drug candidates have failed in clinical trials over the past 30 years. These many trial failures have highlighted a need for the incorporation of biomarkers in clinical trials to help improve the trial design. Fluid biomarkers measured in cerebrospinal fluid and circulating blood, which can reflect the pathophysiological process in the brain, are becoming increasingly important in AD clinical trials. In this review, we first succinctly outline a panel of fluid biomarkers for neuropathological changes in AD. Then, we provide a comprehensive overview of current and future application of fluid biomarkers in clinical trials for AD. We also summarize the many challenges that have been encountered in efforts to integrate fluid biomarkers in clinical trials, and the barriers that have begun to be overcome. Ongoing research efforts in the field of fluid biomarkers will be critical to make significant progress in ultimately unveiling disease-modifying therapies in AD.
Collapse
Affiliation(s)
- Jianwei Yang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, People's Republic of China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Qiongqiong Qiu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,National Clinical Research Center for Geriatric Diseases, Beijing, People's Republic of China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, People's Republic of China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| |
Collapse
|
23
|
Meikle SR, Sossi V, Roncali E, Cherry SR, Banati R, Mankoff D, Jones T, James M, Sutcliffe J, Ouyang J, Petibon Y, Ma C, El Fakhri G, Surti S, Karp JS, Badawi RD, Yamaya T, Akamatsu G, Schramm G, Rezaei A, Nuyts J, Fulton R, Kyme A, Lois C, Sari H, Price J, Boellaard R, Jeraj R, Bailey DL, Eslick E, Willowson KP, Dutta J. Quantitative PET in the 2020s: a roadmap. Phys Med Biol 2021; 66:06RM01. [PMID: 33339012 PMCID: PMC9358699 DOI: 10.1088/1361-6560/abd4f7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Positron emission tomography (PET) plays an increasingly important role in research and clinical applications, catalysed by remarkable technical advances and a growing appreciation of the need for reliable, sensitive biomarkers of human function in health and disease. Over the last 30 years, a large amount of the physics and engineering effort in PET has been motivated by the dominant clinical application during that period, oncology. This has led to important developments such as PET/CT, whole-body PET, 3D PET, accelerated statistical image reconstruction, and time-of-flight PET. Despite impressive improvements in image quality as a result of these advances, the emphasis on static, semi-quantitative 'hot spot' imaging for oncologic applications has meant that the capability of PET to quantify biologically relevant parameters based on tracer kinetics has not been fully exploited. More recent advances, such as PET/MR and total-body PET, have opened up the ability to address a vast range of new research questions, from which a future expansion of applications and radiotracers appears highly likely. Many of these new applications and tracers will, at least initially, require quantitative analyses that more fully exploit the exquisite sensitivity of PET and the tracer principle on which it is based. It is also expected that they will require more sophisticated quantitative analysis methods than those that are currently available. At the same time, artificial intelligence is revolutionizing data analysis and impacting the relationship between the statistical quality of the acquired data and the information we can extract from the data. In this roadmap, leaders of the key sub-disciplines of the field identify the challenges and opportunities to be addressed over the next ten years that will enable PET to realise its full quantitative potential, initially in research laboratories and, ultimately, in clinical practice.
Collapse
Affiliation(s)
- Steven R Meikle
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Canada
| | - Emilie Roncali
- Department of Biomedical Engineering, University of California, Davis, United States of America
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Richard Banati
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
- Australian Nuclear Science and Technology Organisation, Sydney, Australia
| | - David Mankoff
- Department of Radiology, University of Pennsylvania, United States of America
| | - Terry Jones
- Department of Radiology, University of California, Davis, United States of America
| | - Michelle James
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), CA, United States of America
- Department of Neurology and Neurological Sciences, Stanford University, CA, United States of America
| | - Julie Sutcliffe
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Internal Medicine, University of California, Davis, CA, United States of America
| | - Jinsong Ouyang
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Yoann Petibon
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Chao Ma
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Suleman Surti
- Department of Radiology, University of Pennsylvania, United States of America
| | - Joel S Karp
- Department of Radiology, University of Pennsylvania, United States of America
| | - Ramsey D Badawi
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Taiga Yamaya
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Go Akamatsu
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Georg Schramm
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Ahmadreza Rezaei
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Johan Nuyts
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Roger Fulton
- Brain and Mind Centre, The University of Sydney, Australia
- Department of Medical Physics, Westmead Hospital, Sydney, Australia
| | - André Kyme
- Brain and Mind Centre, The University of Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Australia
| | - Cristina Lois
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Hasan Sari
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Julie Price
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, location VUMC, Netherlands
| | - Robert Jeraj
- Departments of Medical Physics, Human Oncology and Radiology, University of Wisconsin, United States of America
- Faculty of Mathematics and Physics, University of Ljubljana, Slovenia
| | - Dale L Bailey
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Enid Eslick
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Kathy P Willowson
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Joyita Dutta
- Department of Electrical and Computer Engineering, University of Massachusetts Lowell, United States of America
| |
Collapse
|
24
|
Friedman LG, McKeehan N, Hara Y, Cummings JL, Matthews DC, Zhu J, Mohs RC, Wang D, Hendrix SB, Quintana M, Schneider LS, Grundman M, Dickson SP, Feldman HH, Jaeger J, Finger EC, Ryan JM, Niehoff D, Dickinson SLJ, Markowitz JT, Owen M, Travaglia A, Fillit HM. Value-Generating Exploratory Trials in Neurodegenerative Dementias. Neurology 2021; 96:944-954. [PMID: 33674360 PMCID: PMC8205472 DOI: 10.1212/wnl.0000000000011774] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/12/2021] [Indexed: 11/25/2022] Open
Abstract
Drug development for Alzheimer disease and other neurodegenerative dementias, including frontotemporal dementia, has experienced a long history of phase 2 and phase 3 clinical trials that failed to show efficacy of investigational drugs. Despite differences in clinical and behavioral characteristics, these disorders have shared pathologies and face common challenges in designing early-phase trials that are predictive of late-stage success. Here, we discuss exploratory clinical trials in neurodegenerative dementias. These are generally phase 1b or phase 2a trials that are designed to assess pharmacologic effects and rely on biomarker outcomes, with shorter treatment durations and fewer patients than traditional phase 2 studies. Exploratory trials can establish go/no-go decision points, support proof of concept and dose selection, and terminate drugs that fail to show target engagement with suitable exposure and acceptable safety profiles. Early failure saves valuable resources including opportunity costs. This is especially important for programs in academia and small biotechnology companies but may be applied to high-risk projects in large pharmaceutical companies to achieve proof of concept more rapidly at lower costs than traditional approaches. Exploratory studies in a staged clinical development program may provide promising data to warrant the substantial resources needed to advance compounds through late-stage development. To optimize the design and application of exploratory trials, the Alzheimer's Drug Discovery Foundation and the Association for Frontotemporal Degeneration convened an advisory panel to provide recommendations on outcome measures and statistical considerations for these types of studies and study designs that can improve efficiency in clinical development.
Collapse
Affiliation(s)
- Lauren G Friedman
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Nicholas McKeehan
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Yuko Hara
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Jeffrey L Cummings
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Dawn C Matthews
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Jian Zhu
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Richard C Mohs
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Deli Wang
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Suzanne B Hendrix
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Melanie Quintana
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Lon S Schneider
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Michael Grundman
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Samuel P Dickson
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Howard H Feldman
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Judith Jaeger
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Elizabeth C Finger
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - J Michael Ryan
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Debra Niehoff
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Susan L-J Dickinson
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Jessica T Markowitz
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Meriel Owen
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Alessio Travaglia
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA
| | - Howard M Fillit
- From the Alzheimer's Drug Discovery Foundation (L.G.F., N.M., Y.H., M.O., A.T., H.M.F.), New York; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; ADM Diagnostics (D.C.M.), Inc. Northbrook, IL; Servier Pharmaceuticals (J.Z.), Boston, MA; Global Alzheimer's Platform Foundation (R.C.M.), Washington, DC; AgeneBio (R.C.M.), Inc. Baltimore, MD; AbbVie Inc. (D.W.), North Chicago, IL; Pentara Corporation (S.B.H., S.P.D.), Salt Lake City, UT; Berry Consultants (M.Q.), Austin TX; Keck School of Medicine of the University of Southern California (L.S.S.), Los Angeles; Global R&D Partners (M.G.), LLC, University of California, San Diego, La Jolla; Department of Neurosciences (H.H.F.), University of California, San Diego, La Jolla; Albert Einstein College of Medicine (J.J.), Bronx, NY; CognitionMetrics (J.J.), LLC; Department of Clinical Neurological Sciences and Robarts Research Institute (E.C.F.), Schulich School of Medicine and Dentistry, University of Western Ontario; Parkwood Institute (E.C.F.), Lawson Health Research Institute, St. Josephs Health Care, London, Ontario, Canada; Rodin Therapeutics (J.M.R.), Boston, MA; Association for Frontotemporal Degeneration (D.N., S.L.-J.D.), Radnor, PA; and Modus Outcomes LLP (J.T.M.), Cambridge, MA.
| |
Collapse
|
25
|
Tarawneh R. Biomarkers: Our Path Towards a Cure for Alzheimer Disease. Biomark Insights 2020; 15:1177271920976367. [PMID: 33293784 PMCID: PMC7705771 DOI: 10.1177/1177271920976367] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Over the last decade, biomarkers have significantly improved our understanding of
the pathophysiology of Alzheimer disease (AD) and provided valuable tools to
examine different disease mechanisms and their progression over time. While
several markers of amyloid, tau, neuronal, synaptic, and axonal injury,
inflammation, and immune dysregulation in AD have been identified, there is a
relative paucity of biomarkers which reflect other disease mechanisms such as
oxidative stress, mitochondrial injury, vascular or endothelial injury, and
calcium-mediated excitotoxicity. Importantly, there is an urgent need to
standardize methods for biomarker assessments across different centers, and to
identify dynamic biomarkers which can monitor disease progression over time
and/or response to potential disease-modifying treatments. The updated research
framework for AD, proposed by the National Institute of Aging- Alzheimer’s
Association (NIA-AA) Work Group, emphasizes the importance of incorporating
biomarkers in AD research and defines AD as a biological construct consisting of
amyloid, tau, and neurodegeneration which spans pre-symptomatic and symptomatic
stages. As results of clinical trials of AD therapeutics have been
disappointing, it has become increasingly clear that the success of future AD
trials will require the incorporation of biomarkers in participant selection,
prognostication, monitoring disease progression, and assessing response to
treatments. We here review the current state of fluid AD biomarkers, and discuss
the advantages and limitations of the updated NIA-AA research framework.
