1
|
Wang MY, Chen KL, Huang YY, Chen SF, Wang RZ, Zhang Y, Hu HY, Ma LZ, Liu WS, Wang J, Xin JW, Zhang X, Li MM, Guo Y, Dong Q, Cheng W, Tan L, Cui M, Zhang YR, Yu JT. Clinical utility of cerebrospinal fluid Alzheimer's disease biomarkers in the diagnostic workup of complex patients with cognitive impairment. Transl Psychiatry 2025; 15:130. [PMID: 40195333 PMCID: PMC11976989 DOI: 10.1038/s41398-025-03345-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers have been widely adopted in Alzheimer's disease (AD) diagnosis. However, no studies focused on the application of CSF biomarkers in the clinical practice of complex and atypical patients with cognitive impairment in China. This study aimed to evaluate the added value of CSF AD biomarkers in cognitively impaired patients with complex conditions in a memory clinical setting. A total of 633 participants were included from the National Center for Neurological Disorders in Shanghai, China. The CSF AD biomarkers were measured with ELISA. Cutoff values were firstly identified using Youden's index. The neurologists proposed etiology diagnosis with a percentage estimate of their confidence and prescribed medication before and after CSF disclosure. Changes in etiological diagnosis, diagnostic confidence, and management plan were compared across the groups. Of the 633 patients (mean [SD] age, 61.1 [11.3] years; 295 males [46.6%]), 372 (58.8%) were diagnosed with dementia, 103 (16.3%) with mild cognitive impairment, and 158 (24.9%) with subjective cognitive decline. Using those pre-defined cutoffs, we categorized patients into 3 groups: Alzheimer's continuum (68.1%), non-AD pathologic change (11.1%), and normal AD biomarkers (20.8%). After CSF disclosure, the proposed etiology changed in 158 (25.0%) participants and the prescribed medication changed in 200 (31.6%) patients. Mean diagnostic confidence increased from 69.5-83.0% (+13.5%; P < 0.001). In conclusion, CSF AD biomarkers significantly impacted the diagnosis, diagnostic confidence, and treatment plans for Chinese patients with complex cognitive impairment. CSF AD biomarkers are a useful tool for clinicians beyond routine clinical assessment.
Collapse
Affiliation(s)
- Ming-Yu Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Weifang People's Hospital, Weifang, China
| | - Ke-Liang Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shu-Fen Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong-Ze Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ling-Zhi Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jia-Wei Xin
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xue Zhang
- Department of Neurology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Meng-Meng Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Mei Cui
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Kale MB, Bhondge HM, Wankhede NL, Shende PV, Thanekaer RP, Aglawe MM, Rahangdale SR, Taksande BG, Pandit SB, Upaganlawar AB, Umekar MJ, Kopalli SR, Koppula S. Navigating the intersection: Diabetes and Alzheimer's intertwined relationship. Ageing Res Rev 2024; 100:102415. [PMID: 39002642 DOI: 10.1016/j.arr.2024.102415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) and Diabetes mellitus (DM) exhibit comparable pathophysiological pathways. Genetic abnormalities in APP, PS-1, and PS-2 are linked to AD, with diagnostic aid from CSF and blood biomarkers. Insulin dysfunction, termed "type 3 diabetes mellitus" in AD, involves altered insulin signalling and neuronal shrinkage. Insulin influences beta-amyloid metabolism, exacerbating neurotoxicity in AD and amyloid production in DM. Both disorders display impaired glucose transporter expression, hastening cognitive decline. Mitochondrial dysfunction and Toll-like receptor 4-mediated inflammation worsen neurodegeneration in both diseases. ApoE4 raises disease risk, especially when coupled with dyslipidemia common in DM. Targeting shared pathways like insulin-degrading enzyme activation and HSP60 holds promise for therapeutic intervention. Recognizing these interconnected mechanisms underscores the imperative for developing tailored treatments addressing the overlapping pathophysiology of AD and DM, offering potential avenues for more effective management of both conditions.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | | | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Rushikesh P Thanekaer
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sunil B Pandit
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
3
|
Parvizi T, Wurm R, König T, Silvaieh S, Altmann P, Klotz S, Regelsberger G, Traub‐Weidinger T, Gelpi E, Stögmann E. Real-world performance of plasma p-tau181 in a heterogeneous memory clinic cohort. Ann Clin Transl Neurol 2024; 11:1988-1998. [PMID: 38965832 PMCID: PMC11330220 DOI: 10.1002/acn3.52116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/22/2024] [Accepted: 05/23/2024] [Indexed: 07/06/2024] Open
Abstract
OBJECTIVE In light of clinical trials and disease-modifying therapies, an early identification of patients at-risk of developing Alzheimer's disease (AD) is crucial. Blood-based biomarkers have shown promising results regarding the in vivo detection of the earliest neuropathological changes in AD. Herein, we investigated the ability of plasma p-tau181 to act as a prescreening marker for amyloid positivity in a heterogeneous memory clinic-based cohort. METHODS In this retrospective cross-sectional study, we included a total of 115 patients along the clinical AD continuum (mild cognitive impairment [MCI] due to AD, n = 62, probable AD dementia, n = 53). Based on their biomarker status, they were stratified into an amyloid-positive (Aβ+, n = 88) or amyloid-negative cohort (Aβ-, n = 27). Plasma and CSF p-tau181 concentrations were quantified using an ultrasensitive single-molecule array (SIMOA©). Furthermore, age- and sex-adjusted receiver operating characteristic (ROC) curves were calculated and the area under the curve (AUC) of each model was compared using DeLong's test for correlated AUC curves. RESULTS The median (interquartile range [IQR]) concentration of plasma p-tau181 was significantly higher in Aβ+ patients (3.6 pg/mL [2.5-4.6]), compared with Aβ- patients (1.7 pg/mL [1.2-1.9], p < 0.001). Regarding the distinction between Aβ+ and Aβ- patients and the prediction of amyloid positivity, a high diagnostic accuracy for plasma p-tau181 with an AUC of 0.89 (95% CI = 0.82-0.95) was calculated. Adding the risk factors, age and APOE4, to the model did not significantly improve its performance. INTERPRETATION Our findings demonstrate that plasma p-tau181 could be a noninvasive and feasible prescreening marker for amyloid positivity in a heterogeneous clinical AD cohort and therefore help in identifying those who would benefit from more invasive assessment of amyloid pathology.
Collapse
Affiliation(s)
- Tandis Parvizi
- Department of NeurologyMedical University of ViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| | - Raphael Wurm
- Department of NeurologyMedical University of ViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| | - Theresa König
- Department of NeurologyMedical University of ViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| | - Sara Silvaieh
- Department of NeurologyMedical University of ViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| | - Patrick Altmann
- Department of NeurologyMedical University of ViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| | - Sigrid Klotz
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| | - Guenther Regelsberger
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| | - Tatjana Traub‐Weidinger
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
| | - Ellen Gelpi
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| | - Elisabeth Stögmann
- Department of NeurologyMedical University of ViennaViennaAustria
- Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| |
Collapse
|
4
|
Patel KJ, Yang D, Best JR, Chambers C, Lee PE, Henri-Bhargava A, Funnell CR, Foti DJ, Pettersen JA, Feldman HH, Nygaard HB, Hsiung GYR, DeMarco ML. Clinical value of Alzheimer's disease biomarker testing. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12464. [PMID: 38596484 PMCID: PMC10999950 DOI: 10.1002/trc2.12464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION In the Investigating the Impact of Alzheimer's Disease Diagnostics in British Columbia (IMPACT-AD BC) study, we aimed to understand how Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarker testing-used in medical care-impacted medical decision-making (medical utility), personal decision-making (personal utility), and health system economics. METHODS The study was designed as an observational, longitudinal cohort study. A total of 149 patients were enrolled between February 2019 and July 2021. Patients referred to memory clinics were approached to participate if their dementia specialist ordered AD CSF biomarker testing as part of their routine medical care, and the clinical scenario met the appropriate use criteria for lumbar puncture and AD CSF biomarker testing. For the medical utility pillar, detailed clinical management plans were collected via physician questionnaires pre- and post-biomarker disclosure. RESULTS Patients with completed management questionnaires (n = 142) had a median age of 64 (interquartile range: 59-69) years, 48% were female, and 60% had CSF biomarker profiles on the AD continuum. Clinical management changed in 89.4% of cases. AD biomarker testing was associated with decreased need for other diagnostic procedures, including brain imaging (-52.0%) and detailed neuropsychological assessments (-63.2%), increased referrals and counseling (57.0%), and guided AD-related drug prescriptions (+88.4% and -50.0% in biomarker-positive and -negative cases, respectively). DISCUSSION AD biomarker testing was associated with significant and positive changes in clinical management, including decreased health care resource use, therapy optimization, and increased patient and family member counseling. While certain changes in management were linked to the AD biomarker profile (e.g., referral to clinical trials), the majority of changes were independent of baseline clinical presentation and level of cognitive impairment, demonstrating a broad value for AD biomarker testing in individuals meeting the appropriate use criteria for testing.
Collapse
Affiliation(s)
- Khushbu J Patel
- Department of Pathology and Laboratory Medicine University of British Columbia Vancouver Canada
| | - David Yang
- Department of Pathology and Laboratory Medicine University of British Columbia Vancouver Canada
| | - John R Best
- Gerontology Research Centre Simon Fraser University Vancouver Canada
| | - Colleen Chambers
- Department of Pathology and Laboratory Medicine University of British Columbia Vancouver Canada
| | - Philip E Lee
- Division of Neurology Department of Medicine University of British Columbia Vancouver Canada
- Djavad Mowafaghian Centre for Brain Health Department of Medicine University of British Columbia Vancouver Canada
- UBC Hospital Clinic for Alzheimer Disease and Related Disorders University of British Columbia Vancouver Canada
| | - Alexandre Henri-Bhargava
- Division of Neurology Department of Medicine University of British Columbia Vancouver Canada
- Division of Medical Sciences University of Victoria Victoria Canada
| | - Clark R Funnell
- Division of Neurology Department of Medicine University of British Columbia Vancouver Canada
- Djavad Mowafaghian Centre for Brain Health Department of Medicine University of British Columbia Vancouver Canada
- UBC Hospital Clinic for Alzheimer Disease and Related Disorders University of British Columbia Vancouver Canada
| | - Dean J Foti
- Division of Neurology Department of Medicine University of British Columbia Vancouver Canada
- Djavad Mowafaghian Centre for Brain Health Department of Medicine University of British Columbia Vancouver Canada
- UBC Hospital Clinic for Alzheimer Disease and Related Disorders University of British Columbia Vancouver Canada
| | - Jacqueline A Pettersen
- Division of Neurology Department of Medicine University of British Columbia Vancouver Canada
- Division of Medical Sciences University of Northern British Columbia Prince George Canada
| | - Howard H Feldman
- Department of Neurosciences University of California San Diego San Diego California USA
- Alzheimer Disease Cooperative Study University of California San Diego San Diego California USA
- Alzheimer's and Related Neurodegenerative Research University of California San Diego San Diego California USA
| | - Haakon B Nygaard
- Division of Neurology Department of Medicine University of British Columbia Vancouver Canada
- Djavad Mowafaghian Centre for Brain Health Department of Medicine University of British Columbia Vancouver Canada
- UBC Hospital Clinic for Alzheimer Disease and Related Disorders University of British Columbia Vancouver Canada
| | - Ging-Yuek R Hsiung
- Division of Neurology Department of Medicine University of British Columbia Vancouver Canada
- Djavad Mowafaghian Centre for Brain Health Department of Medicine University of British Columbia Vancouver Canada
- UBC Hospital Clinic for Alzheimer Disease and Related Disorders University of British Columbia Vancouver Canada
| | - Mari L DeMarco
- Department of Pathology and Laboratory Medicine University of British Columbia Vancouver Canada
- Department of Pathology and Laboratory Medicine St. Paul's Hospital Providence Health Care Vancouver Canada
| |
Collapse
|
5
|
Moonen JEF, Haan R, Bos I, Teunissen C, van de Giessen E, Tomassen J, den Braber A, van der Landen SM, de Geus EJC, Legdeur N, van Harten AC, Trieu C, de Boer C, Kroeze L, Barkhof F, Visser PJ, van der Flier WM. Contributions of amyloid beta and cerebral small vessel disease in clinical decline. Alzheimers Dement 2024; 20:1868-1880. [PMID: 38146222 PMCID: PMC10984432 DOI: 10.1002/alz.13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION We assessed whether co-morbid small vessel disease (SVD) has clinical predictive value in preclinical or prodromal Alzheimer's disease. METHODS In 1090 non-demented participants (65.4 ± 10.7 years) SVD was assessed with magnetic resonance imaging and amyloid beta (Aβ) with lumbar puncture and/or positron emission tomography scan (mean follow-up for cognitive function 3.1 ± 2.4 years). RESULTS Thirty-nine percent had neither Aβ nor SVD (A-V-), 21% had SVD only (A-V+), 23% Aβ only (A+V-), and 17% had both (A+V+). Pooled cohort linear mixed model analyses demonstrated that compared to A-V- (reference), A+V- had a faster rate of cognitive decline. Co-morbid SVD (A+V+) did not further increase rate of decline. Cox regression showed that dementia risk was modestly increased in A-V+ (hazard ratio [95% confidence interval: 1.8 [1.0-3.2]) and most strongly in A+ groups. Also, mortality risk was increased in A+ groups. DISCUSSION In non-demented persons Aβ was predictive of cognitive decline, dementia, and mortality. SVD modestly predicts dementia in A-, but did not increase deleterious effects in A+. HIGHLIGHTS Amyloid beta (Aβ; A) was predictive for cognitive decline, dementia, and mortality. Small vessel disease (SVD) had no additional deleterious effects in A+. SVD modestly predicted dementia in A-. Aβ should be assessed even when magnetic resonance imaging indicates vascular cognitive impairment.
Collapse
Affiliation(s)
- Justine E. F. Moonen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Renée Haan
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Isabelle Bos
- Nivel, Research Institute for Better CareUtrechtthe Netherlands
| | - Charlotte Teunissen
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Neurochemistry LaboratoryDepartment of Clinical ChemistryAmsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
| | - Jori Tomassen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Sophie M. van der Landen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Eco J. C. de Geus
- Department of Biological PsychologyVU UniversityAmsterdamthe Netherlands
| | - Nienke Legdeur
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Argonde C. van Harten
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Calvin Trieu
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Casper de Boer
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Lior Kroeze
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Institute of Healthcare Engineering and the Institute of Neurology, University College LondonLondonUK
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNS), Maastricht UniversityMaastrichtthe Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of NeurogeriatricsKarolinska InstitutetSolnaSweden
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| |
Collapse
|
6
|
Sánchez‐Soblechero A, Berbel A, Villarejo A, Palmí‐Cortés I, Vieira A, Gil‐Moreno MJ, Fernández C, Martín‐Montes Ã, Carreras MT, Fernández Y, Puertas C, Blanco‐Palmero V, Llamas S, González‐Sánchez M, Lapeña T, de Luis P, Manzano S, Olazarán J. Translating NIA-AA criteria into usual practice: Report from the ReDeMa Project. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12451. [PMID: 38505833 PMCID: PMC10948948 DOI: 10.1002/trc2.12451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/23/2023] [Accepted: 12/30/2023] [Indexed: 03/21/2024]
Abstract
INTRODUCTION Biomarker-informed criteria were proposed for the diagnosis of Alzheimer's disease (AD) by the National Institute on Aging and the Alzheimer's Association (NIA-AA) in 2011; however, the adequacy of this criteria has not been sufficiently evaluated. METHODS ReDeMa (Red de Demencias de Madrid) is a regional cohort of patients attending memory and neurology clinics. Core cerebrospinal fluid biomarkers were obtained, NIA-AA diagnostic criteria were considered, and changes in diagnosis and management were evaluated. RESULTS A total of 233 patients were analyzed (mean age 70 years, 50% women, 73% AD). The diagnostic language was modified significantly, with a majority assumption of NIA-AA definitions (69%). Confidence in diagnosis increased from 70% to 92% (p < 0.0005) and management was changed in 71% of patient/caregivers. The influence of neurologist's age or expertise on study results was minimal. DISCUSSION The NIA-AA criteria are adequate and utile for usual practice in memory and neurology clinics, improving diagnostic confidence and significantly modifying patient management. HIGHLIGHTS Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers increase diagnostic certainty regardless of the neurologist.AD CSF biomarkers lead to changes in disease management .Biomarker-enriched, 2011 NIA-AA diagnostic criteria are adequate for usual practice.
Collapse
Affiliation(s)
| | | | | | - Itziar Palmí‐Cortés
- Neurology ServiceUniversity Hospital Infanta Sofía, San Sebastián de los ReyesMadridSpain
| | - Alba Vieira
- Neurology ServiceUniversity Hospital la PrincesaMadridSpain
| | | | | | - Ãngel Martín‐Montes
- Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital – Universidad Autónoma de Madrid)MadridSpain
| | | | - Yolanda Fernández
- Memory Disorders Clinic ‐ HM Hospitals and Neurology Service ‐ University Hospital Gregorio MarañónMadridSpain
| | - Carolina Puertas
- Clinical Biochemistry ServiceUniversity Hospital Gregorio MarañónMadridSpain
| | | | - Sara Llamas
- Neurology ServiceUniversity Hospital 12 de OctubreMadridSpain
| | - Marta González‐Sánchez
- Neurology ServiceUniversity Hospital 12 de OctubreMadridSpain
- Group of Neurodegenerative DiseasesUniversity Hospital 12 de Octubre Research Institute (imas12), and Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | | | | | | | - Javier Olazarán
- Memory Disorders Clinic ‐ HM HospitalsNeurology Service ‐ University Hospital Gregorio Marañón, and Maria Wolff FoundationMadridSpain
| |
Collapse
|
7
|
Coomans EM, de Koning LA, Rikken RM, Verfaillie SCJ, Visser D, den Braber A, Tomassen J, van de Beek M, Collij LE, Lemstra AW, Windhorst AD, Barkhof F, Golla SSV, Visser PJ, Scheltens P, van der Flier WM, Ossenkoppele R, van Berckel BNM, van de Giessen E. Performance of a [ 18F]Flortaucipir PET Visual Read Method Across the Alzheimer Disease Continuum and in Dementia With Lewy Bodies. Neurology 2023; 101:e1850-e1862. [PMID: 37748892 PMCID: PMC10663007 DOI: 10.1212/wnl.0000000000207794] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/24/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Recently, the US Food and Drug Administration approved the tau-binding radiotracer [18F]flortaucipir and an accompanying visual read method to support the diagnostic process in cognitively impaired patients assessed for Alzheimer disease (AD). Studies evaluating this visual read method are limited. In this study, we evaluated the performance of the visual read method in participants along the AD continuum and dementia with Lewy bodies (DLB) by determining its reliability, accordance with semiquantitative analyses, and associations with clinically relevant variables. METHODS We included participants who underwent tau-PET at Amsterdam University Medical Center. A subset underwent follow-up tau-PET. Two trained nuclear medicine physicians visually assessed all scans. Inter-reader agreement was calculated using Cohen κ. To examine the concordance of visual read tau positivity with semiquantification, we defined standardized uptake value ratio (SUVr) positivity using different threshold approaches. To evaluate the prognostic value of tau-PET visual read, we performed linear mixed models with longitudinal Mini-Mental State Examination (MMSE). RESULTS We included 263 participants (mean age 68.5 years, 45.6% female), including 147 cognitively unimpaired (CU) participants, 97 amyloid-positive participants with mild cognitive impairment or AD dementia (AD), and 19 participants with DLB. The visual read inter-reader agreement was excellent (κ = 0.95, CI 0.91-0.99). None of the amyloid-negative CU participants (0/92 [0%]) and 1 amyloid-negative participant with DLB (1/12 [8.3%]) were tau-positive. Among amyloid-positive participants, 13 CU participants (13/52 [25.0%]), 85 with AD (85/97 [87.6%]), and 3 with DLB (3/7 [42.9%]) were tau-positive. Two-year follow-up visual read status was identical to baseline. Tau-PET visual read corresponded strongly to SUVr status, with up to 90.4% concordance. Visual read tau positivity was associated with a decline on the MMSE in CU participants (β = -0.52, CI -0.74 to -0.30, p < 0.001) and participants with AD (β = -0.30, CI -0.58 to -0.02, p = 0.04). DISCUSSION The excellent inter-reader agreement, strong correspondence with SUVr, and longitudinal stability indicate that the visual read method is reliable and robust, supporting clinical application. Furthermore, visual read tau positivity was associated with prospective cognitive decline, highlighting its additional prognostic potential. Future studies in unselected cohorts are needed for a better generalizability to the clinical population. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that [18F]flortaucipir visual read accurately distinguishes patients with low tau-tracer binding from those with high tau-tracer binding and is associated with amyloid positivity and cognitive decline.
Collapse
Affiliation(s)
- Emma M Coomans
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden.
| | - Lotte A de Koning
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Roos M Rikken
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Sander C J Verfaillie
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Denise Visser
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Anouk den Braber
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Jori Tomassen
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Marleen van de Beek
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Lyduine E Collij
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Afina W Lemstra
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Albert D Windhorst
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Frederik Barkhof
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Sandeep S V Golla
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Pieter Jelle Visser
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Philip Scheltens
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Wiesje M van der Flier
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Rik Ossenkoppele
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Bart N M van Berckel
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| | - Elsmarieke van de Giessen
- From the Radiology & Nuclear Medicine (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Brain Imaging (E.M.C., L.A.d.K., R.M.R., S.C.J.V., D.V., L.E.C., A.D.W., F.B., S.S.V.G., B.N.M.v.B., E.v.d.G.), Amsterdam Neuroscience; Medical Psychology (S.C.J.V.), Amsterdam UMC location University of Amsterdam; Alzheimer Center Amsterdam (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Neurodegeneration (A.d.B., J.T., M.v.d.B., A.W.L., P.J.V., P.S., W.M.v.d.F., R.O.), Amsterdam Neuroscience; Department of Biological Psychology (A.d.B.), Vrije Universiteit Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, United Kingdom; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology & Data Science (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and Clinical Memory Research Unit (R.O.), Lund University, Sweden
| |
Collapse
|
8
|
Mank A, van Maurik IS, Rijnhart JJM, Rhodius‐Meester HFM, Visser LNC, Lemstra AW, Sikkes SAM, Teunissen CE, van Giessen EM, Berkhof J, van der Flier WM. Determinants of informal care time, distress, depression, and quality of life in care partners along the trajectory of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12418. [PMID: 37114014 PMCID: PMC10126754 DOI: 10.1002/dad2.12418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/19/2023] [Accepted: 03/01/2023] [Indexed: 04/29/2023]
Abstract
Introduction We evaluated determinants associated with care partner outcomes along the Alzheimer's disease (AD) stages. Methods We included n = 270 care partners of amyloid-positive patients in the pre-dementia and dementia stages of AD. Using linear regression analysis, we examined determinants of four care partner outcomes: informal care time, caregiver distress, depression, and quality of life (QoL). Results More behavioral symptoms and functional impairment in patients were associated with more informal care time and depressive symptoms in care partners. More behavioral symptoms were related with more caregiver distress. Spouse care partners spent more time on informal care and QoL was lower in female care partners. Behavioral problems and subtle functional impairment of the patient predisposed for worse care partner outcomes already in the pre-dementia stages. Discussion Both patient and care partner determinants contribute to the care partner outcomes, already in early disease stages. This study provides red flags for high care partner burden.