Importantly, the integration of biomarker data with clinical, cognitive, and
imaging domains through a systems biology approach will be essential to
adequately capture the molecular, genetic, and pathological heterogeneity of AD
and its spatiotemporal evolution over time.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
26
|
Hong S, Prokopenko D, Dobricic V, Kilpert F, Bos I, Vos SJB, Tijms BM, Andreasson U, Blennow K, Vandenberghe R, Cleynen I, Gabel S, Schaeverbeke J, Scheltens P, Teunissen CE, Niemantsverdriet E, Engelborghs S, Frisoni G, Blin O, Richardson JC, Bordet R, Molinuevo JL, Rami L, Kettunen P, Wallin A, Lleó A, Sala I, Popp J, Peyratout G, Martinez-Lage P, Tainta M, Dobson RJB, Legido-Quigley C, Sleegers K, Van Broeckhoven C, Ten Kate M, Barkhof F, Zetterberg H, Lovestone S, Streffer J, Wittig M, Franke A, Tanzi RE, Visser PJ, Bertram L. Genome-wide association study of Alzheimer's disease CSF biomarkers in the EMIF-AD Multimodal Biomarker Discovery dataset. Transl Psychiatry 2020; 10:403. [PMID: 33223526 PMCID: PMC7680793 DOI: 10.1038/s41398-020-01074-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/23/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and the most common form of dementia in the elderly. Susceptibility to AD is considerably determined by genetic factors which hitherto were primarily identified using case-control designs. Elucidating the genetic architecture of additional AD-related phenotypic traits, ideally those linked to the underlying disease process, holds great promise in gaining deeper insights into the genetic basis of AD and in developing better clinical prediction models. To this end, we generated genome-wide single-nucleotide polymorphism (SNP) genotyping data in 931 participants of the European Medical Information Framework Alzheimer's Disease Multimodal Biomarker Discovery (EMIF-AD MBD) sample to search for novel genetic determinants of AD biomarker variability. Specifically, we performed genome-wide association study (GWAS) analyses on 16 traits, including 14 measures derived from quantifications of five separate amyloid-beta (Aβ) and tau-protein species in the cerebrospinal fluid (CSF). In addition to confirming the well-established effects of apolipoprotein E (APOE) on diagnostic outcome and phenotypes related to Aβ42, we detected novel potential signals in the zinc finger homeobox 3 (ZFHX3) for CSF-Aβ38 and CSF-Aβ40 levels, and confirmed the previously described sex-specific association between SNPs in geminin coiled-coil domain containing (GMNC) and CSF-tau. Utilizing the results from independent case-control AD GWAS to construct polygenic risk scores (PRS) revealed that AD risk variants only explain a small fraction of CSF biomarker variability. In conclusion, our study represents a detailed first account of GWAS analyses on CSF-Aβ and -tau-related traits in the EMIF-AD MBD dataset. In subsequent work, we will utilize the genomics data generated here in GWAS of other AD-relevant clinical outcomes ascertained in this unique dataset.
Collapse
Affiliation(s)
- Shengjun Hong
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Dmitry Prokopenko
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Fabian Kilpert
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Isabelle Bos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands
| | - Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands
| | - Ulf Andreasson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Neurology Service, University Hospital Leuven, Leuven, Belgium
| | - Isabelle Cleynen
- Laboratory for Complex Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Silvy Gabel
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Jolien Schaeverbeke
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ellis Niemantsverdriet
- Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Center for Neurosciences, UZ Brussel and Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Giovanni Frisoni
- University of Geneva, Geneva, Switzerland
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Olivier Blin
- AIX Marseille University, INS, Ap-hm, Marseille, France
| | | | - Regis Bordet
- University of Lille, Inserm, CHU Lille, Lille, France
| | - José Luis Molinuevo
- Alzheimer's disease and other cognitive disorders unit, Hospital Clinic I Universitari, Barcelona, Spain
| | - Lorena Rami
- Alzheimer's disease and other cognitive disorders unit, Hospital Clinic I Universitari, Barcelona, Spain
| | - Petronella Kettunen
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neuropathology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alberto Lleó
- Memory Unit, Neurology Department, Hospital de Sant Pau, Barcelona and Centro de Investigación Biomédica en Red en enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Isabel Sala
- Memory Unit, Neurology Department, Hospital de Sant Pau, Barcelona and Centro de Investigación Biomédica en Red en enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Julius Popp
- Geriatric Psychiatry, Department of Mental Health and Psychiatry, Geneva University Hospitals, Geneva, Switzerland
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Gwendoline Peyratout
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Pablo Martinez-Lage
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, San Sebastian, Spain
| | - Mikel Tainta
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, San Sebastian, Spain
| | - Richard J B Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
- Health Data Research UK London, University College London, 222 Euston Road, London, UK
- Institute of Health Informatics, University College London, 222 Euston Road, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, 222 Euston Road, London, UK
| | - Cristina Legido-Quigley
- Steno Diabetes Center, Copenhagen, Denmark
- Institute of Pharmaceutical Sciences, King's College London, London, UK
| | - Kristel Sleegers
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Mara Ten Kate
- Alzheimer Center and Department of Neurology, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Institutes of Neurology and Healthcare Engineering, University College London, London, UK
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | | | - Johannes Streffer
- Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Translational Medicine Neuroscience, UCB Biopharma SPRL, Braine l'Alleud, Belgium
| | - Michael Wittig
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Rudolph E Tanzi
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, The Netherlands
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany.
- Department of Psychology, University of Oslo, Oslo, Norway.
| |
Collapse
|
27
|
Shi WY, Wang ZT, Sun FR, Ma YH, Xu W, Shen XN, Dong Q, Tan L, Yu JT, Yu Y. High pulse pressure is a risk factor for prodromal Alzheimer's disease: a longitudinal study. Aging (Albany NY) 2020; 12:18221-18237. [PMID: 32960784 PMCID: PMC7585106 DOI: 10.18632/aging.103678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023]
Abstract
It has been increasingly evident that pulse pressure (PP) is associated with Alzheimer's disease (AD) but whether PP increases AD risk and the mechanism responsible for this association remains unclear. To investigate the effects of PP in the process of AD, we have evaluated the cross-sectional and longitudinal associations of PP with AD biomarkers, brain structure and cognition and have assessed the effect of PP on AD risk in a large sample (n= 1,375) from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Multiple linear regression and mixed-model regression were employed in cross-sectional and longitudinal analyses respectively. Clinical disease progression was assessed using Cox proportional hazards models. High PP was associated with lower β-amyloid 42 (Aβ42) (P= .015), and higher total tau (T-tau) (P= .011), phosphorylated tau (P-tau) (P= .003), T-tau/Aβ42 (P= .004) and P-tau/Aβ42 (P = .001), as well as heavier cortical amyloid-beta burden (P= .011). Longitudinally, baseline high PP was significantly associated with hippocampal atrophy (P= .039), entorhinal atrophy (P= .031) and worse memory performance (P= .058). Baseline high PP showed more rapid progression than those with normal PP (P <.001). These results suggest PP elevation could increase AD risk, which may be driven by amyloid plaques and subclinical neurodegeneration.