Collapse
Affiliation(s)
- Arenda Mank
- Alzheimer Center Amsterdam, Department of NeurologyVrije Universiteit AmsterdamAmsterdam UMC VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
- Amsterdam UMCVrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamthe Netherlands
| | - Ingrid S. van Maurik
- Alzheimer Center Amsterdam, Department of NeurologyVrije Universiteit AmsterdamAmsterdam UMC VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
- Amsterdam UMCVrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamthe Netherlands
| | | | - Hanneke F. M. Rhodius‐Meester
- Alzheimer Center Amsterdam, Department of NeurologyVrije Universiteit AmsterdamAmsterdam UMC VUmcAmsterdamthe Netherlands
- Department of Internal MedicineGeriatric Medicine SectionVrije Universiteit AmsterdamAmsterdam UMCAmsterdamthe Netherlands
- Department of Geriatric MedicineThe Memory ClinicOslo University HospitalOsloNorway
| | - Leonie N. C. Visser
- Department of Medical PsychologyAmsterdam UMC, AMCUniversity of AmsterdamAmsterdamthe Netherlands
- Amsterdam Public Health Research InstituteQuality of CareAmsterdamthe Netherlands
| | - Afina W. Lemstra
- Alzheimer Center Amsterdam, Department of NeurologyVrije Universiteit AmsterdamAmsterdam UMC VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
| | - Sietske A. M. Sikkes
- Alzheimer Center Amsterdam, Department of NeurologyVrije Universiteit AmsterdamAmsterdam UMC VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
| | - Charlotte E. Teunissen
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
- Neurochemistry LaboratoryDepartment of Clinical ChemistryVrije UniversiteitAmsterdam UMC, VUmcAmsterdamthe Netherlands
| | - Elsmarieke M. van Giessen
- Department of Radiology & Nuclear Medicine Vrije Universiteit AmsterdamAmsterdam UMC, VUmcAmsterdamthe Netherlands
| | - Johannes Berkhof
- Amsterdam UMCVrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamthe Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Department of NeurologyVrije Universiteit AmsterdamAmsterdam UMC VUmcAmsterdamthe Netherlands
- Amsterdam NeuroscienceNeurodegenerationAmsterdamthe Netherlands
- Amsterdam UMCVrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamthe Netherlands
| |
Collapse
|
9
|
Hazan J, Wing M, Liu KY, Reeves S, Howard R. Clinical utility of cerebrospinal fluid biomarkers in the evaluation of cognitive impairment: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 2023; 94:113-120. [PMID: 36096664 DOI: 10.1136/jnnp-2022-329530] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/29/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND The analytical and clinical validity of cerebrospinal (CSF) biomarkers has been extensively researched in dementia. Further work is needed to assess the ability of these biomarkers to improve diagnosis, management and health outcomes in the clinical setting OBJECTIVES: To assess the added value and clinical utility of CSF biomarkers in the diagnostic assessment of cognitively impaired patients under evaluation for Alzheimer's disease (AD). METHODS Systematic literature searches of Medline, EMBASE, PsycINFO and Web of Science research databases were conducted on 17 December 2022. Data from relevant studies were extracted and independently screened for quality using a tool for bias. Clinical utility was measured by clinicians' changes in diagnosis, diagnostic confidence and patient management (when available), after their examination of patients' CSF biomarkers. Cost-effectiveness was assessed by consideration of additional cost per patient and quality-adjusted life years. RESULTS Searches identified 17 studies comprising 2090 patient participants and 593 clinicians. The meta-analysis revealed that clinicians' use of CSF biomarkers resulted in a pooled percentage change in diagnosis of 25% (95% CI 14 to 37), an increase in diagnostic confidence of 14% (95% CI 9 to 18) and a pooled proportion of patients whose management changed of 31% (95% CI 12 to 50). CSF biomarkers were deemed cost-effective, particularly in memory services, where pre-test AD prevalence is higher compared with a primary care setting. CONCLUSIONS CSF biomarkers can be a helpful additional diagnostic tool for clinicians assessing patients with cognitive impairment. In particular, CSF biomarkers consistently improved clinicians' confidence in diagnosing AD and influenced on diagnostic change and patient management. Further research is needed to study the clinical utility of blood-based biomarkers in the clinical setting.
Collapse
Affiliation(s)
- Jemma Hazan
- Division of Psychiatry, University College London, London, UK
| | - Michelle Wing
- Division of Psychiatry, University College London, London, UK
| | - Kathy Y Liu
- Division of Psychiatry, University College London, London, UK
| | - Suzanne Reeves
- Division of Psychiatry, University College London, London, UK
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
10
|
Mank A, van Maurik IS, van Harten AC, Rhodius‐Meester HFM, Teunissen CE, van Berckel BNM, Berkhof J, van der Flier WM, Rijnhart JJM. Life satisfaction across the entire trajectory of Alzheimer's disease: A mediation analysis. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12389. [PMID: 36579132 PMCID: PMC9780509 DOI: 10.1002/dad2.12389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Introduction We studied life satisfaction across Alzheimer's disease (AD) stages and studied mobility and meaningful activities as mediators of the associations between these AD stages and life satisfaction. Methods In this cross-sectional study, we included n = 269 amyloid-positive patients with subjective cognitive decline (SCD), mild cognitive impairment (MCI), and AD dementia from the Amsterdam Dementia Cohort. Life satisfaction was measured with the satisfaction with life scale. The mediating role of transportation, work, sports, and hobbies on life satisfaction was examined in single and multiple mediator models. Results Patients with dementia are less satisfied with life compared to SCD and MCI. These differences in life satisfaction are explained by reduced participation in meaningful activities, which in turn, was largely attributable to decreased transportation use. Discussion Our findings suggest that improving access to transportation, therewith allowing participation in meaningful activities help to maintain life satisfaction and may be an important target for intervention.
Collapse
Affiliation(s)
- Arenda Mank
- Alzheimer Center Amsterdam, NeurologyAmsterdam UMC location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamThe Netherlands
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamThe Netherlands
| | - Ingrid S. van Maurik
- Alzheimer Center Amsterdam, NeurologyAmsterdam UMC location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamThe Netherlands
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamThe Netherlands
| | - Argonde C. van Harten
- Alzheimer Center Amsterdam, NeurologyAmsterdam UMC location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamThe Netherlands
| | - Hanneke F. M. Rhodius‐Meester
- Alzheimer Center Amsterdam, NeurologyAmsterdam UMC location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Internal MedicineGeriatric Medicine SectionVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Clinical ChemistryAmsterdam UMC location VUmcVrije UniversiteitAmsterdamThe Netherlands
| | - Bart N. M. van Berckel
- Alzheimer Center Amsterdam, NeurologyAmsterdam UMC location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamThe Netherlands
- Department of Radiology & Nuclear Medicine Amsterdam Neuroscience Vrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
| | - Johannes Berkhof
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamThe Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, NeurologyAmsterdam UMC location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamThe Netherlands
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamThe Netherlands
| | - Judith J. M. Rijnhart
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of Epidemiology and Data ScienceAmsterdam Public Health InstituteAmsterdamThe Netherlands
| |
Collapse
|
11
|
Mank A, Rijnhart JJM, van Maurik IS, Jönsson L, Handels R, Bakker ED, Teunissen CE, van Berckel BNM, van Harten AC, Berkhof J, van der Flier WM. A longitudinal study on quality of life along the spectrum of Alzheimer's disease. Alzheimers Res Ther 2022; 14:132. [PMID: 36109800 PMCID: PMC9476356 DOI: 10.1186/s13195-022-01075-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Quality of life (QoL) is an important outcome from the perspective of patients and their caregivers, in both dementia and pre-dementia stages. Yet, little is known about the long-term changes in QoL over time. We aimed to compare the trajectories of QoL between amyloid-positive and amyloid-negative SCD or MCI patients and to evaluate QoL trajectories along the Alzheimer's disease (AD) continuum of cognitively normal to dementia. METHODS We included longitudinal data of 447 subjective cognitive decline (SCD), 276 mild cognitive impairment (MCI), and 417 AD dementia patients from the Amsterdam Dementia Cohort. We compared QoL trajectories (EQ-5D and visual analog scale (VAS)) between (1) amyloid-positive and amyloid-negative SCD or MCI patients and (2) amyloid-positive SCD, MCI, and dementia patients with linear mixed-effect models. The models were adjusted for age, sex, Charlson Comorbidity Index (CCI), education, and EQ-5D scale (3 or 5 level). RESULTS In SCD, amyloid-positive participants had a higher VAS at baseline but showed a steeper decline over time in EQ-5D and VAS than amyloid-negative participants. Also, in MCI, amyloid-positive patients had higher QoL at baseline but subsequently showed a steeper decline in QoL over time compared to amyloid-negative patients. When we compared amyloid-positive patients along the Alzheimer continuum, we found no difference between SCD, MCI, or dementia in baseline QoL, but QoL decreased at a faster rate in the dementia stage compared with the of SCD and MCI stages. CONCLUSIONS QoL decreased at a faster rate over time in amyloid-positive SCD or MCI patients than amyloid-negative patients. QoL decreases over time along the entire AD continuum of SCD, MCI and dementia, with the strongest decrease in dementia patients. Knowledge of QoL trajectories is essential for the future evaluation of treatments in AD.
Collapse
Affiliation(s)
- Arenda Mank
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands. .,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands. .,Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam, The Netherlands.
| | - Judith J M Rijnhart
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam, The Netherlands
| | - Ingrid S van Maurik
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam, The Netherlands
| | - Linus Jönsson
- Department for Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
| | - Ron Handels
- Department for Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden.,Department of Psychiatry and Neuropsychology, Alzheimer Centre Limburg, School for Mental Health and Neurosciences, Maastricht University, Maastricht, The Netherlands
| | - Els D Bakker
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands.,Department of Radiology & Nuclear Medicine Amsterdam Neuroscience Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Argonde C van Harten
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Johannes Berkhof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, De Boelelaan 1118, 1081, HZ, Amsterdam, The Netherlands.,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Parvizi T, König T, Wurm R, Silvaieh S, Altmann P, Klotz S, Rommer PS, Furtner J, Regelsberger G, Lehrner J, Traub-Weidinger T, Gelpi E, Stögmann E. Real-world applicability of glial fibrillary acidic protein and neurofilament light chain in Alzheimer's disease. Front Aging Neurosci 2022; 14:887498. [PMID: 36072480 PMCID: PMC9441692 DOI: 10.3389/fnagi.2022.887498] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Blood-based biomarkers may add a great benefit in detecting the earliest neuropathological changes in patients with Alzheimer's disease (AD). We examined the utility of neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) regarding clinical diagnosis and differentiation between amyloid positive and negative patients. To evaluate the practical application of these biomarkers in a routine clinical setting, we conducted this study in a heterogeneous memory-clinic population. Methods: We included 167 patients in this retrospective cross-sectional study, 123 patients with an objective cognitive decline [mild cognitive impairment (MCI) due to AD, n = 63, and AD-dementia, n = 60] and 44 age-matched healthy controls (HC). Cerebrospinal fluid (CSF) and plasma concentrations of NfL and GFAP were measured with single molecule array (SIMOA®) technology using the Neurology 2-Plex B kit from Quanterix. To assess the discriminatory potential of different biomarkers, age- and sex-adjusted receiver operating characteristic (ROC) curves were calculated and the area under the curve (AUC) of each model was compared. Results: We constructed a panel combining plasma NfL and GFAP with known AD risk factors (Combination panel: age+sex+APOE4+GFAP+NfL). With an AUC of 91.6% (95%CI = 0.85-0.98) for HC vs. AD and 81.7% (95%CI = 0.73-0.90) for HC vs. MCI as well as an AUC of 87.5% (95%CI = 0.73-0.96) in terms of predicting amyloid positivity, this panel showed a promising discriminatory power to differentiate these populations. Conclusion: The combination of plasma GFAP and NfL with well-established risk factors discerns amyloid positive from negative patients and could potentially be applied to identify patients who would benefit from a more invasive assessment of amyloid pathology. In the future, improved prediction of amyloid positivity with a noninvasive test may decrease the number and costs of a more invasive or expensive diagnostic approach.
Collapse
Affiliation(s)
- Tandis Parvizi
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Theresa König
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Raphael Wurm
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Sara Silvaieh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Patrick Altmann
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Sigrid Klotz
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Julia Furtner
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Günther Regelsberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Johann Lehrner
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Tatjana Traub-Weidinger
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, University of Vienna, Vienna, Austria
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
13
|
Mank A, van Maurik IS, Rijnhart JJM, bakker ED, Bouteloup V, Le Scouarnec L, Teunissen CE, Barkhof F, Scheltens P, Berkhof J, van der Flier WM. Development of multivariable prediction models for institutionalization and mortality in the full spectrum of Alzheimer’s disease. Alzheimers Res Ther 2022; 14:110. [PMID: 35932034 PMCID: PMC9354423 DOI: 10.1186/s13195-022-01053-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022]
Abstract
Background Patients and caregivers express a desire for accurate prognostic information about time to institutionalization and mortality. Previous studies predicting institutionalization and mortality focused on the dementia stage. However, Alzheimer’s disease (AD) is characterized by a long pre-dementia stage. Therefore, we developed prediction models to predict institutionalization and mortality along the AD continuum of cognitively normal to dementia. Methods This study included SCD/MCI patients (subjective cognitive decline (SCD) or mild cognitive impairment (MCI)) and patients with AD dementia from the Amsterdam Dementia Cohort. We developed internally and externally validated prediction models with biomarkers and without biomarkers, stratified by dementia status. Determinants were selected using backward selection (p<0.10). All models included age and sex. Discriminative performance of the models was assessed with Harrell’s C statistics. Results We included n=1418 SCD/MCI patients (n=123 died, n=74 were institutionalized) and n=1179 patients with AD dementia (n=413 died, n=453 were institutionalized). For both SCD/MCI and dementia stages, the models for institutionalization and mortality included after backward selection clinical characteristics, imaging, and cerebrospinal fluid (CSF) biomarkers. In SCD/MCI, the Harrell’s C-statistics of the models were 0.81 (model without biomarkers: 0.76) for institutionalization and 0.79 (model without biomarker: 0.76) for mortality. In AD-dementia, the Harrell’s C-statistics of the models were 0.68 (model without biomarkers: 0.67) for institutionalization and 0.65 (model without biomarker: 0.65) for mortality. Models based on data from amyloid-positive patients only had similar discrimination. Conclusions We constructed prediction models to predict institutionalization and mortality with good accuracy for SCD/MCI patients and moderate accuracy for patients with AD dementia. The developed prediction models can be used to provide patients and their caregivers with prognostic information on time to institutionalization and mortality along the cognitive continuum of AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01053-0.
Collapse
|
14
|
Ebenau JL, Pelkmans W, Verberk IMW, Verfaillie SCJ, van den Bosch KA, van Leeuwenstijn M, Collij LE, Scheltens P, Prins ND, Barkhof F, van Berckel BNM, Teunissen CE, van der Flier WM. Association of CSF, Plasma, and Imaging Markers of Neurodegeneration With Clinical Progression in People With Subjective Cognitive Decline. Neurology 2022; 98:e1315-e1326. [PMID: 35110378 PMCID: PMC8967429 DOI: 10.1212/wnl.0000000000200035] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Multiple biomarkers have been suggested to measure neurodegeneration (N) in the AT(N) framework, leading to inconsistencies between studies. We investigated the association of 5 N biomarkers with clinical progression and cognitive decline in individuals with subjective cognitive decline (SCD). METHODS We included individuals with SCD from the Amsterdam Dementia Cohort and SCIENCe project, a longitudinal cohort study (follow-up 4±3 years). We used the following N biomarkers: CSF total tau (t-tau), medial temporal atrophy visual rating on MRI, hippocampal volume (HV), serum neurofilament light (NfL), and serum glial fibrillary acidic protein (GFAP). We determined correlations between biomarkers. We assessed associations between N biomarkers and clinical progression to mild cognitive impairment or dementia (Cox regression) and Mini-Mental State Examination (MMSE) over time (linear mixed models). Models included age, sex, CSF β-amyloid (Aβ) (A), and CSF p-tau (T) as covariates, in addition to the N biomarker. RESULT We included 401 individuals (61±9 years, 42% female, MMSE 28 ± 2, vascular comorbidities 8%-19%). N biomarkers were modestly to moderately correlated (range r -0.28 - 0.58). Serum NfL and GFAP correlated most strongly (r 0.58, p < 0.01). T-tau was strongly correlated with p-tau (r 0.89, p < 0.01), although these biomarkers supposedly represent separate biomarker groups. All N biomarkers individually predicted clinical progression, but only HV, NfL, and GFAP added predictive value beyond Aβ and p-tau (hazard ratio 1.52 [95% CI 1.11-2.09]; 1.51 [1.05-2.17]; 1.50 [1.04-2.15]). T-tau, HV, and GFAP individually predicted MMSE slope (range β -0.17 to -0.11, p < 0.05), but only HV remained associated beyond Aβ and p-tau (β -0.13 [SE 0.04]; p < 0.05). DISCUSSION In cognitively unimpaired older adults, correlations between different N biomarkers were only moderate, indicating they reflect different aspects of neurodegeneration and should not be used interchangeably. T-tau was strongly associated with p-tau (T), which makes it less desirable to use as a measure for N. HV, NfL, and GFAP predicted clinical progression beyond A and T. Our results do not allow to choose one most suitable biomarker for N, but illustrate the added prognostic value of N beyond A and T. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that HV, NfL, and GFAP predicted clinical progression beyond A and T in individuals with SCD.
Collapse
Affiliation(s)
- Jarith L Ebenau
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK.
| | - Wiesje Pelkmans
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Inge M W Verberk
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Sander C J Verfaillie
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Karlijn A van den Bosch
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Mardou van Leeuwenstijn
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Lyduine E Collij
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Philip Scheltens
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Niels D Prins
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Frederik Barkhof
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Bart N M van Berckel
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Charlotte E Teunissen
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Wiesje M van der Flier
- From the Alzheimer Center, Departments of Neurology (J.L.E., W.P., I.M.W.V., K.A.v.d.B., M.v.L., P.S., N.D.P., B.N.M.v.B., W.M.V.d.F.) and Radiology & Nuclear Medicine (S.C.J.V., L.E.C., F.B., B.N.M.v.B.), Amsterdam Neuroscience, and Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| |
Collapse
|
15
|
The approval of a disease-modifying treatment for Alzheimer's disease: impact and consequences for the nuclear medicine community. Eur J Nucl Med Mol Imaging 2021; 48:3033-3036. [PMID: 34272989 DOI: 10.1007/s00259-021-05485-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
16
|
Willemse EAJ, Scheltens P, Teunissen CE, Vijverberg EGB. A neurologist's perspective on serum neurofilament light in the memory clinic: a prospective implementation study. ALZHEIMERS RESEARCH & THERAPY 2021; 13:101. [PMID: 34006321 PMCID: PMC8132439 DOI: 10.1186/s13195-021-00841-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/04/2021] [Indexed: 11/10/2022]
Abstract
Background Neurofilament light in serum (sNfL) is a biomarker for axonal damage with elevated levels in many neurological disorders, including neurodegenerative dementias. Since within-group variation of sNfL is large and concentrations increase with aging, sNfL’s clinical use in memory clinic practice remains to be established. The objective of the current study was to evaluate the clinical use of serum neurofilament light (sNfL), a cross-disease biomarker for axonal damage, in a tertiary memory clinic cohort. Methods Six neurologists completed questionnaires regarding the usefulness of sNfL (n = 5–42 questionnaires/neurologist). Patients that visited the Alzheimer Center Amsterdam for the first time between May and October 2019 (n = 109) were prospectively included in this single-center implementation study. SNfL levels were analyzed on Simoa and reported together with normal values in relation to age, as part of routine diagnostic work-up and in addition to cerebrospinal fluid (CSF) biomarker analysis. Results SNfL was perceived as useful in 53% (n = 58) of the cases. SNfL was more often perceived as useful in patients < 62 years (29/48, 60%, p = 0.05) and males (41/65, 63%, p < 0.01). Availability of CSF biomarker results at time of result discussion had no influence. We observed non-significant trends for increased perceived usefulness of sNfL for patients with the diagnosis subjective cognitive decline (64%), psychiatric disorder (71%), or uncertain diagnosis (67%). SNfL was mostly helpful to neurologists in confirming or excluding neurodegeneration. Whether sNfL was regarded as useful strongly depended on which neurologist filled out the questionnaire (ranging from 0 to 73% of useful cases/neurologist). Discussion Regardless of the availability of CSF biomarker results, sNfL was perceived as a useful tool in more than half of the evaluated cases in a tertiary memory clinic practice. Based on our results, we recommend the analysis of the biomarker sNfL to confirm or exclude neurodegeneration in patients below 62 years old and in males. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00841-4.
Collapse
Affiliation(s)
- E A J Willemse
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Center, Vrije Universiteit, De Boelelaan, 1117, Amsterdam, The Netherlands.
| | - P Scheltens
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, Vrije Universiteit, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - C E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Center, Vrije Universiteit, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - E G B Vijverberg
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, Vrije Universiteit, De Boelelaan, 1117, Amsterdam, The Netherlands.,Brain Research Center, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Ossenkoppele R, Hansson O. Towards clinical application of tau PET tracers for diagnosing dementia due to Alzheimer's disease. Alzheimers Dement 2021; 17:1998-2008. [PMID: 33984177 DOI: 10.1002/alz.12356] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/22/2021] [Accepted: 03/28/2021] [Indexed: 11/07/2022]
Abstract
The recent development of several tau positron emission tomography (PET) tracers represents a major milestone for the Alzheimer's disease (AD) field. These tau PET tracers bind tau neurofibrillary tangles, a key neuropathological characteristic of AD that is tightly linked to synaptic loss, brain atrophy, and cognitive decline. It is notable that these tau PET tracers show low uptake in most non-AD tauopathies and other neurodegenerative disorders, resulting in a diagnostic specificity that is superior to that of amyloid beta (Aβ) PET and biofluid markers, especially at an older age when incidental Aβ pathology is common. Furthermore, tau PET tracers diagnostically outperform widely used MRI markers. Given its excellent diagnostic performance due to the combination of high sensitivity and specificity for detecting tau pathology in AD dementia, we hypothesize that tau PET can become an important diagnostic tool in specialized clinics for the differential diagnosis of dementia syndromes where AD is among the major possible underlying diseases.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Lund University, Clinical Memory Research Unit, Lund, Sweden.,Department of Neurology, Amsterdam Neuroscience, Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Oskar Hansson
- Lund University, Clinical Memory Research Unit, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
18
|
Rosenberg A, Solomon A, Soininen H, Visser PJ, Blennow K, Hartmann T, Kivipelto M. Research diagnostic criteria for Alzheimer's disease: findings from the LipiDiDiet randomized controlled trial. ALZHEIMERS RESEARCH & THERAPY 2021; 13:64. [PMID: 33766132 PMCID: PMC7995792 DOI: 10.1186/s13195-021-00799-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/23/2021] [Indexed: 12/18/2022]
Abstract
Background To explore the utility of the International Working Group (IWG)-1 criteria in recruitment for Alzheimer’s disease (AD) clinical trials, we applied the more recently proposed research diagnostic criteria to individuals enrolled in a randomized controlled prevention trial (RCT) and assessed their disease progression. Methods The multinational LipiDiDiet RCT targeted 311 individuals with IWG-1 defined prodromal AD. Based on centrally analyzed baseline biomarkers, participants were classified according to the IWG-2 and National Institute on Aging–Alzheimer’s Association (NIA-AA) 2011 and 2018 criteria. Linear mixed models were used to investigate the 2-year change in cognitive and functional performance (Neuropsychological Test Battery NTB Z scores, Clinical Dementia Rating-Sum of Boxes CDR-SB) (criteria × time interactions; baseline score, randomization group, sex, Mini-Mental State Examination (MMSE), and age also included in the models). Cox models adjusted for randomization group, MMSE, sex, age, and study site were used to investigate the risk of progression to dementia over 2 years. Results In total, 88%, 86%, and 69% of participants had abnormal cerebrospinal fluid (CSF) β-amyloid, total tau, and phosphorylated tau, respectively; 64% had an A+T+N+ profile (CSF available for N = 107). Cognitive-functional decline appeared to be more pronounced in the IWG-2 prodromal AD, NIA-AA 2011 high and intermediate AD likelihood, and NIA-AA 2018 AD groups, but few significant differences were observed between the groups within each set of criteria. Hazard ratio (95% CI) for dementia was 4.6 (1.6–13.7) for IWG-2 prodromal AD (reference group no prodromal AD), 7.4 (1.0–54.7) for NIA-AA 2011 high AD likelihood (reference group suspected non-AD pathology SNAP), and 9.4 (1.2–72.7) for NIA-AA 2018 AD (reference group non-Alzheimer’s pathologic change). Compared with the NIA-AA 2011 high AD likelihood group (abnormal β-amyloid and neuronal injury markers), disease progression was similar in the intermediate AD likelihood group (medial temporal lobe atrophy; no CSF available). Conclusions Despite being less restrictive than the other criteria, the IWG-1 criteria reliably identified individuals with AD pathology. More pragmatic and easily applicable selection criteria might be preferred due to feasibility in certain situations, e.g., in multidomain prevention trials that do not specifically target β-amyloid/tau pathologies. Trial registration Netherlands Trial Register, NL1620. Registered on 9 March 2009
Collapse
Affiliation(s)
- Anna Rosenberg
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.