Collapse
Affiliation(s)
- Wen-Yan Shi
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Zuo-Teng Wang
- College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Fu-Rong Sun
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China,College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China,Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Yu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | | |
Collapse
|
28
|
Canevelli M, Remoli G, Bacigalupo I, Valletta M, Toccaceli Blasi M, Sciancalepore F, Bruno G, Cesari M, Vanacore N. Use of Biomarkers in Ongoing Research Protocols on Alzheimer's Disease. J Pers Med 2020; 10:jpm10030068. [PMID: 32722106 PMCID: PMC7564515 DOI: 10.3390/jpm10030068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
The present study aimed to describe and discuss the state of the art of biomarker use in ongoing Alzheimer’s disease (AD) research. A review of 222 ongoing phase 1, 2, 3, and 4 protocols registered in the clinicaltrials.gov database was performed. All the trials (i) enrolling subjects with clinical disturbances and/or preclinical diagnoses falling within the AD continuum; and (ii) testing the efficacy and/or safety/tolerability of a therapeutic intervention, were analyzed. The use of biomarkers of amyloid deposition, tau pathology, and neurodegeneration among the eligibility criteria and/or study outcomes was assessed. Overall, 58.2% of ongoing interventional studies on AD adopt candidate biomarkers. They are mostly adopted by studies at the preliminary stages of the drug development process to explore the safety profile of novel therapies, and to provide evidence of target engagement and disease-modifying properties. The biologically supported selection of participants is mostly based on biomarkers of amyloid deposition, whereas the use of biomarkers as study outcomes mostly relies on markers of neurodegeneration. Biomarkers play an important role in the design and conduction of research protocols targeting AD. Nevertheless, their clinical validity, utility, and cost-effectiveness in the “real world” remain to be clarified.
Collapse
Affiliation(s)
- Marco Canevelli
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, 00161 Rome, Italy; (I.B.); (N.V.)
- Correspondence: ; Tel./Fax: +39-(06)-4991-4604
| | - Giulia Remoli
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Ilaria Bacigalupo
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, 00161 Rome, Italy; (I.B.); (N.V.)
| | - Martina Valletta
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Marco Toccaceli Blasi
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Francesco Sciancalepore
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Giuseppe Bruno
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Nicola Vanacore
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, 00161 Rome, Italy; (I.B.); (N.V.)
| |
Collapse
|
29
|
Villa C, Lavitrano M, Salvatore E, Combi R. Molecular and Imaging Biomarkers in Alzheimer's Disease: A Focus on Recent Insights. J Pers Med 2020; 10:jpm10030061. [PMID: 32664352 PMCID: PMC7565667 DOI: 10.3390/jpm10030061] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/28/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease among the elderly, affecting millions of people worldwide and clinically characterized by a progressive and irreversible cognitive decline. The rapid increase in the incidence of AD highlights the need for an easy, efficient and accurate diagnosis of the disease in its initial stages in order to halt or delay the progression. The currently used diagnostic methods rely on measures of amyloid-β (Aβ), phosphorylated (p-tau) and total tau (t-tau) protein levels in the cerebrospinal fluid (CSF) aided by advanced neuroimaging techniques like positron emission tomography (PET) and magnetic resonance imaging (MRI). However, the invasiveness of these procedures and the high cost restrict their utilization. Hence, biomarkers from biological fluids obtained using non-invasive methods and novel neuroimaging approaches provide an attractive alternative for the early diagnosis of AD. Such biomarkers may also be helpful for better understanding of the molecular mechanisms underlying the disease, allowing differential diagnosis or at least prolonging the pre-symptomatic stage in patients suffering from AD. Herein, we discuss the advantages and limits of the conventional biomarkers as well as recent promising candidates from alternative body fluids and new imaging techniques.
Collapse
Affiliation(s)
- Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Correspondence: (C.V.); (R.C.)
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Institute for the Experimental Endocrinology and Oncology, National Research Council (IEOS-CNR), 80131 Naples, Italy;
| | - Elena Salvatore
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II University, 80131 Naples, Italy;
| | - Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Correspondence: (C.V.); (R.C.)
| |
Collapse
|
30
|
Stylianaki I, Polizopoulou ZS, Theodoridis A, Koutouzidou G, Baka R, Papaioannou NG. Amyloid-beta plasma and cerebrospinal fluid biomarkers in aged dogs with cognitive dysfunction syndrome. J Vet Intern Med 2020; 34:1532-1540. [PMID: 32557873 PMCID: PMC7379053 DOI: 10.1111/jvim.15812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 01/03/2023] Open
Abstract
Background Cognitive dysfunction syndrome (CDS) is a common progressive neurodegenerative disease that is poorly defined. Specific multitargeted protocols do not exist for setting the diagnosis and the prognosis of the syndrome. Hypothesis/Objectives To quantify Aβ42 and Aβ40 peptides in blood and cerebrospinal fluid (CSF) and to investigate their contribution to CCDS. Animals A total of 61 dogs from a hospital population. Methods Case‐control study. Six young (YG: 0‐4 years old), 8 middle‐aged (4‐8 years old), 17 cognitively unimpaired and aged (CU: 8‐20 years old), and 30 cognitively impaired and aged (CI: 8‐17 years). From the CI group, 10 dogs exhibited mild impairment (CI‐MCI) and 20 exhibited severe impairment (CI‐SCI). Cognitive status was assessed using a validated owner‐based questionnaire. Direct and indirect Aβ markers were determined in plasma fractions (total‐TP, free‐FP, bound to plasma components‐CP) and CSF using commercial ELISA assays (AΒtest, Araclon Biotech). Results TPAβ42/40 facilitated discrimination between CI‐MCI and CU aged dogs with area under curve ≥ 0.79. CSFAβ42 levels were higher (P = .09) in CU (1.25 ± 0.28 ng/mL) than in MCI (1.04 ± 0.32 ng/mL) dogs. CSF Aβ42 levels were correlated with the CP fragment (CPAβ40: P = .02, CPAβ42: P = .02). CPAβ42 was higher in the CI‐MCI (23.03 ± 11.79 pg/μL) group compared to the other aged dogs (CU: 10.42 ± 7.18 pg/μL, P = .02, SCI: 11.40 ± 12.98 pg/μL, P = .26). Conclusion and Clinical Importance The Aβ should be determined in all of the 3 plasma fractions (TP, FP, CP). In the clinical approach, TPAβ42/40 could be used as an efficient preselection tool for the aged canine population targeting dogs with mild cognitive impairment.
Collapse
Affiliation(s)
- Ioanna Stylianaki
- Department of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Zoe S Polizopoulou
- Diagnostic Laboratory, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandros Theodoridis
- Laboratory of Animal Production Economics, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Koutouzidou
- Department of Applied Informatics, University of Macedonia, Thessaloniki, Greece
| | - Rania Baka
- Diagnostic Laboratory, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos G Papaioannou
- Department of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
31
|
SWATH-MS analysis of cerebrospinal fluid to generate a robust battery of biomarkers for Alzheimer's disease. Sci Rep 2020; 10:7423. [PMID: 32366888 PMCID: PMC7198522 DOI: 10.1038/s41598-020-64461-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 04/16/2020] [Indexed: 12/14/2022] Open
Abstract
Cerebrospinal fluid (CSF) Aβ42 and tau protein levels are established diagnostic biomarkers of Alzheimer's disease (AD). However, their inadequacy to represent clinical efficacy in drug trials indicates the need for new biomarkers. Sequential window acquisition of all theoretical fragment ion spectra (SWATH)-based mass spectrometry (MS) is an advanced proteomic tool for large-scale, high-quality quantification. In this study, SWATH-MS showed that VGF, chromogranin-A, secretogranin-1, and opioid-binding protein/cell adhesion molecule were significantly decreased in 42 AD patients compared to 39 controls, whereas 14-3-3ζ was increased (FDR < 0.05). In addition, 16 other proteins showed substantial changes (FDR < 0.2). The expressions of the top 21 analytes were closely interconnected, but were poorly correlated with CSF Aβ42, tTau, and pTau181 levels. Logistic regression analysis and data mining were used to establish the best algorithm for AD, which created novel biomarker panels with high diagnostic value (AUC = 0.889 and 0.924) and a strong correlation with clinical severity (all p < 0.001). Targeted proteomics was used to validate their usefulness in a different cohort (n = 36) that included patients with other brain disorders (all p < 0.05). This study provides a list of proteins (and combinations thereof) that could serve as new AD biomarkers.
Collapse
|
32
|
Insel PS, Donohue MC, Sperling R, Hansson O, Mattsson-Carlgren N. The A4 study: β-amyloid and cognition in 4432 cognitively unimpaired adults. Ann Clin Transl Neurol 2020; 7:776-785. [PMID: 32315118 PMCID: PMC7261742 DOI: 10.1002/acn3.51048] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/30/2022] Open
Abstract
Objective To clarify the preclinical stage of Alzheimer’s disease by estimating when β‐amyloid accumulation first becomes associated with changes in cognition. Methods Here we studied a large group (N = 4432) of cognitively unimpaired individuals who were screened for inclusion in the A4 trial (age 65–85) to assess the effect of subthreshold levels of β‐amyloid on cognition and to identify which cognitive domains first become affected. Results β‐amyloid accumulation was linked to significant cognitive dysfunction in cognitively unimpaired participants with subthreshold levels of β‐amyloid in multiple measures of memory (Logical Memory Delayed Recall, P = 0.03; Free and Cued Selective Reminding Test, P < 0.001), the Preclinical Alzheimer’s Cognitive Composite (P = 0.01), and was marginally associated with decreased executive function (Digit Symbol Substitution, P = 0.07). Significantly, decreased cognitive scores were associated with suprathreshold levels of β‐amyloid, across all measures (P < 0.05). The Free and Cued Selective Reminding Test, a list recall memory test, appeared most sensitive to β‐amyloid ‐related decreases in average cognitive scores, outperforming all other cognitive domains, including the narrative recall memory test, Logical Memory. Interpretation Clinical trials for cognitively unimpaired β‐amyloid‐positive individuals will include a large number of individuals where mechanisms downstream from β‐amyloid pathology are already activated. These findings have implications for primary and secondary prevention of Alzheimer’s disease.