| | - Alina Solomon
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Hilkka Soininen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Neurocenter, Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centre Limburg, University of Maastricht, Maastricht, Netherlands.,Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience, VU University Medical Centre, Amsterdam, Netherlands
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Tobias Hartmann
- Deutsches Institut für Demenz Prävention (DIDP), Medical Faculty, and Department of Experimental Neurology, Saarland University, Homburg, Germany
| | - Miia Kivipelto
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | | |
Collapse
|
19
|
Gjerum L, Andersen BB, Bruun M, Simonsen AH, Henriksen OM, Law I, Hasselbalch SG, Frederiksen KS. Comparison of the clinical impact of 2-[18F]FDG-PET and cerebrospinal fluid biomarkers in patients suspected of Alzheimer's disease. PLoS One 2021; 16:e0248413. [PMID: 33711065 PMCID: PMC7954298 DOI: 10.1371/journal.pone.0248413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/26/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The two biomarkers 2-[18F]FDG-PET and cerebrospinal fluid biomarkers are both recommended to support the diagnosis of Alzheimer's disease. However, there is a lack of knowledge for the comparison of the two biomarkers in a routine clinical setting. OBJECTIVE The aim was to compare the clinical impact of 2-[18F]FDG-PET and cerebrospinal fluid biomarkers on diagnosis, prognosis, and patient management in patients suspected of Alzheimer's disease. METHODS Eighty-one patients clinically suspected of Alzheimer's disease were retrospectively included from the Copenhagen Memory Clinic. As part of the clinical work-up all patients had a standard diagnostic program examination including MRI and ancillary investigations with 2-[18F]FDG-PET and cerebrospinal fluid biomarkers. An incremental study design was used to evaluate the clinical impact of the biomarkers. First, the diagnostic evaluation was based on the standard diagnostic program, then the diagnostic evaluation was revised after addition of either cerebrospinal fluid biomarkers or 2-[18F]FDG-PET. At each diagnostic evaluation, two blinded dementia specialists made a consensus decision on diagnosis, prediction of disease course, and change in patient management. Confidence in the decision was measured on a visual analogue scale (0-100). After 6 months, the diagnostic evaluation was performed with addition of the other biomarker. A clinical follow-up after 12 months was used as reference for diagnosis and disease course. RESULTS The two biomarkers had a similar clinical value across all diagnosis when added individually to the standard diagnostic program. However, for the correctly diagnosed patient with Alzheimer's disease cerebrospinal fluid biomarkers had a significantly higher impact on diagnostic confidence (mean scores±SD: 88±11 vs. 82±11, p = 0.046) and a significant reduction in the need for ancillary investigations (23 vs. 18 patients, p = 0.049) compared to 2-[18F]FDG-PET. CONCLUSION The two biomarkers had similar clinical impact on diagnosis, but cerebrospinal fluid biomarkers had a more significant value in corroborating the diagnosis of Alzheimer's disease compared to 2-[18F]FDG-PET.
Collapse
Affiliation(s)
- Le Gjerum
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Bo Andersen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Marie Bruun
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anja Hviid Simonsen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Otto Mølby Henriksen
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Steen Frederiksen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
2020 update on the clinical validity of cerebrospinal fluid amyloid, tau, and phospho-tau as biomarkers for Alzheimer's disease in the context of a structured 5-phase development framework. Eur J Nucl Med Mol Imaging 2021; 48:2121-2139. [PMID: 33674895 PMCID: PMC8175301 DOI: 10.1007/s00259-021-05258-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022]
Abstract
Purpose In the last decade, the research community has focused on defining reliable biomarkers for the early detection of Alzheimer’s disease (AD) pathology. In 2017, the Geneva AD Biomarker Roadmap Initiative adapted a framework for the systematic validation of oncological biomarkers to cerebrospinal fluid (CSF) AD biomarkers—encompassing the 42 amino-acid isoform of amyloid-β (Aβ42), phosphorylated-tau (P-tau), and Total-tau (T-tau)—with the aim to accelerate their development and clinical implementation. The aim of this work is to update the current validation status of CSF AD biomarkers based on the Biomarker Roadmap methodology. Methods A panel of experts in AD biomarkers convened in November 2019 at a 2-day workshop in Geneva. The level of maturity (fully achieved, partly achieved, preliminary evidence, not achieved, unsuccessful) of CSF AD biomarkers was assessed based on the Biomarker Roadmap methodology before the meeting and presented and discussed during the workshop. Results By comparison to the previous 2017 Geneva Roadmap meeting, the primary advances in CSF AD biomarkers have been in the area of a unified protocol for CSF sampling, handling and storage, the introduction of certified reference methods and materials for Aβ42, and the introduction of fully automated assays. Additional advances have occurred in the form of defining thresholds for biomarker positivity and assessing the impact of covariates on their discriminatory ability. Conclusions Though much has been achieved for phases one through three, much work remains in phases four (real world performance) and five (assessment of impact/cost). To a large degree, this will depend on the availability of disease-modifying treatments for AD, given these will make accurate and generally available diagnostic tools key to initiate therapy. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05258-7.
Collapse
|
21
|
Ye LQ, Gao PR, Zhang YB, Cheng HR, Tao QQ, Wu ZY, Li HL. Application of Cerebrospinal Fluid AT(N) Framework on the Diagnosis of AD and Related Cognitive Disorders in Chinese Han Population. Clin Interv Aging 2021; 16:311-323. [PMID: 33654388 PMCID: PMC7910151 DOI: 10.2147/cia.s294756] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/21/2021] [Indexed: 11/23/2022] Open
Abstract
Background Studies concerning the impact of the AT(N) framework on diagnostic capability in the dementia population are lacking. We aimed to explore the diagnostic application of CSF AT(N) framework in clinical routines of Alzheimer's disease (AD) as well as differential diagnosis of other cognitive diseases in the Chinese Han population. Patients and Methods A total of 137 patients with cognitive disorders received CSF tests of Aβ42, t-tau and p-tau181. Their CSF biomarker results were categorized and interpreted by the AT(N) framework. Neurologists provided a diagnosis both pre- and post-CSF biomarker disclosure with corresponding diagnostic confidence. Results The total initial diagnosis included 79 patients with AD and 58 patients with non-AD (NAD). The results of CSF biomarkers led to a diagnostic change of 28% in the cohort. Approximately 81.5% (n=53) of 65 patients whose CSF biomarker showed an underlying AD pathology were finally diagnosed as AD, with an increase of 17.5% in diagnostic confidence. Thirty-seven CSF results indicating NAD pathologic changes contributed to an exclusion of AD in 56.8% (n=21) of the patients along with a modest increase of 9.8% in average confidence. Thirty-five patients with normal CSF biomarkers maintained the diagnosis of NAD in 68.6% (n=24) of the group, leading to a slight elevation of 7.6% in confidence. Conclusion We found that the presence of amyloid pathology (A+) is contributable to diagnosing AD and improving confidence. On occasion of negative amyloid pathology (A-), with or without tau pathology, gaining uncertainty of the primary AD diagnosis would diminish the corresponding confidence. To the best of our knowledge, this is the first study performed in the Chinese Han population with cognitive disorders that explores the clinical capability of CSF AT(N) framework in a quantitative way.
Collapse
Affiliation(s)
- Ling-Qi Ye
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Rehabilitation Medicine and Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, People's Republic of China
| | - Pei-Rong Gao
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yan-Bin Zhang
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Neurology and Institute of Neurology in First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Hong-Rong Cheng
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Department of Neurology in Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, People's Republic of China
| | - Qing-Qing Tao
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Hong-Lei Li
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
22
|
Brisson M, Brodeur C, Létourneau‐Guillon L, Masellis M, Stoessl J, Tamm A, Zukotynski K, Ismail Z, Gauthier S, Rosa‐Neto P, Soucy J. CCCDTD5: Clinical role of neuroimaging and liquid biomarkers in patients with cognitive impairment. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 6:e12098. [PMID: 33532543 PMCID: PMC7821956 DOI: 10.1002/trc2.12098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 09/11/2020] [Indexed: 04/21/2023]
Abstract
Since 1989, four Canadian Consensus Conferences on the Diagnosis and Treatment of Dementia (CCCDTDs) have provided evidence-based dementia diagnostic and treatment guidelines for Canadian clinicians and researchers. We present the results from the Neuroimaging and Fluid Biomarkers Group of the 5th CCCDTD (CCCDTD5), which addressed topics chosen by the steering committee to reflect advances in the field and build on our previous guidelines. Recommendations on Imaging and Fluid Biomarker Use from this Conference cover a series of different fields. Prior structural imaging recommendations for both computerized tomography (CT) and magnetic resonance imaging (MRI) remain largely unchanged, but MRI is now more central to the evaluation than before, with suggested sequences described here. The use of visual rating scales for both atrophy and white matter anomalies is now included in our recommendations. Molecular imaging with [18F]-fluorodeoxyglucose ([18F]-FDG) Positron Emisson Tomography (PET) or [99mTc]-hexamethylpropyleneamine oxime/ethylene cysteinate dimer ([99mTc]-HMPAO/ECD) Single Photon Emission Tomography (SPECT), should now decidedly favor PET. The value of [18F]-FDG PET in the assessment of neurodegenerative conditions has been established with greater certainty since the previous conference, and it has now been recognized as a useful biomarker to establish the presence of neurodegeneration by a number of professional organizations around the world. Furthermore, the role of amyloid PET has been clarified and our recommendations follow those from other groups in multiple countries. SPECT with [123I]-ioflupane (DaTscanTM) is now included as a useful study in differentiating Alzheimer's disease (AD) from Lewy body disease. Finally, liquid biomarkers are in a rapid phase of development and, could lead to a revolution in the assessment AD and other neurodegenerative conditions at a reasonable cost. We hope these guidelines will be useful for clinicians, researchers, policy makers, and the lay public, to inform a current and evidence-based approach to the use of neuroimaging and liquid biomarkers in clinical dementia evaluation and management.
Collapse
Affiliation(s)
- Mélanie Brisson
- Centre hospitalier de l'université de QuébecQuebec CityCanada
| | | | | | | | - Jon Stoessl
- Vancouver Coastal Health, University of British‐ColumbiaVancouverCanada
| | | | | | - Zahinoor Ismail
- Department of Psychiatry, Hotchkiss Brain Institute and O'Brien Institute for Public HealthUniversity of CalgaryCalgaryCanada
| | | | - Pedro Rosa‐Neto
- McGill Center for Studies in AgingCanada
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
| | - Jean‐Paul Soucy
- Centre hospitalier de l'université de MontréalMontrealCanada
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMontrealCanada
- PERFORM Center, Concordia UniversityMontrealCanada
| |
Collapse
|
23
|
van Maurik IS, Rhodius-Meester HFM, Teunissen CE, Scheltens P, Barkhof F, Palmqvist S, Hansson O, van der Flier WM, Berkhof J. Biomarker testing in MCI patients-deciding who to test. ALZHEIMERS RESEARCH & THERAPY 2021; 13:14. [PMID: 33413634 PMCID: PMC7792312 DOI: 10.1186/s13195-020-00763-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/23/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND We aimed to derive an algorithm to define the optimal proportion of patients with mild cognitive impairment (MCI) in whom cerebrospinal fluid (CSF) testing is of added prognostic value. METHODS MCI patients were selected from the Amsterdam Dementia Cohort (n = 402). Three-year progression probabilities to dementia were predicted using previously published models with and without CSF data (amyloid-beta1-42 (Abeta), phosphorylated tau (p-tau)). We incrementally augmented the proportion of patients undergoing CSF, starting with the 10% patients with prognostic probabilities based on clinical data around the median (percentile 45-55), until all patients received CSF. The optimal proportion was defined as the proportion where the stepwise algorithm showed similar prognostic discrimination (Harrell's C) and accuracy (three-year Brier scores) compared to CSF testing of all patients. We used the BioFINDER study (n = 221) for validation. RESULTS The optimal proportion of MCI patients to receive CSF testing selected by the stepwise approach was 50%. CSF testing in only this proportion improved the performance of the model with clinical data only from Harrell's C = 0.60, Brier = 0.198 (Harrell's C = 0.61, Brier = 0.197 if the information on magnetic resonance imaging was available) to Harrell's C = 0.67 and Brier = 0.190, and performed similarly to a model in which all patients received CSF testing. Applying the stepwise approach in the BioFINDER study would again select half of the MCI patients and yielded robust results with respect to prognostic performance. INTERPRETATION CSF biomarker testing adds prognostic value in half of the MCI patients. As such, we achieve a CSF saving recommendation while simultaneously retaining optimal prognostic accuracy.