Collapse
Affiliation(s)
- Philip S Insel
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Psychiatry, University of California, San Francisco, California
| | - Michael C Donohue
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, California
| | - Reisa Sperling
- Department of Neurology, Harvard Aging Brain Study, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
33
|
Lance EI, Barron-Casella E, Everett AD, Casella JF. Brain-derived neurotrophic factor levels in pediatric sickle cell disease. Pediatr Blood Cancer 2020; 67:e28076. [PMID: 31736231 PMCID: PMC7171877 DOI: 10.1002/pbc.28076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Children with sickle cell disease (SCD) have an increased risk of neurological complications, particularly stroke and silent cerebral infarction (SCI). Brain-derived neurotrophic factor (BDNF) is a nerve growth factor associated with neuronal survival, synaptic plasticity, elevated transcranial Doppler (TCD) velocities and increased risk of stroke in patients with SCD. The objective of this study was to analyze plasma BDNF protein levels in children with SCD participating in the Silent Cerebral Infarct Transfusion Multi-Center Clinical Trial (SIT Trial), comparing plasma samples of children with SCD and SCI to plasma samples from children with SCD without SCI, as well as healthy pediatric control participants. PROCEDURE Entry, exit, and longitudinal blood samples were collected from 190 SIT Trial participants with SCD and healthy pediatric controls over time. BDNF levels were measured by enzyme-linked immunosorbent assay. Sample collection was not optimized for measurements of BDNF, but factors affecting BDNF levels were accounted for in analyses. RESULTS BDNF levels were significantly higher in children with SCD in comparison to healthy pediatric control subjects. BDNF levels significantly increased over time in SCD participants. BDNF levels did not show any significant associations with the presence or absence of SCI or new/progressive SCI/stroke or TCD velocities. CONCLUSIONS Plasma BDNF levels are elevated and increase over time in children with SCD. Additional studies with more longitudinal samples are needed to address the reasons for those increased levels.
Collapse
Affiliation(s)
- Eboni I. Lance
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute,Department of Neurology, the Johns Hopkins University School of Medicine
| | - Emily Barron-Casella
- Department of Pediatrics, Division of Hematology, the Johns Hopkins University School of Medicine
| | - Allen D. Everett
- Department of Pediatrics, Division of Cardiology, the Johns Hopkins University School of Medicine
| | - James F. Casella
- Department of Pediatrics, Division of Hematology, the Johns Hopkins University School of Medicine
| |
Collapse
|
34
|
Ou YN, Hu H, Wang ZT, Xu W, Tan L, Yu JT. Plasma neurofilament light as a longitudinal biomarker of neurodegeneration in Alzheimer’s disease. BRAIN SCIENCE ADVANCES 2020. [DOI: 10.1177/2096595820902582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: To examine whether plasma neurofilament light (NFL) might be a potential longitudinal biomarker for Alzheimer’s disease (AD). Methods: A total of 835 individuals from the Alzheimer’s Disease Neuroimaging Initiative were involved. Correlations of the rate of change in plasma NFL with cerebrospinal fluid biomarkers, cognition, and brain structure were investigated. Cox proportional hazards models were used to assess the associations between quartiles of plasma NFL and the risk of AD conversion. Results: Participants were further divided into β amyloid-positive (Aβ+) versus β amyloid-negative (Aβ−), resulting in five biomarker group combinations, which are CN Aβ−, CN Aβ+, MCI Aβ−, MCI Aβ+ and AD Aβ+. Plasma NFL concentration markedly increased in the five groups longitudinally ( p < 0.001) with the greatest rate of change in AD Aβ+ group. The rate of change in plasma NFL was associated with cognitive deficits and neuroimaging hallmarks of AD over time ( p < 0.005). Compared with the bottom quartile, the top quartile of change rate was associated with a 5.41-fold increased risk of AD (95% CI = 1.83−16.01) in the multivariate model. Conclusion: Our finding implies the potential of plasma NFL as a longitudinal noninvasive biomarker in AD.
Collapse
Affiliation(s)
- Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Zuo-Teng Wang
- College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266100, Shandong, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, Shandong, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200433, China
| | | |
Collapse
|
35
|
James HJ, Van Houtven CH, Lippmann S, Burke JR, Shepherd-Banigan M, Belanger E, Wetle TF, Plassman BL. How Accurately Do Patients and Their Care Partners Report Results of Amyloid-β PET Scans for Alzheimer's Disease Assessment? J Alzheimers Dis 2020; 74:625-636. [PMID: 32065790 PMCID: PMC7183243 DOI: 10.3233/jad-190922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Amyloid-β PET scans will likely become an integral part of the diagnostic evaluation for Alzheimer's disease if Medicare approves reimbursement for the scans. However, little is known about patients' and their care partners' interpretation of scan results. OBJECTIVE This study seeks to understand how accurately patients with mild cognitive impairment (MCI) or dementia and their care partners report results of amyloid-β PET scans and factors related to correct reporting. METHODS A mixed-methods approach was used to analyze survey data from 1,845 patient-care partner dyads and responses to open-ended questions about interpretation of scan results from a sub-sample of 200 dyads. RESULTS Eighty-three percent of patients and 85% of care partners correctly reported amyloid-β PET scan results. Patients' higher cognitive function was associated with a small but significant decrease in the predicted probability of not only patients accurately reporting scan results (ME: -0.004, 95% CI: -0.007, -0.000), but also care partners accurately reporting scan results (ME: -0.006, 95% CI: -0.007, -0.001), as well as decreased concordance between patient and care partner reports (ME: -0.004, 95% CI: -0.007, -0.001). Content analysis of open-ended responses found that participants who reported the scan results incorrectly exhibited more confusion about diagnostic terminology than those who correctly reported the scan results. CONCLUSION Overall, patients with MCI or dementia showed high rates of accurate reporting of amyloid-β PET scan results. However, responses to questions about the meaning of the scan results highlight the need for improved provider communication, including providing written explanations and better prognostic information.
Collapse
Affiliation(s)
- Hailey J. James
- Department of Health Policy and Management, University of North Carolina, Chapel Hill, NC, USA
- Department of Population Health Sciences, Duke University, Durham, NC, USA
| | - Courtney Harold Van Houtven
- Department of Population Health Sciences, Duke University, Durham, NC, USA
- Health Services Research and Development in Primary Care, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Steven Lippmann
- Department of Population Health Sciences, Duke University, Durham, NC, USA
| | - James R. Burke
- Department of Neurology, School of Medicine, Duke University, Durham, NC, USA
| | - Megan Shepherd-Banigan
- Department of Population Health Sciences, Duke University, Durham, NC, USA
- Health Services Research and Development in Primary Care, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Emmanuelle Belanger
- Center for Gerontology and Healthcare Research, School of Public Health, Brown University, Providence, RI, USA
- Department of Health Services, Policy and Practice, School of Public Health, Brown University, Providence, RI, USA
| | - Terrie Fox Wetle
- Department of Health Services, Policy and Practice, School of Public Health, Brown University, Providence, RI, USA
| | - Brenda L. Plassman
- Department of Neurology, School of Medicine, Duke University, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
36
|
Llibre-Guerra JJ, Li Y, Schindler SE, Gordon BA, Fagan AM, Morris JC, Benzinger TLS, Hassenstab J, Wang G, Allegri R, Berman SB, Chhatwal J, Farlow MR, Holtzman DM, Jucker M, Levin J, Noble JM, Salloway S, Schofield P, Karch C, Fox NC, Xiong C, Bateman RJ, McDade E. Association of Longitudinal Changes in Cerebrospinal Fluid Total Tau and Phosphorylated Tau 181 and Brain Atrophy With Disease Progression in Patients With Alzheimer Disease. JAMA Netw Open 2019; 2:e1917126. [PMID: 31825500 PMCID: PMC6991202 DOI: 10.1001/jamanetworkopen.2019.17126] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IMPORTANCE The amyloid/tau/neurodegeneration (A/T/N) framework uses cerebrospinal fluid (CSF) levels of total tau (tTau) as a marker of neurodegeneration and CSF levels of phosphorylated tau 181 (pTau181) as a marker of tau tangles. However, it is unclear whether CSF levels of tTau and pTau181 have similar or different trajectories over the course of Alzheimer disease. OBJECTIVES To examine the rates of change in CSF levels of tTau and pTau181 across the Alzheimer disease course and how the rates of change are associated with brain atrophy as measured by magnetic resonance imaging. DESIGN, SETTING, AND PARTICIPANTS This cohort study was set in tertiary research clinics. Each participant was a member of a pedigree with a known mutation for dominantly inherited Alzheimer disease. Participants were divided into 3 groups on the basis of the presence of a mutation and their Clinical Dementia Rating score. Data analysis was performed in June 2019. MAIN OUTCOMES AND MEASURES Rates of change of CSF tTau and pTau181 levels and their association with the rate of change of brain volume. RESULTS Data from 465 participants (283 mutation carriers and 182 noncarriers) were analyzed. The mean (SD) age of the cohort was 37.8 (11.3) years, and 262 (56.3%) were women. The mean (SD) follow-up duration was 2.7 (1.5) years. Two or more longitudinal CSF and magnetic resonance imaging assessments were available for 160 and 247 participants, respectively. Sixty-five percent of mutation carriers (183) did not have symptoms at baseline (Clinical Dementia Rating score, 0). For mutation carriers, the annual rates of change for CSF tTau and pTau181 became significantly different from 0 approximately 10 years before the estimated year of onset (mean [SE] rates of change, 5.5 [2.8] for tTau [P = .05] and 0.7 [0.3] for pTau 181 [P = .04]) and 15 years before onset (mean [SE] rates of change, 5.4 [3.9] for tTau [P = .17] and 1.1 [0.5] for pTau181 [P = .03]), respectively. The rate of change of pTau181 was positive and increased at the early stages of the disease, showing a positive rate of change starting at 15 estimated years before onset until 5 estimated years before onset (mean [SE], 0.4 [0.3]), followed by a positive but decreasing rate of change at year 0 (mean [SE], 0.1 [0.3]) and then negative rates of change at 5 years (mean [SE], -0.3 [0.4]) and 10 years (mean [SE], -0.6 [0.6]) after symptom onset. In individuals without symptoms (Clinical Dementia Rating score, 0), the rates of change of CSF tTau and pTau181 were negatively associated with brain atrophy (high rates of change in CSF measures were associated with low rates of change in brain volume in asymptomatic stages). After symptom onset (Clinical Dementia Rating score, >0), an increased rate of brain atrophy was not associated with rates of change of levels of both CSF tTau and pTau181. CONCLUSIONS AND RELEVANCE These findings suggest that CSF tTau and pTau181 may have different associations with brain atrophy across the disease time course. These results have implications for understanding the dynamics of disease pathobiology and interpreting neuronal injury biomarker concentrations in response to Alzheimer disease progression and disease-modifying therapies.
Collapse
Affiliation(s)
| | - Yan Li
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | | | - Brian A. Gordon
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
| | - Anne M. Fagan
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - John C. Morris
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Hope Center for Neurological Disorders, St Louis, Missouri
- Knight Alzheimer’s Disease Research Center, St Louis, Missouri
| | | | - Jason Hassenstab
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Guoqiao Wang
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Tübingen, Germany
| | - Ricardo Allegri
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Sarah B. Berman
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | - David M. Holtzman
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Hope Center for Neurological Disorders, St Louis, Missouri
- Knight Alzheimer’s Disease Research Center, St Louis, Missouri
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Tübingen, Germany
- DZNE-German Center for Neurodegenerative Diseases, Tübingen, Tübingen, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
- DZNE-German Center for Neurodegenerative Diseases, Munich, Munich, Germany
- SyNergy, Munich Cluster for Systems Neurology, Munich, Germany
| | - James M. Noble
- Taub Institute for Research on Alzheimer’s Disease, Aging Brain G.H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, New York
| | - Stephen Salloway
- Memory & Aging Program, Butler Hospital, Brown University, Providence, Rhode Island
| | - Peter Schofield
- Neuroscience Research Australia, Randwick, Sydney, New South Wales, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Celeste Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Nick C. Fox
- Dementia Research Centre, University College London, London, United Kingdom
| | - Chengjie Xiong
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | - Randall J. Bateman
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| |
Collapse
|
37
|
Abstract
Currently established and employed biomarkers of Alzheimer's disease (AD) predominantly mirror AD-associated molecular and structural brain changes. While they are necessary for identifying disease-specific neuropathology, they lack a clear and robust relationship with the clinical presentation of dementia; they can be altered in healthy individuals, while they often inadequately mirror the degree of cognitive and functional deficits in affected subjects. There is growing evidence that synaptic loss and dysfunction are early events during the trajectory of AD pathogenesis that best correlate with the clinical symptoms, suggesting measures of brain functional deficits as candidate early markers of AD. Resting-state electroencephalography (EEG) is a widely available and noninvasive diagnostic method that provides direct insight into brain synaptic activity in real time. Quantitative EEG (qEEG) analysis additionally provides information on physiologically meaningful frequency components, dynamic alterations and topography of EEG signal generators, i.e. neuronal signaling. Numerous studies have shown that qEEG measures can detect disruptions in activity, topographical distribution and synchronization of neuronal (synaptic) activity such as generalized EEG slowing, reduced global synchronization and anteriorization of neuronal generators of fast-frequency resting-state EEG activity in patients along the AD continuum. Moreover, qEEG measures appear to correlate well with surrogate markers of AD neuropathology and discriminate between different types of dementia, making them promising low-cost and noninvasive markers of AD. Future large-scale longitudinal clinical studies are needed to elucidate the diagnostic and prognostic potential of qEEG measures as early functional markers of AD on an individual subject level.
Collapse
Affiliation(s)
- Una Smailovic
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.
| | - Vesna Jelic
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Clinic for Cognitive Disorders, Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
38
|
Schindler SE, Bollinger JG, Ovod V, Mawuenyega KG, Li Y, Gordon BA, Holtzman DM, Morris JC, Benzinger TLS, Xiong C, Fagan AM, Bateman RJ. High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology 2019; 93:e1647-e1659. [PMID: 31371569 PMCID: PMC6946467 DOI: 10.1212/wnl.0000000000008081] [Citation(s) in RCA: 533] [Impact Index Per Article: 88.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/17/2019] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE We examined whether plasma β-amyloid (Aβ)42/Aβ40, as measured by a high-precision assay, accurately diagnosed brain amyloidosis using amyloid PET or CSF p-tau181/Aβ42 as reference standards. METHODS Using an immunoprecipitation and liquid chromatography-mass spectrometry assay, we measured Aβ42/Aβ40 in plasma and CSF samples from 158 mostly cognitively normal individuals that were collected within 18 months of an amyloid PET scan. RESULTS Plasma Aβ42/Aβ40 had a high correspondence with amyloid PET status (receiver operating characteristic area under the curve [AUC] 0.88, 95% confidence interval [CI] 0.82-0.93) and CSF p-tau181/Aβ42 (AUC 0.85, 95% CI 0.79-0.92). The combination of plasma Aβ42/Aβ40, age, and APOE ε4 status had a very high correspondence with amyloid PET (AUC 0.94, 95% CI 0.90-0.97). Individuals with a negative amyloid PET scan at baseline and a positive plasma Aβ42/Aβ40 (<0.1218) had a 15-fold greater risk of conversion to amyloid PET-positive compared to individuals with a negative plasma Aβ42/Aβ40 (p = 0.01). CONCLUSIONS Plasma Aβ42/Aβ40, especially when combined with age and APOE ε4 status, accurately diagnoses brain amyloidosis and can be used to screen cognitively normal individuals for brain amyloidosis. Individuals with a negative amyloid PET scan and positive plasma Aβ42/Aβ40 are at increased risk for converting to amyloid PET-positive. Plasma Aβ42/Aβ40 could be used in prevention trials to screen for individuals likely to be amyloid PET-positive and at risk for Alzheimer disease dementia. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that plasma Aβ42/Aβ40 levels accurately determine amyloid PET status in cognitively normal research participants.
Collapse
Affiliation(s)
- Suzanne E Schindler
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - James G Bollinger
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - Vitaliy Ovod
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - Kwasi G Mawuenyega
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - Yan Li
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - Brian A Gordon
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - David M Holtzman
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - John C Morris
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - Tammie L S Benzinger
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - Chengjie Xiong
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - Anne M Fagan
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO
| | - Randall J Bateman
- From the Department of Neurology (S.E.S., J.G.B., V.O., K.G.M., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Knight Alzheimer's Disease Research Center (S.E.S., B.A.G., D.M.H., J.C.M., T.L.S.B., C.X., A.M.F., R.J.B.), Division of Biostatistics (Y.L., C.X.), Mallinckrodt Institute of Radiology (B.A.G., T.L.S.B.), and Hope Center for Neurological Disorders (D.M.H., A.M.F., R.J.B.), Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
39
|
Silajdžić E, Björkqvist M. A Critical Evaluation of Wet Biomarkers for Huntington's Disease: Current Status and Ways Forward. J Huntingtons Dis 2019; 7:109-135. [PMID: 29614689 PMCID: PMC6004896 DOI: 10.3233/jhd-170273] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
There is an unmet clinical need for objective biomarkers to monitor disease progression and treatment response in Huntington's disease (HD). The aim of this review is, therefore, to provide practical advice for biomarker discovery and to summarise studies on biofluid markers for HD. A PubMed search was performed to review literature with regard to candidate saliva, urine, blood and cerebrospinal fluid biomarkers for HD. Information has been organised into tables to allow a pragmatic approach to the discussion of the evidence and generation of practical recommendations for future studies. Many of the markers published converge on metabolic and inflammatory pathways, although changes in other analytes representing antioxidant and growth factor pathways have also been found. The most promising markers reflect neuronal and glial degeneration, particularly neurofilament light chain. International collaboration to standardise assays and study protocols, as well as to recruit sufficiently large cohorts, will facilitate future biomarker discovery and development.