Collapse
Affiliation(s)
- Ingrid S van Maurik
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands. .,Department of Epidemiology and Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Hanneke F M Rhodius-Meester
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.,Department of Internal Medicine, Geriatric Medicine Section, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Institutes of Neurology and Healthcare Engineering, University College London, London, England
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.,Department of Epidemiology and Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Johannes Berkhof
- Department of Epidemiology and Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Chételat G, Arbizu J, Barthel H, Garibotto V, Law I, Morbelli S, van de Giessen E, Agosta F, Barkhof F, Brooks DJ, Carrillo MC, Dubois B, Fjell AM, Frisoni GB, Hansson O, Herholz K, Hutton BF, Jack CR, Lammertsma AA, Landau SM, Minoshima S, Nobili F, Nordberg A, Ossenkoppele R, Oyen WJG, Perani D, Rabinovici GD, Scheltens P, Villemagne VL, Zetterberg H, Drzezga A. Amyloid-PET and 18F-FDG-PET in the diagnostic investigation of Alzheimer's disease and other dementias. Lancet Neurol 2020; 19:951-962. [PMID: 33098804 DOI: 10.1016/s1474-4422(20)30314-8] [Citation(s) in RCA: 278] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 07/22/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
Abstract
Various biomarkers are available to support the diagnosis of neurodegenerative diseases in clinical and research settings. Among the molecular imaging biomarkers, amyloid-PET, which assesses brain amyloid deposition, and 18F-fluorodeoxyglucose (18F-FDG) PET, which assesses glucose metabolism, provide valuable and complementary information. However, uncertainty remains regarding the optimal timepoint, combination, and an order in which these PET biomarkers should be used in diagnostic evaluations because conclusive evidence is missing. Following an expert panel discussion, we reached an agreement on the specific use of the individual biomarkers, based on available evidence and clinical expertise. We propose a diagnostic algorithm with optimal timepoints for these PET biomarkers, also taking into account evidence from other biomarkers, for early and differential diagnosis of neurodegenerative diseases that can lead to dementia. We propose three main diagnostic pathways with distinct biomarker sequences, in which amyloid-PET and 18F-FDG-PET are placed at different positions in the order of diagnostic evaluations, depending on clinical presentation. We hope that this algorithm can support diagnostic decision making in specialist clinical settings with access to these biomarkers and might stimulate further research towards optimal diagnostic strategies.
Collapse
Affiliation(s)
- Gaël Chételat
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237, Groupement d'Intérêt Public Cyceron, Caen, France.
| | - Javier Arbizu
- Department of Nuclear Medicine, University of Navarra, Clinica Universidad de Navarra, Pamplona, Spain
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and NIMTlab, Geneva University, Geneva, Switzerland
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Silvia Morbelli
- Nuclear Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genova, Italy
| | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Institutes of Neurology and Healthcare Engineering, University College London, London, UK
| | - David J Brooks
- Institute of Neuroscience, Newcastle University, Newcastle, UK; Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | | | - Bruno Dubois
- Centre des Maladies Cognitives et Comportementales, University Hospital of Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne-Université, Paris, France
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway, Oslo; Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Giovanni B Frisoni
- Memory Clinic, Department of Rehabilitation and Geriatrics, Geneva University and University Hospitals, Geneva, Switzerland
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Brian F Hutton
- Institute of Nuclear Medicine, University College London, London, UK
| | | | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Flavio Nobili
- UO Clinica Neurologica, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genova, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Child and Mother Health, University of Genoa, Genova, Italy
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Rik Ossenkoppele
- Department of Neurology, Alzheimer Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Wim J G Oyen
- Humanitas University and Humanitas Clinical and Research Center, Department of Nuclear Medicine, Milan, Italy; Rijnstate, Department of Radiology and Nuclear Medicine, Arnhem, Netherlands; Radboud UMC, Department of Radiology and Nuclear Medicine, Nijmegen, Netherlands
| | - Daniela Perani
- Vita-Salute San Raffaele University, Nuclear Medicine Unit, San Raffaele Hospital, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Gil D Rabinovici
- Departments of Neurology, Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC, Australia; School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK; 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; UK Dementia Research Institute at University College London, London, UK
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; German Center for Neurodegenerative Diseases, Bonn-Cologne, Germany; Institute of Neuroscience and Medicine, Molecular Organization of the Brain, Forschungszentrum Jülich, Germany
| |
Collapse
|
25
|
Behfar Q, Behfar SK, von Reutern B, Richter N, Dronse J, Fassbender R, Fink GR, Onur OA. Graph Theory Analysis Reveals Resting-State Compensatory Mechanisms in Healthy Aging and Prodromal Alzheimer's Disease. Front Aging Neurosci 2020; 12:576627. [PMID: 33192468 PMCID: PMC7642892 DOI: 10.3389/fnagi.2020.576627] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/29/2020] [Indexed: 01/20/2023] Open
Abstract
Several theories of cognitive compensation have been suggested to explain sustained cognitive abilities in healthy brain aging and early neurodegenerative processes. The growing number of studies investigating various aspects of task-based compensation in these conditions is contrasted by the shortage of data about resting-state compensatory mechanisms. Using our proposed criterion-based framework for compensation, we investigated 45 participants in three groups: (i) patients with mild cognitive impairment (MCI) and positive biomarkers indicative of Alzheimer's disease (AD); (ii) cognitively normal young adults; (iii) cognitively normal older adults. To increase reliability, three sessions of resting-state functional magnetic resonance imaging for each participant were performed on different days (135 scans in total). To elucidate the dimensions and dynamics of resting-state compensatory mechanisms, we used graph theory analysis along with volumetric analysis. Graph theory analysis was applied based on the Brainnetome atlas, which provides a connectivity-based parcellation framework. Comprehensive neuropsychological examinations including the Rey Auditory Verbal Learning Test (RAVLT) and the Trail Making Test (TMT) were performed, to relate graph measures of compensatory nodes to cognition. To avoid false-positive findings, results were corrected for multiple comparisons. First, we observed an increase of degree centrality in cognition related brain regions of the middle frontal gyrus, precentral gyrus and superior parietal lobe despite local atrophy in MCI and healthy aging, indicating a resting-state connectivity increase with positive biomarkers. When relating the degree centrality measures to cognitive performance, we observed that greater connectivity led to better RAVLT and TMT scores in MCI and, hence, might constitute a compensatory mechanism. The detection and improved understanding of the compensatory dynamics in healthy aging and prodromal AD is mandatory for implementing and tailoring preventive interventions aiming at preserved overall cognitive functioning and delayed clinical onset of dementia.
Collapse
Affiliation(s)
- Qumars Behfar
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Research Centre Jülich, Institute of Neuroscience and Medicine (INM-3), Jülich, Germany
| | - Stefan Kambiz Behfar
- Laboratory for Innovation Science at Harvard (LISH), Harvard University, Cambridge, MA, United States
| | - Boris von Reutern
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nils Richter
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Research Centre Jülich, Institute of Neuroscience and Medicine (INM-3), Jülich, Germany
| | - Julian Dronse
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Research Centre Jülich, Institute of Neuroscience and Medicine (INM-3), Jülich, Germany
| | - Ronja Fassbender
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Research Centre Jülich, Institute of Neuroscience and Medicine (INM-3), Jülich, Germany
| | - Oezguer A Onur
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Research Centre Jülich, Institute of Neuroscience and Medicine (INM-3), Jülich, Germany
| |
Collapse
|
26
|
Diagnostic Impact of Cerebrospinal Fluid Biomarkers in Atypical Dementias in Canada. Can J Neurol Sci 2020; 48:312-320. [PMID: 32892784 DOI: 10.1017/cjn.2020.196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND In Canada, standard dementia workup consists of clinical, neurological, and cognitive evaluation, as well as structural brain imaging. For atypical dementia presentations, additional FDG-PET brain imaging is recommended. Cerebrospinal fluid (CSF) biomarkers have recently been proposed as the gold standard for in vivo detection of Alzheimer's disease (AD) pathophysiology (NIA-AA research framework, 2018). As clinical implementation of CSF assessment is still limited in Canada, the present study assessed its impact on diagnostic accuracy in atypical neurodegenerative disorders in the clinical practice. METHODS This retrospective clinical chart review included patients with cognitive complaints who underwent lumbar puncture (LP) in addition to the standard diagnostic workup. CSF analysis determined the presence of biological AD based on reduced amyloid-β42-to-total-tau index (ATI) and increased phosphorylated-tau (p-tau) levels. CSF-based diagnoses were compared to standard workup and FDG-PET-based diagnoses. RESULTS A total of 28 patients with atypical dementia presentations were included in the present study after evaluation for cognitive complaints at a specialized dementia clinic between November 2017 and July 2019. CSF analysis changed or better specified the initial clinical diagnosis in 43.0% of cases (alternative diagnosis revealed in 25% and excluded in 18%). In patients with additional FDG-PET imaging (n = 23), FDG-PET and CSF-based diagnosis did not correspond in 35% of patients, even though FDG-PET appeared to increase diagnostic accuracy compared to the initial clinical diagnosis. CONCLUSION CSF biomarkers improved diagnostic accuracy in atypical cognitively-impaired patients beyond standard workup and FDG-PET imaging. These results support CSF analysis implementation for atypical dementias in Canada, in addition to the standard diagnostic workup.
Collapse
|
27
|
Ebenau JL, Timmers T, Wesselman LMP, Verberk IMW, Verfaillie SCJ, Slot RER, van Harten AC, Teunissen CE, Barkhof F, van den Bosch KA, van Leeuwenstijn M, Tomassen J, Braber AD, Visser PJ, Prins ND, Sikkes SAM, Scheltens P, van Berckel BNM, van der Flier WM. ATN classification and clinical progression in subjective cognitive decline: The SCIENCe project. Neurology 2020; 95:e46-e58. [PMID: 32522798 PMCID: PMC7371376 DOI: 10.1212/wnl.0000000000009724] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Objective To investigate the relationship between the ATN classification system (amyloid, tau, neurodegeneration) and risk of dementia and cognitive decline in individuals with subjective cognitive decline (SCD). Methods We classified 693 participants with SCD (60 ± 9 years, 41% women, Mini-Mental State Examination score 28 ± 2) from the Amsterdam Dementia Cohort and Subjective Cognitive Impairment Cohort (SCIENCe) project according to the ATN model, as determined by amyloid PET or CSF β-amyloid (A), CSF p-tau (T), and MRI-based medial temporal lobe atrophy (N). All underwent extensive neuropsychological assessment. For 342 participants, follow-up was available (3 ± 2 years). As a control population, we included 124 participants without SCD. Results Fifty-six (n = 385) participants had normal Alzheimer disease (AD) biomarkers (A–T–N–), 27% (n = 186) had non-AD pathologic change (A–T–N+, A–T+N–, A–T+N+), 18% (n = 122) fell within the Alzheimer continuum (A+T–N–, A+T–N+, A+T+N–, A+T+N+). ATN profiles were unevenly distributed, with A–T+N+, A+T–N+, and A+T+N+ containing very few participants. Cox regression showed that compared to A–T–N–, participants in A+ profiles had a higher risk of dementia with a dose–response pattern for number of biomarkers affected. Linear mixed models showed participants in A+ profiles showed a steeper decline on tests addressing memory, attention, language, and executive functions. In the control group, there was no association between ATN and cognition. Conclusions Among individuals presenting with SCD at a memory clinic, those with a biomarker profile A–T+N+, A+T–N–, A+T+N–, and A+T+N+ were at increased risk of dementia, and showed steeper cognitive decline compared to A–T–N– individuals. These results suggest a future where biomarker results could be used for individualized risk profiling in cognitively normal individuals presenting at a memory clinic.