Collapse
Affiliation(s)
- Edina Silajdžić
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Maria Björkqvist
- Department of Experimental Medical Science, Brain Disease Biomarker Unit, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
40
|
Palmqvist S, Janelidze S, Stomrud E, Zetterberg H, Karl J, Zink K, Bittner T, Mattsson N, Eichenlaub U, Blennow K, Hansson O. Performance of Fully Automated Plasma Assays as Screening Tests for Alzheimer Disease-Related β-Amyloid Status. JAMA Neurol 2019; 76:1060-1069. [PMID: 31233127 PMCID: PMC6593637 DOI: 10.1001/jamaneurol.2019.1632] [Citation(s) in RCA: 289] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Importance Accurate blood-based biomarkers for Alzheimer disease (AD) might improve the diagnostic accuracy in primary care, referrals to memory clinics, and screenings for AD trials. Objective To examine the accuracy of plasma β-amyloid (Aβ) and tau measured using fully automated assays together with other blood-based biomarkers to detect cerebral Aβ. Design, Setting, and Participants Two prospective, cross-sectional, multicenter studies. Study participants were consecutively enrolled between July 6, 2009, and February 11, 2015 (cohort 1), and between January 29, 2000, and October 11, 2006 (cohort 2). Data were analyzed in 2018. The first cohort comprised 842 participants (513 cognitively unimpaired [CU], 265 with mild cognitive impairment [MCI], and 64 with AD dementia) from the Swedish BioFINDER study. The validation cohort comprised 237 participants (34 CU, 109 MCI, and 94 AD dementia) from a German biomarker study. Main Outcome and Measures The cerebrospinal fluid (CSF) Aβ42/Aβ40 ratio was used as the reference standard for brain Aβ status. Plasma Aβ42, Aβ40 and tau were measured using Elecsys immunoassays (Roche Diagnostics) and examined as predictors of Aβ status in logistic regression models in cohort 1 and replicated in cohort 2. Plasma neurofilament light chain (NFL) and heavy chain (NFH) and APOE genotype were also examined in cohort 1. Results The mean (SD) age of the 842 participants in cohort 1 was 72 (5.6) years, with a range of 59 to 88 years, and 446 (52.5%) were female. For the 237 in cohort 2, mean (SD) age was 66 (10) years with a range of 23 to 85 years, and 120 (50.6%) were female. In cohort 1, plasma Aβ42 and Aβ40 predicted Aβ status with an area under the receiver operating characteristic curve (AUC) of 0.80 (95% CI, 0.77-0.83). When adding APOE, the AUC increased significantly to 0.85 (95% CI, 0.82-0.88). Slight improvements were seen when adding plasma tau (AUC, 0.86; 95% CI, 0.83-0.88) or tau and NFL (AUC, 0.87; 95% CI, 0.84-0.89) to Aβ42, Aβ40 and APOE. The results were similar in CU and cognitively impaired participants, and in younger and older participants. Applying the plasma Aβ42 and Aβ40 model from cohort 1 in cohort 2 resulted in slightly higher AUC (0.86; 95% CI, 0.81-0.91), but plasma tau did not contribute. Using plasma Aβ42, Aβ40, and APOE in an AD trial screening scenario reduced positron emission tomography costs up to 30% to 50% depending on cutoff. Conclusions and Relevance Plasma Aβ42 and Aβ40 measured using Elecsys immunoassays predict Aβ status in all stages of AD with similar accuracy in a validation cohort. Their accuracy can be further increased by analyzing APOE genotype. Potential future applications of these blood tests include prescreening of Aβ positivity in clinical AD trials to lower the costs and number of positron emission tomography scans or lumbar punctures.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom.,UK Dementia Research Institute at UCL, London, United Kingdom
| | | | | | - Tobias Bittner
- Genentech, a Member of the Roche Group, Basel, Switzerland
| | - Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Malmö, Sweden
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
41
|
Prospects and challenges of imaging neuroinflammation beyond TSPO in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2019; 46:2831-2847. [PMID: 31396666 PMCID: PMC6879435 DOI: 10.1007/s00259-019-04462-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation, as defined by the activation of microglia and astrocytes, has emerged in the last years as a key element of the pathogenesis of neurodegenerative diseases based on genetic findings and preclinical and human studies. This has raised the need for new methodologies to assess and follow glial activation in patients, prompting the development of PET ligands for molecular imaging of glial cells and novel structural MRI and DTI tools leading to a multimodal approach. The present review describes the recent advancements in microglia and astrocyte biology in the context of health, ageing, and Alzheimer's disease, the most common dementia worldwide. The review further delves in molecular imaging discussing the challenges associated with past and present targets, including conflicting findings, and finally, presenting novel methodologies currently explored to improve our in vivo knowledge of the neuroinflammatory patterns in Alzheimer's disease. With glial cell activation as a potential therapeutic target in neurodegenerative diseases, the translational research between cell biologists, chemists, physicists, radiologists, and neurologists should be strengthened.
Collapse
|
42
|
Identifying an Optimal Cutoff of the Montreal Cognitive Assessment to Predict Amyloid-PET Positivity in a Referral Memory Clinic. Alzheimer Dis Assoc Disord 2019; 33:194-199. [PMID: 31305321 DOI: 10.1097/wad.0000000000000330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Brain amyloid- positron emission tomography (PET) imaging is highly sensitive for identifying Alzheimer disease. Currently, there is a lack of insight on the association between amyloid-PET status and the widely used Montreal cognitive assessment (MoCA). Studying this relationship may optimize the clinical use of amyloid-PET imaging. OBJECTIVES To evaluate the relationship between amyloid-PET status and MoCA scores and to identify a MoCA score cutoff that translates to amyloid-PET positivity. METHODS Using retrospective chart review, patients from 2010 to 2017 with amyloid-PET scans (positive or negative) and MoCA test scores were included. We studied the relationship between amyloid-PET status and MoCA scores and the influence of age, sex, education, and race. A MoCA score cutoff for amyloid-PET positivity was estimated. RESULTS Among the 684 clinic patients with dementia, 99 fulfilled inclusion criteria. Amyloid-PET positivity was associated significantly with lower MoCA scores (median=19, U=847, P=0.01). The MoCA score cutoff (25) used for minimal cognitive impairment (MCI) predicted amyloid-PET positivity suboptimally (sensitivity=94.6%, specificity=13.9%). A MoCA score cutoff of 20 patients had optimal sensitivity (64.2%) and specificity (67.4%). CONCLUSIONS Amyloid-PET positivity is associated with lower MoCA scores. Clinical utility of amyloid-PET scan is likely to be suboptimal at the MoCA score cutoff for minimal cognitive impairment.
Collapse
|
43
|
Ou YN, Hu H, Wang ZT, Xu W, Tan L, Yu JT. Plasma neurofilament light as a longitudinal biomarker of neurodegeneration in Alzheimer’s disease. BRAIN SCIENCE ADVANCES 2019. [DOI: 10.26599/bsa.2019.9050011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
44
|
Bereczki E, Branca RM, Francis PT, Pereira JB, Baek JH, Hortobágyi T, Winblad B, Ballard C, Lehtiö J, Aarsland D. Synaptic markers of cognitive decline in neurodegenerative diseases: a proteomic approach. Brain 2019; 141:582-595. [PMID: 29324989 DOI: 10.1093/brain/awx352] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 10/30/2017] [Indexed: 01/12/2023] Open
Abstract
See Attems and Jellinger (doi:10.1093/brain/awx360) for a scientific commentary on this article.Cognitive changes occurring throughout the pathogenesis of neurodegenerative diseases are directly linked to synaptic loss. We used in-depth proteomics to compare 32 post-mortem human brains in the prefrontal cortex of prospectively followed patients with Alzheimer's disease, Parkinson's disease with dementia, dementia with Lewy bodies and older adults without dementia. In total, we identified 10 325 proteins, 851 of which were synaptic proteins. Levels of 25 synaptic proteins were significantly altered in the various dementia groups. Significant loss of SNAP47, GAP43, SYBU (syntabulin), LRFN2, SV2C, SYT2 (synaptotagmin 2), GRIA3 and GRIA4 were further validated on a larger cohort comprised of 92 brain samples using ELISA or western blot. Cognitive impairment before death and rate of cognitive decline significantly correlated with loss of SNAP47, SYBU, LRFN2, SV2C and GRIA3 proteins. Besides differentiating Parkinson's disease dementia, dementia with Lewy bodies, and Alzheimer's disease from controls with high sensitivity and specificity, synaptic proteins also reliably discriminated Parkinson's disease dementia from Alzheimer's disease patients. Our results suggest that these particular synaptic proteins have an important predictive and discriminative molecular fingerprint in neurodegenerative diseases and could be a potential target for early disease intervention.