Collapse
Affiliation(s)
- Jarith L Ebenau
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden.
| | - Tessa Timmers
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Linda M P Wesselman
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Inge M W Verberk
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Sander C J Verfaillie
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Rosalinde E R Slot
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Argonde C van Harten
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Charlotte E Teunissen
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Frederik Barkhof
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Karlijn A van den Bosch
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Mardou van Leeuwenstijn
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Jori Tomassen
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Anouk den Braber
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Pieter Jelle Visser
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Niels D Prins
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Sietske A M Sikkes
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Philip Scheltens
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Bart N M van Berckel
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| | - Wiesje M van der Flier
- From the Alzheimer Center, Department of Neurology (J.L.E., T.T., L.M.P.W., I.M.W.V., R.E.R.S., A.C.v.H., K.A.v.d.B., M.v.L., J.T., A.d.B., P.J.V., N.D.P., S.A.M.S., P.S., B.N.M.v.B., W.M.v.d.F.), and Department of Radiology & Nuclear Medicine (S.C.J.V., F.B., B.N.v.B.), Amsterdam Neuroscience, Neurochemistry Laboratory, Department of Clinical Chemistry (I.M.W.V., C.E.T.), and Department of Epidemiology & Biostatistics (W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK; Department of Biological Psychology (A.d.B.), Neuroscience Amsterdam, VU University Amsterdam; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; and Department of Neurobiology, Care Sciences and Society (P.J.V.), Division of Neurogeriatrics, Karolinska Institutet, Stockholm Sweden
| |
Collapse
|
28
|
Bruun M, Frederiksen KS, Rhodius-Meester HFM, Baroni M, Gjerum L, Koikkalainen J, Urhemaa T, Tolonen A, van Gils M, Tong T, Guerrero R, Rueckert D, Dyremose N, Andersen BB, Simonsen AH, Lemstra A, Hallikainen M, Kurl S, Herukka SK, Remes AM, Waldemar G, Soininen H, Mecocci P, van der Flier WM, Lötjönen J, Hasselbalch SG. Impact of a Clinical Decision Support Tool on Dementia Diagnostics in Memory Clinics: The PredictND Validation Study. Curr Alzheimer Res 2020; 16:91-101. [PMID: 30605060 DOI: 10.2174/1567205016666190103152425] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/04/2018] [Accepted: 12/13/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Determining the underlying etiology of dementia can be challenging. Computer- based Clinical Decision Support Systems (CDSS) have the potential to provide an objective comparison of data and assist clinicians. OBJECTIVES To assess the diagnostic impact of a CDSS, the PredictND tool, for differential diagnosis of dementia in memory clinics. METHODS In this prospective multicenter study, we recruited 779 patients with either subjective cognitive decline (n=252), mild cognitive impairment (n=219) or any type of dementia (n=274) and followed them for minimum 12 months. Based on all available patient baseline data (demographics, neuropsychological tests, cerebrospinal fluid biomarkers, and MRI visual and computed ratings), the PredictND tool provides a comprehensive overview and analysis of the data with a likelihood index for five diagnostic groups; Alzheimer´s disease, vascular dementia, dementia with Lewy bodies, frontotemporal dementia and subjective cognitive decline. At baseline, a clinician defined an etiological diagnosis and confidence in the diagnosis, first without and subsequently with the PredictND tool. The follow-up diagnosis was used as the reference diagnosis. RESULTS In total, 747 patients completed the follow-up visits (53% female, 69±10 years). The etiological diagnosis changed in 13% of all cases when using the PredictND tool, but the diagnostic accuracy did not change significantly. Confidence in the diagnosis, measured by a visual analogue scale (VAS, 0-100%) increased (ΔVAS=3.0%, p<0.0001), especially in correctly changed diagnoses (ΔVAS=7.2%, p=0.0011). CONCLUSION Adding the PredictND tool to the diagnostic evaluation affected the diagnosis and increased clinicians' confidence in the diagnosis indicating that CDSSs could aid clinicians in the differential diagnosis of dementia.
Collapse
Affiliation(s)
- Marie Bruun
- Department of Neurology, Danish Dementia Research Centre, University of Copenhagen, Rigshospitalet, Denmark
| | - Kristian S Frederiksen
- Department of Neurology, Danish Dementia Research Centre, University of Copenhagen, Rigshospitalet, Denmark
| | | | - Marta Baroni
- Institute of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Le Gjerum
- Department of Neurology, Danish Dementia Research Centre, University of Copenhagen, Rigshospitalet, Denmark
| | | | - Timo Urhemaa
- VTT Technical Research Centre of Finland Ltd., Tampere, Finland
| | - Antti Tolonen
- VTT Technical Research Centre of Finland Ltd., Tampere, Finland
| | - Mark van Gils
- VTT Technical Research Centre of Finland Ltd., Tampere, Finland
| | - Tong Tong
- Institutes of Neurology and Healthcare Engineering, University College London, London, United Kingdom
| | - Ricardo Guerrero
- Institutes of Neurology and Healthcare Engineering, University College London, London, United Kingdom
| | - Daniel Rueckert
- Institutes of Neurology and Healthcare Engineering, University College London, London, United Kingdom
| | - Nadia Dyremose
- Department of Neurology, Danish Dementia Research Centre, University of Copenhagen, Rigshospitalet, Denmark
| | - Birgitte Bo Andersen
- Department of Neurology, Danish Dementia Research Centre, University of Copenhagen, Rigshospitalet, Denmark
| | - Anja H Simonsen
- Department of Neurology, Danish Dementia Research Centre, University of Copenhagen, Rigshospitalet, Denmark
| | - Afina Lemstra
- Alzheimer Center, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Merja Hallikainen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Sudhir Kurl
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Anne M Remes
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland and Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland
| | - Gunhild Waldemar
- Department of Neurology, Danish Dementia Research Centre, University of Copenhagen, Rigshospitalet, Denmark
| | - Hilkka Soininen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Patrizia Mecocci
- Institute of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Wiesje M van der Flier
- Alzheimer Center, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands.,Department of Epidemiology and Biostatistics, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | | | - Steen G Hasselbalch
- Department of Neurology, Danish Dementia Research Centre, University of Copenhagen, Rigshospitalet, Denmark
| |
Collapse
|
29
|
Barthélemy NR, Bateman RJ, Hirtz C, Marin P, Becher F, Sato C, Gabelle A, Lehmann S. Cerebrospinal fluid phospho-tau T217 outperforms T181 as a biomarker for the differential diagnosis of Alzheimer's disease and PET amyloid-positive patient identification. Alzheimers Res Ther 2020; 12:26. [PMID: 32183883 PMCID: PMC7079453 DOI: 10.1186/s13195-020-00596-4] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/09/2020] [Indexed: 11/14/2023]
Abstract
BACKGROUND Cerebrospinal fluid biomarker profiles characterized by decreased amyloid-beta peptide levels and increased total and phosphorylated tau levels at threonine 181 (pT181) are currently used to discriminate between Alzheimer's disease and other neurodegenerative diseases. However, these changes are not entirely specific to Alzheimer's disease, and it is noteworthy that other phosphorylated isoforms of tau, possibly more specific for the disease process, have been described in the brain parenchyma of patients. The precise detection of these isoforms in biological fluids remains however a challenge. METHODS In the present study, we used the latest quantitative mass spectrometry approach, which achieves a sensitive detection in cerebrospinal fluid biomarker of two phosphorylated tau isoforms, pT181 and pT217, and first analyzed a cohort of probable Alzheimer's disease patients and patients with other neurological disorders, including tauopathies, and a set of cognitively normal controls. We then checked the validity of our results on a second cohort comprising cognitively normal individuals and patients with mild cognitive impairments and AD stratified in terms of their amyloid status based on PiB-PET imaging methods. RESULTS In the first cohort, pT217 but not pT181 differentiated between Alzheimer's disease patients and those with other neurodegenerative diseases and control subjects much more specificity and sensitivity than pT181. T217 phosphorylation was increased by 6.0-fold in patients with Alzheimer's disease whereas T181 phosphorylation was only increased by 1.3-fold, when compared with control subjects. These results were confirmed in the case of a second cohort, in which the pT217 cerebrospinal fluid levels marked out amyloid-positive patients with a sensitivity and a specificity of more than 90% (AUC 0.961; CI 0.874 to 0.995). The pT217 concentrations were also highly correlated with the PiB-PET values (correlation coefficient 0.72; P < 0.001). CONCLUSIONS Increased cerebrospinal fluid pT217 levels, more than those of pT181, are highly specific biomarkers for detecting both the preclinical and advanced forms of Alzheimer's disease. This finding should greatly improve the diagnosis of Alzheimer's disease, along with the correlations found to exist between pT217 levels and PiB-PET data. It also suggests that pT217 is a promising potential target for therapeutic applications and that a link exists between amyloid and tau pathology.
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Laboratoire de Biochimie Protéomique Clinique, Plateforme de Protéomique Clinique, CHU de Montpellier, INSERM, Université de Montpellier, Montpellier, France.
- Department of Neurology, Washington University School of Medicine, Saint-Louis, MO, USA.
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, Saint-Louis, MO, USA
| | - Christophe Hirtz
- Laboratoire de Biochimie Protéomique Clinique, Plateforme de Protéomique Clinique, CHU de Montpellier, INSERM, Université de Montpellier, Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, CNRS UMR5203, INSERM U1191, Université de Montpellier, Montpellier, France
| | - François Becher
- Service de Pharmacologie et d'Immunoanalyse (SPI), Laboratoire d'Etude du Métabolisme des Médicaments (LEMM), CEA, INRA, Université Paris Saclay, F-91191, Gif-sur-Yvette cedex, France
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, Saint-Louis, MO, USA
| | - Audrey Gabelle
- Memory Resources and Research Center of Montpellier, Department of Neurology, Gui de Chauliac Hospital, Montpellier, France.
| | - Sylvain Lehmann
- Laboratoire de Biochimie Protéomique Clinique, Plateforme de Protéomique Clinique, CHU de Montpellier, INSERM, Université de Montpellier, Montpellier, France.
| |
Collapse
|
30
|
Faura J, Bustamante A, Penalba A, Giralt D, Simats A, Martínez-Sáez E, Alcolea D, Fortea J, Lleó A, Teunissen CE, van der Flier WM, Ibañez L, Harari O, Cruchaga C, Hernández-Guillamón M, Delgado P, Montaner J. CCL23: A Chemokine Associated with Progression from Mild Cognitive Impairment to Alzheimer's Disease. J Alzheimers Dis 2020; 73:1585-1595. [PMID: 31958084 PMCID: PMC8010612 DOI: 10.3233/jad-190753] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
CCL23 is a chemokine implicated in inflammation and host defense responses. It has been recently associated with acquired brain damage and stroke outcomes. In this study, we reported the role of CCL23 in Alzheimer's disease (AD). We evaluated the levels of CCL23 in 659 individuals: cognitively normal, mild cognitive impaired (MCI), and AD patients. Two cross-sectional (study 1, n = 53; study 2, n = 200) and two longitudinal (study 3, n = 74; study 4, n = 332) studies were analyzed separately. CCL23 levels in the blood and/or cerebrospinal fluid (CSF) of each study were measured by immunoassays. Globally, our results suggest a predictive role of CCL23 protein levels both in the plasma in study 3 (hazard ratio (HR) = 2.5 (confidence interval (CI) 95% : 1.2-5.3), p = 0.02) and in the CSF in study 4 (HR = 3.05 (CI 95% : 1.02-5), p = 0.04) in cases of MCI that progress to AD. Moreover, we observed that the APOEɛ4 allele was associated with higher levels of CCL23 in study 2 (470.33 pg/mL (interquartile range (IQR): 303.33-597.76) versus 377.94 pg/mL (IQR: 267.16-529.19), p = 0.01) (APOE genotypes were available in studies 2 and 4). Together, these findings support the role of CCL23 in neuroinflammation in the early stages of AD, suggesting that CCL23 might be a candidate blood biomarker for MCI to AD progression.