Collapse
Affiliation(s)
- Erika Bereczki
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Novum, Stockholm, Sweden
| | - Rui M Branca
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Paul T Francis
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, UK
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Novum, 14186 Stockholm, Sweden
| | - Jean-Ha Baek
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Novum, Stockholm, Sweden
| | - Tibor Hortobágyi
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, University of Debrecen, Debrecen, Hungary.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Novum, Stockholm, Sweden
| | - Clive Ballard
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Janne Lehtiö
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Novum, Stockholm, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
45
|
Rabinovici GD, Gatsonis C, Apgar C, Chaudhary K, Gareen I, Hanna L, Hendrix J, Hillner BE, Olson C, Lesman-Segev OH, Romanoff J, Siegel BA, Whitmer RA, Carrillo MC. Association of Amyloid Positron Emission Tomography With Subsequent Change in Clinical Management Among Medicare Beneficiaries With Mild Cognitive Impairment or Dementia. JAMA 2019; 321:1286-1294. [PMID: 30938796 PMCID: PMC6450276 DOI: 10.1001/jama.2019.2000] [Citation(s) in RCA: 388] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
IMPORTANCE Amyloid positron emission tomography (PET) detects amyloid plaques in the brain, a core neuropathological feature of Alzheimer disease. OBJECTIVE To determine if amyloid PET is associated with subsequent changes in the management of patients with mild cognitive impairment (MCI) or dementia of uncertain etiology. DESIGN, SETTING, AND PARTICIPANTS The Imaging Dementia-Evidence for Amyloid Scanning (IDEAS) study was a single-group, multisite longitudinal study that assessed the association between amyloid PET and subsequent changes in clinical management for Medicare beneficiaries with MCI or dementia. Participants were required to meet published appropriate use criteria stating that etiology of cognitive impairment was unknown, Alzheimer disease was a diagnostic consideration, and knowledge of PET results was expected to change diagnosis and management. A total of 946 dementia specialists at 595 US sites enrolled 16 008 patients between February 2016 and September 2017. Patients were followed up through January 2018. Dementia specialists documented their diagnosis and management plan before PET and again 90 (±30) days after PET. EXPOSURES Participants underwent amyloid PET at 343 imaging centers. MAIN OUTCOMES AND MEASURES The primary end point was change in management between the pre- and post-PET visits, as assessed by a composite outcome that included Alzheimer disease drug therapy, other drug therapy, and counseling about safety and future planning. The study was powered to detect a 30% or greater change in the MCI and dementia groups. One of 2 secondary end points is reported: the proportion of changes in diagnosis (from Alzheimer disease to non-Alzheimer disease and vice versa) between pre- and post-PET visits. RESULTS Among 16 008 registered participants, 11 409 (71.3%) completed study procedures and were included in the analysis (median age, 75 years [interquartile range, 71-80]; 50.9% women; 60.5% with MCI). Amyloid PET results were positive in 3817 patients with MCI (55.3%) and 3154 patients with dementia (70.1%). The composite end point changed in 4159 of 6905 patients with MCI (60.2% [95% CI, 59.1%-61.4%]) and 2859 of 4504 patients with dementia (63.5% [95% CI, 62.1%-64.9%]), significantly exceeding the 30% threshold in each group (P < .001, 1-sided). The etiologic diagnosis changed from Alzheimer disease to non-Alzheimer disease in 2860 of 11 409 patients (25.1% [95% CI, 24.3%-25.9%]) and from non-Alzheimer disease to Alzheimer disease in 1201 of 11 409 (10.5% [95% CI, 10.0%-11.1%]). CONCLUSIONS AND RELEVANCE Among Medicare beneficiaries with MCI or dementia of uncertain etiology evaluated by dementia specialists, the use of amyloid PET was associated with changes in clinical management within 90 days. Further research is needed to determine whether amyloid PET is associated with improved clinical outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02420756.
Collapse
Affiliation(s)
- Gil D. Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
- Associate Editor, JAMA Neurology
| | - Constantine Gatsonis
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
- Department of Biostatistics, Brown University School of Public Health, Providence, Rhode Island
| | | | - Kiran Chaudhary
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Ilana Gareen
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
- Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island
| | - Lucy Hanna
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
| | | | - Bruce E. Hillner
- Department of Medicine, Virginia Commonwealth University, Richmond
| | | | - Orit H. Lesman-Segev
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Justin Romanoff
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
| | - Barry A. Siegel
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Rachel A. Whitmer
- Division of Research, Kaiser Permanente, Oakland, California
- Department of Public Health Sciences, University of California, Davis
| | | |
Collapse
|
46
|
Aghakhanyan G, Vergallo A, Gennaro M, Mazzarri S, Guidoccio F, Radicchi C, Ceravolo R, Tognoni G, Bonuccelli U, Volterrani D. The Precuneus – A Witness for Excessive Aβ Gathering in Alzheimer’s Disease Pathology. NEURODEGENER DIS 2019; 18:302-309. [DOI: 10.1159/000492945] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 08/15/2018] [Indexed: 11/19/2022] Open
Abstract
Evidence of cortical beta-amyloid (Aβ) load, assessed by Aβ positron emission tomography (Aβ-PET), is an established in vivo biomarker of Alzheimer’s disease (AD)-related pathophysiology. Qualitative assessment of Aβ-PET provides binary information; meanwhile semiquantitative approaches require a parcellation of PET image either manually or by placement of atlas-based volumes of interest. We supposed that a whole-brain approach with voxel-by-voxel standardized uptake value ratio (SUVr) parametric images may better elucidate the spatial trajectories of Aβ burden along the continuum of AD. Methods: We recruited 32 subjects with a diagnosis of probable AD dementia (ADD, n = 20) and mild cognitive impairment due to AD (MCI-AD, n = 12) according to the NIA-AA 2011 criteria. We also enrolled a control group of 6 cognitively healthy individuals (HCs) with preserved cognitive functions and negative Aβ-PET scan. The PET images were spatially normalized using the AV45 PET template in the MNI brain space. Subsequently, parametric SUVr images were calculated using the whole cerebellum as a reference region. A voxel-wise analysis of covariance was used to compare (between groups) the Αβ distribution pattern considering age as a nuisance covariate. Results: Both ADD and MCI-AD subjects showed a widespread increase in radiotracer uptake when compared with HC participants (p < 0.001, uncorrected). After applying a multiple comparison correction (p < 0.05, corrected), a relative large cluster of increased [18F]-florbetapir uptake was observed in the precuneus in the ADD and MCI-AD groups compared to HCs. Voxel-wise regression analysis showed a significant positive linear association between the voxel-wise SUVr values and the disease duration. Conclusions: The voxel-wise semiquantitative analysis shows that the precuneus is a region with higher vulnerability to Aβ depositions when compared to other cortical regions in both MCI-AD and ADD subjects. We think that the precuneus is a promising PET-based outcome measure for clinical trials of drugs targeting brain Aβ. We found a positive association between the overall Aβ-PET SUVr and the disease duration suggesting that the region-specific slow saturation of Aβ deposition continuously takes place as the disease progresses.
Collapse
|
47
|
Bouter C, Vogelgsang J, Wiltfang J. Comparison between amyloid-PET and CSF amyloid-β biomarkers in a clinical cohort with memory deficits. Clin Chim Acta 2019; 492:62-68. [PMID: 30735665 DOI: 10.1016/j.cca.2019.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 11/25/2022]
Abstract
With increasing prevalence of Alzheimer's disease (AD) and advances in research of therapeutic approaches, an early and accurate in-vivo diagnosis is crucial. Different biomarkers that are able to identify AD are currently in focus. However, whether and to which extend results of cerebrospinal fluid (CSF) and imaging biomarkers are comparable, is unclear. This study aims to correlate CSF and amyloid imaging biomarkers comparing them to cognitive measurements in order to determine whether these methods provide identical or complementary information. The study comprises 33 consecutive patients with suspected cognitive decline that underwent lumbar puncture for CSF biomarker analysis and Amyloid-PET/CT within the diagnostic evaluation of memory impairment. Amyloid PET/CTs were evaluated visually and quantitatively. CSF and imaging data were retrospectively evaluated and results were compared to cognition tests, age, gender, and ApoE status. Global cortex SUVr levels correlated highly with CSF Aβ42/40 and moderately with Aβ42 but not with Aβ40. Global cortex SUVr and Aβ42/40 correlated with mini mental status examination. This study indicates that Amyloid-PET and CSF biomarkers might not reflect identical clinical information and a combination of both seems to be the most accurate way to characterize clinically unclear cognitive decline.
Collapse
Affiliation(s)
- Caroline Bouter
- University Medical Center Goettingen (UMG), Georg-August-University, Dept. of Nuclear Medicine, Robert-Koch-Str. 40, D-37075 Goettingen, Germany.
| | - Jonathan Vogelgsang
- University Medical Center Goettingen (UMG), Georg-August-University, Dept. of Psychiatry and Psychotherapy, Von-Siebold-Str. 5, D-37075 Goettingen, Germany
| | - Jens Wiltfang
- University Medical Center Goettingen (UMG), Georg-August-University, Dept. of Psychiatry and Psychotherapy, Von-Siebold-Str. 5, D-37075 Goettingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, D-37075 Goettingen, Germany; iBiMED, Medical Science Department, University of Aveiro, Portugal
| |
Collapse
|
48
|
Molinuevo JL, Ayton S, Batrla R, Bednar MM, Bittner T, Cummings J, Fagan AM, Hampel H, Mielke MM, Mikulskis A, O'Bryant S, Scheltens P, Sevigny J, Shaw LM, Soares HD, Tong G, Trojanowski JQ, Zetterberg H, Blennow K. Current state of Alzheimer's fluid biomarkers. Acta Neuropathol 2018; 136:821-853. [PMID: 30488277 PMCID: PMC6280827 DOI: 10.1007/s00401-018-1932-x] [Citation(s) in RCA: 356] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with a complex and heterogeneous pathophysiology. The number of people living with AD is predicted to increase; however, there are no disease-modifying therapies currently available and none have been successful in late-stage clinical trials. Fluid biomarkers measured in cerebrospinal fluid (CSF) or blood hold promise for enabling more effective drug development and establishing a more personalized medicine approach for AD diagnosis and treatment. Biomarkers used in drug development programmes should be qualified for a specific context of use (COU). These COUs include, but are not limited to, subject/patient selection, assessment of disease state and/or prognosis, assessment of mechanism of action, dose optimization, drug response monitoring, efficacy maximization, and toxicity/adverse reactions identification and minimization. The core AD CSF biomarkers Aβ42, t-tau, and p-tau are recognized by research guidelines for their diagnostic utility and are being considered for qualification for subject selection in clinical trials. However, there is a need to better understand their potential for other COUs, as well as identify additional fluid biomarkers reflecting other aspects of AD pathophysiology. Several novel fluid biomarkers have been proposed, but their role in AD pathology and their use as AD biomarkers have yet to be validated. In this review, we summarize some of the pathological mechanisms implicated in the sporadic AD and highlight the data for several established and novel fluid biomarkers (including BACE1, TREM2, YKL-40, IP-10, neurogranin, SNAP-25, synaptotagmin, α-synuclein, TDP-43, ferritin, VILIP-1, and NF-L) associated with each mechanism. We discuss the potential COUs for each biomarker.
Collapse
Affiliation(s)
- José Luis Molinuevo
- BarcelonaBeta Brain Research Center, Fundació Pasqual Maragall, Universitat Pompeu Fabra, Barcelona, Spain
- Unidad de Alzheimer y otros trastornos cognitivos, Hospital Clinic-IDIBAPS, Barcelona, Spain
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Richard Batrla
- Roche Centralised and Point of Care Solutions, Roche Diagnostics International, Rotkreuz, Switzerland
| | - Martin M Bednar
- Neuroscience Therapeutic Area Unit, Takeda Development Centre Americas Ltd, Cambridge, MA, USA
| | - Tobias Bittner
- Genentech, A Member of the Roche Group, Basel, Switzerland
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Harald Hampel
- AXA Research Fund and Sorbonne University Chair, Paris, France
- Sorbonne University, GRC No 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain and Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Paris, France
- Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Michelle M Mielke
- Departments of Epidemiology and Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Sid O'Bryant
- Department of Pharmacology and Neuroscience; Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeffrey Sevigny
- Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Holly D Soares
- Clinical Development Neurology, AbbVie, North Chicago, IL, USA
| | | | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal Campus, Sahlgrenska University Hospital, 431 80, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal Campus, Sahlgrenska University Hospital, 431 80, Mölndal, Sweden.
| |
Collapse
|
49
|
de Rojas I, Romero J, Rodríguez-Gomez O, Pesini P, Sanabria A, Pérez-Cordon A, Abdelnour C, Hernández I, Rosende-Roca M, Mauleón A, Vargas L, Alegret M, Espinosa A, Ortega G, Gil S, Guitart M, Gailhajanet A, Santos-Santos MA, Moreno-Grau S, Sotolongo-Grau O, Ruiz S, Montrreal L, Martín E, Pelejà E, Lomeña F, Campos F, Vivas A, Gómez-Chiari M, Tejero MA, Giménez J, Pérez-Grijalba V, Marquié GM, Monté-Rubio G, Valero S, Orellana A, Tárraga L, Sarasa M, Ruiz A, Boada M. Correlations between plasma and PET beta-amyloid levels in individuals with subjective cognitive decline: the Fundació ACE Healthy Brain Initiative (FACEHBI). Alzheimers Res Ther 2018; 10:119. [PMID: 30497535 PMCID: PMC6267075 DOI: 10.1186/s13195-018-0444-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/29/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND Peripheral biomarkers that identify individuals at risk of developing Alzheimer's disease (AD) or predicting high amyloid beta (Aβ) brain burden would be highly valuable. To facilitate clinical trials of disease-modifying therapies, plasma concentrations of Aβ species are good candidates for peripheral AD biomarkers, but studies to date have generated conflicting results. METHODS The Fundació ACE Healthy Brain Initiative (FACEHBI) study uses a convenience sample of 200 individuals diagnosed with subjective cognitive decline (SCD) at the Fundació ACE (Barcelona, Spain) who underwent amyloid florbetaben(18F) (FBB) positron emission tomography (PET) brain imaging. Baseline plasma samples from FACEHBI subjects (aged 65.9 ± 7.2 years) were analyzed using the ABtest (Araclon Biotech). This test directly determines the free plasma (FP) and total plasma (TP) levels of Aβ40 and Aβ42 peptides. The association between Aβ40 and Aβ42 plasma levels and FBB-PET global standardized uptake value ratio (SUVR) was determined using correlations and linear regression-based methods. The effect of the APOE genotype on plasma Aβ levels and FBB-PET was also assessed. Finally, various models including different combinations of demographics, genetics, and Aβ plasma levels were constructed using logistic regression and area under the receiver operating characteristic curve (AUROC) analyses to evaluate their ability for discriminating which subjects presented brain amyloidosis. RESULTS FBB-PET global SUVR correlated weakly but significantly with Aβ42/40 plasma ratios. For TP42/40, this observation persisted after controlling for age and APOE ε4 allele carrier status (R2 = 0.193, p = 1.01E-09). The ROC curve demonstrated that plasma Aβ measurements are not superior to APOE and age in combination in predicting brain amyloidosis. It is noteworthy that using a simple preselection tool (the TP42/40 ratio with an empirical cut-off value of 0.08) optimizes the sensitivity and reduces the number of individuals subjected to Aβ FBB-PET scanners to 52.8%. No significant dependency was observed between APOE genotype and plasma Aβ measurements (p value for interaction = 0.105). CONCLUSION Brain and plasma Aβ levels are partially correlated in individuals diagnosed with SCD. Aβ plasma measurements, particularly the TP42/40 ratio, could generate a new recruitment strategy independent of the APOE genotype that would improve identification of SCD subjects with brain amyloidosis and reduce the rate of screening failures in preclinical AD studies. Independent replication of these findings is warranted.
Collapse
Affiliation(s)
- Itziar de Rojas
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | | | - O. Rodríguez-Gomez
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | | | - A. Sanabria
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - A. Pérez-Cordon
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - C. Abdelnour
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - I. Hernández
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - M. Rosende-Roca
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - A. Mauleón
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - L. Vargas
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - M. Alegret
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - A. Espinosa
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - G. Ortega
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - S. Gil
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - M. Guitart
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - A. Gailhajanet
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - M. A. Santos-Santos
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - Sonia Moreno-Grau
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - O. Sotolongo-Grau
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - S. Ruiz
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - L. Montrreal
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - E. Martín
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - E. Pelejà
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - F. Lomeña
- Servei de Medicina Nuclear, Hospital Clínic i Provincial, Barcelona, Spain
| | - F. Campos
- Servei de Medicina Nuclear, Hospital Clínic i Provincial, Barcelona, Spain
| | - A. Vivas
- Departament de Diagnòstic per la Imatge, Clínica Corachan, Barcelona, Spain
| | - M. Gómez-Chiari
- Departament de Diagnòstic per la Imatge, Clínica Corachan, Barcelona, Spain
| | - M. A. Tejero
- Departament de Diagnòstic per la Imatge, Clínica Corachan, Barcelona, Spain
| | - J. Giménez
- Departament de Diagnòstic per la Imatge, Clínica Corachan, Barcelona, Spain
| | | | - G. M. Marquié
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - G. Monté-Rubio
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - S. Valero
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - A. Orellana
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - L. Tárraga
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | | | - A. Ruiz
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| | - M. Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, C/ Marquès de Sentmenat, 57, 08029 Barcelona, Spain
| |
Collapse
|
50
|
Palmqvist S, Insel PS, Zetterberg H, Blennow K, Brix B, Stomrud E, Mattsson N, Hansson O. Accurate risk estimation of β-amyloid positivity to identify prodromal Alzheimer's disease: Cross-validation study of practical algorithms. Alzheimers Dement 2018; 15:194-204. [PMID: 30365928 PMCID: PMC6374284 DOI: 10.1016/j.jalz.2018.08.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/14/2018] [Accepted: 08/21/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The aim was to create readily available algorithms that estimate the individual risk of β-amyloid (Aβ) positivity. METHODS The algorithms were tested in BioFINDER (n = 391, subjective cognitive decline or mild cognitive impairment) and validated in Alzheimer's Disease Neuroimaging Initiative (n = 661, subjective cognitive decline or mild cognitive impairment). The examined predictors of Aβ status were demographics; cognitive tests; white matter lesions; apolipoprotein E (APOE); and plasma Aβ42/Aβ40, tau, and neurofilament light. RESULTS Aβ status was accurately estimated in BioFINDER using age, 10-word delayed recall or Mini-Mental State Examination, and APOE (area under the receiver operating characteristics curve = 0.81 [0.77-0.85] to 0.83 [0.79-0.87]). When validated, the models performed almost identical in Alzheimer's Disease Neuroimaging Initiative (area under the receiver operating characteristics curve = 0.80-0.82) and within different age, subjective cognitive decline, and mild cognitive impairment populations. Plasma Aβ42/Aβ40 improved the models slightly. DISCUSSION The algorithms are implemented on http://amyloidrisk.com where the individual probability of being Aβ positive can be calculated. This is useful in the workup of prodromal Alzheimer's disease and can reduce the number needed to screen in Alzheimer's disease trials.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden; Department of Neurology, Skåne University Hospital, Lund, Sweden.
| | - Philip S Insel
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom; UK Dementia Research Institute at UCL, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | | | - Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden; Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|