Collapse
Affiliation(s)
- Júlia Faura
- Neurovascular Research Laboratory. Vall d’Hebron Institut de Recerca (VHIR) – Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory. Vall d’Hebron Institut de Recerca (VHIR) – Universitat Autònoma de Barcelona, Barcelona, Spain
- Neurology Department, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Anna Penalba
- Neurovascular Research Laboratory. Vall d’Hebron Institut de Recerca (VHIR) – Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Dolors Giralt
- Neurovascular Research Laboratory. Vall d’Hebron Institut de Recerca (VHIR) – Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Simats
- Neurovascular Research Laboratory. Vall d’Hebron Institut de Recerca (VHIR) – Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Martínez-Sáez
- Neuropathology Unit, Department of Pathology, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau - Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Spain
| | - Charlotte E. Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory and Biobank, VU University Medical Center, Amsterdam, The Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, P.O. Box 7057, 1007MB, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Laura Ibañez
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders. Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders. Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders. Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, USA
| | - Mar Hernández-Guillamón
- Neurovascular Research Laboratory. Vall d’Hebron Institut de Recerca (VHIR) – Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pilar Delgado
- Neurovascular Research Laboratory. Vall d’Hebron Institut de Recerca (VHIR) – Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory. Vall d’Hebron Institut de Recerca (VHIR) – Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Altomare D, de Wilde A, Ossenkoppele R, Pelkmans W, Bouwman F, Groot C, van Maurik I, Zwan M, Yaqub M, Barkhof F, van Berckel BN, Teunissen CE, Frisoni GB, Scheltens P, van der Flier WM. Applying the ATN scheme in a memory clinic population: The ABIDE project. Neurology 2019; 93:e1635-e1646. [PMID: 31597710 DOI: 10.1212/wnl.0000000000008361] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/21/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To apply the ATN scheme to memory clinic patients, to assess whether it discriminates patient populations with specific features. METHODS We included 305 memory clinic patients (33% subjective cognitive decline [SCD]: 60 ± 9 years, 61% M; 19% mild cognitive impairment [MCI]: 68 ± 9 years, 68% M; 48% dementia: 66 ± 10 years, 58% M) classified for positivity (±) of amyloid (A) ([18F]Florbetaben PET), tau (T) (CSF p-tau), and neurodegeneration (N) (medial temporal lobe atrophy). We assessed ATN profiles' demographic, clinical, and cognitive features at baseline, and cognitive decline over time. RESULTS The proportion of A+T+N+ patients increased with syndrome severity (from 1% in SCD to 14% in MCI and 35% in dementia), while the opposite was true for A-T-N- (from 48% to 19% and 6%). Compared to A-T-N-, patients with the Alzheimer disease profiles (A+T+N- and A+T+N+) were older (both p < 0.05) and had a higher prevalence of APOE ε4 (both p < 0.05) and lower Mini-Mental State Examination (MMSE) (both p < 0.05), memory (both p < 0.05), and visuospatial abilities (both p < 0.05) at baseline. Non-Alzheimer profiles A-T-N+ and A-T+N+ showed more severe white matter hyperintensities (both p < 0.05) and worse language performance (both p < 0.05) than A-T-N-. A linear mixed model showed faster decline on MMSE over time in A+T+N- and A+T+N+ (p = 0.059 and p < 0.001 vs A-T-N-), attributable mainly to patients without dementia. CONCLUSIONS The ATN scheme identified different biomarker profiles with overlapping baseline features and patterns of cognitive decline. The large number of profiles, which may have different implications in patients with vs without dementia, poses a challenge to the application of the ATN scheme.
Collapse
Affiliation(s)
- Daniele Altomare
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Arno de Wilde
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Rik Ossenkoppele
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Wiesje Pelkmans
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Femke Bouwman
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Colin Groot
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Ingrid van Maurik
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Marissa Zwan
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Maqsood Yaqub
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Frederik Barkhof
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Bart N van Berckel
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Charlotte E Teunissen
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Giovanni B Frisoni
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Philip Scheltens
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland
| | - Wiesje M van der Flier
- From the Alzheimer Center Amsterdam, Department of Neurology (D.A., A.d.W., R.O., W.P., F.B., C.G., I.v.M., M.Z., B.N.v.B., P.S., W.M.v.d.F.), Department of Radiology & Nuclear Medicine (R.O., C.G., M.Y., F.B., B.N.v.B.), and Neurochemistry Laboratory, Department of Clinical Chemistry (C.E.T.), Amsterdam Neuroscience, and Department of Epidemiology & Biostatistics (I.v.M., W.M.v.d.F.), Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands; Laboratory of Neuroimaging of Aging (LANVIE) (D.A., G.B.F.), University of Geneva, Switzerland; Memory Clinic (D.A.), University Hospitals of Geneva, Switzerland; Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE) (D.A.), Saint John of God Clinical Research Centre; Department of Molecular and Translational Medicine (D.A.), University of Brescia, Italy; Clinical Memory Research Unit (R.O.), Lund University, Malmö, Sweden; Institutes of Neurology and Healthcare Engineering (F.B.), UCL, London, UK; and Memory Clinic (D.A., G.B.F.), University Hospitals of Geneva, Switzerland.
| |
Collapse
|
32
|
Bertens D, Vos S, Kehoe P, Wolf H, Nobili F, Mendonça A, van Rossum I, Hort J, Molinuevo JL, Heneka M, Petersen R, Scheltens P, Visser PJ. Use of mild cognitive impairment and prodromal AD/MCI due to AD in clinical care: a European survey. ALZHEIMERS RESEARCH & THERAPY 2019; 11:74. [PMID: 31439020 PMCID: PMC6706888 DOI: 10.1186/s13195-019-0525-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 07/22/2019] [Indexed: 01/18/2023]
Abstract
Introduction The diagnosis of mild cognitive impairment (MCI) refers to cognitive impairment not meeting dementia criteria. A survey among members of the American Association of Neurology (AAN) showed that MCI was considered a useful diagnosis. Recently, research criteria have been proposed for the diagnosis of Alzheimer’s disease (AD) in MCI based on AD biomarkers (prodromal AD/MCI due to AD). The aim of this study was to investigate the attitudes of clinicians in Europe on the clinical utility of MCI and prodromal AD/MCI due to AD criteria. We also investigated whether the prodromal AD/MCI due to AD criteria impacted management of MCI patients. Methods An online survey was performed in 2015 among 102 members of the European Academy of Neurology (EAN) and the European Alzheimer’s Disease Consortium (EADC). Questions were asked on how often criteria were used, how they were operationalized, how they changed patient management, and what were considered advantages and limitations of MCI and prodromal AD/MCI due to AD. The questionnaire consisted of 47 questions scored on a Likert scale. Results Almost all respondents (92%) used the MCI diagnosis in clinical practice. Over 80% of the EAN/EADC respondents found a MCI diagnosis useful because it helped to label the cognitive problem, involve patients in planning for the future, and start risk reduction activities. These findings were similar to those reported in the AAN survey. Research criteria for prodromal AD/MCI due to AD were used by 68% of the EAN/EADC respondents. The most common reasons to use the criteria were increased certainty of diagnosis (86%), increased possibilities to provide counseling (51%), facilitation of follow-up planning (48%), start of medical intervention (49%), and response to patients’ wish for a diagnosis (41%). Over 70% of the physicians considered that a diagnosis of prodromal AD/MCI due to AD had an added value over the MCI diagnosis. Conclusions The diagnostic criteria of MCI and prodromal AD/MCI due to AD are commonly used among EAN/EADC members. The prodromal AD/MCI due to AD were considered clinically useful and impacted patient management and communication. Electronic supplementary material The online version of this article (10.1186/s13195-019-0525-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniela Bertens
- Alzheimer Centre, Department of Neurology, VU University Medical Centre, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.
| | - Stephanie Vos
- Alzheimer Centre, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Patrick Kehoe
- Learning and Research, Faculty of Health Sciences, University of Bristol, Bristol, UK
| | - Henrike Wolf
- Department of Psychiatry, University of Zurich, Zürich, Switzerland
| | - Flavio Nobili
- Clinical Neurology Unit, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
| | - Alexandre Mendonça
- Department of Neurology and Laboratory of Neurosciences, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Ineke van Rossum
- Alzheimer Centre, Department of Neurology, VU University Medical Centre, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Jacub Hort
- Department of Neurology, 2nd Faculty of Medicine Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - Jose Luis Molinuevo
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain.,Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Barcelona, Spain
| | - Michael Heneka
- Clinical Neuroscience, Department of Neurology Clinical Neuroscience Unit, and German Center for Neurodegenerative Disease (DZNE), Bonn, Germany
| | - Ron Petersen
- Mayo Clinic Alzheimer's Disease Research Center and the Mayo Clinic Study of Aging, Rochester, MN, USA
| | - Philip Scheltens
- Alzheimer Centre, Department of Neurology, VU University Medical Centre, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Centre, Department of Neurology, VU University Medical Centre, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands. .,Alzheimer Centre, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
33
|
Falgàs N, Tort-Merino A, Balasa M, Borrego-Écija S, Castellví M, Olives J, Bosch B, Férnandez-Villullas G, Antonell A, Augé JM, Lomeña F, Perissinotti A, Bargalló N, Sánchez-Valle R, Lladó A. Clinical applicability of diagnostic biomarkers in early-onset cognitive impairment. Eur J Neurol 2019; 26:1098-1104. [PMID: 30793432 DOI: 10.1111/ene.13945] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/19/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND PURPOSE Several diagnostic biomarkers are currently available for clinical use in early-onset cognitive impairment. The decision on which biomarker is used in each patient depends on several factors such as its predictive value or tolerability. METHODS There were a total of 40 subjects with early-onset cognitive complaints (<65 years of age): 26 with Alzheimer's disease (AD), five with frontotemporal dementia and nine with diagnostic suspicion of non-neurodegenerative disorder. Clinical and neuropsychological evaluation, lumbar puncture for cerebrospinal fluid (CSF) AD core biochemical marker determination, medial temporal atrophy evaluation on magnetic resonance imaging, amyloid-positron emission tomography (PET) and 18 F-fluorodeoxyglucose-PET were performed. Neurologists provided pre- and post-biomarker diagnosis, together with diagnostic confidence and clinical/therapeutic management. Patients scored the tolerability of each procedure. RESULTS Cerebrospinal fluid biomarkers and amyloid-PET increased diagnostic confidence in AD (77.4%-86.2% after CSF, 92.4% after amyloid-PET, P < 0.01) and non-neurodegenerative conditions (53.6%-75% after CSF, 95% after amyloid-PET, P < 0.05). Biomarker results led to diagnostic (32.5%) and treatment (32.5%) changes. All tests were well tolerated. CONCLUSIONS Biomarker procedures are well tolerated and have an important diagnostic/therapeutic impact on early-onset cognitive impairment.
Collapse
Affiliation(s)
- N Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - A Tort-Merino
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - M Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Atlantic Fellow for Equity in Brain Health, Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - S Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - M Castellví
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - J Olives
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - B Bosch
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - G Férnandez-Villullas
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - A Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - J M Augé
- Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, Barcelona
| | - F Lomeña
- Nuclear Medicine Department, Hospital Clínic de Barcelona, Barcelona
| | - A Perissinotti
- Nuclear Medicine Department, Hospital Clínic de Barcelona, Barcelona
| | - N Bargalló
- Image Diagnostic Centre, IDIBAPS, Hospital Clínic de Barcelona, Barcelona, Spain
| | - R Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - A Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
34
|
Ferro DA, van den Brink H, Exalto LG, Boomsma JM, Barkhof F, Prins ND, van der Flier WM, Biessels GJ. Clinical relevance of acute cerebral microinfarcts in vascular cognitive impairment. Neurology 2019; 92:e1558-e1566. [DOI: 10.1212/wnl.0000000000007250] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/24/2018] [Indexed: 11/15/2022] Open
Abstract
ObjectiveTo determine the occurrence of acute cerebral microinfarcts (ACMIs) in memory clinic patients and relate their presence to vascular risk and cognitive profile, CSF and neuroimaging markers, and clinical outcome.MethodsThe TRACE-VCI study is a memory clinic cohort of patients with vascular brain injury on MRI (i.e., possible vascular cognitive impairment [VCI]). We included 783 patients (mean age 67.6 ± 8.5, 46% female) with available 3T diffusion-weighted imaging (DWI). ACMIs were defined as supratentorial DWI hyperintensities <5 mm with a corresponding hypo/isointense apparent diffusion coefficient signal and iso/hyperintense T2*-weighted signal.ResultsA total of 23 ACMIs were found in 16 of the 783 patients (2.0%). Patients with ACMIs did not differ in vascular risk or cognitive profile, but were more often diagnosed with vascular dementia (odds ratio [OR] 5.1; 95% confidence interval [CI] 1.4–18.9, p = 0.014). ACMI presence was associated with lower levels of β-amyloid (p < 0.004) and with vascular imaging markers (lacunar infarcts: OR 3.5, CI 1.3–9.6, p = 0.015; nonlacunar infarcts: OR 4.1, CI 1.4–12.5, p = 0.012; severe white matter hyperintensities: OR 4.8, CI 1.7–13.8, p = 0.004; microbleeds: OR 18.9, CI 2.5–144.0, p = 0.0001). After a median follow-up of 2.1 years, the risk of poor clinical outcome (composite of marked cognitive decline, major vascular event, death, and institutionalization) was increased among patients with ACMIs (hazard ratio 3.0; 1.4–6.0, p = 0.005).ConclusionIn patients with possible VCI, ACMI presence was associated with a high burden of cerebrovascular disease of both small and large vessel etiology and poor clinical outcome. ACMIs may thus be a novel marker of active vascular brain injury in these patients.
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Alzheimer disease (AD) is the most common cause of late-onset dementia. This article describes the epidemiology, genetic and environmental risk factors, clinical diagnosis, biomarkers, and treatment of late-onset AD, defined by age of onset of 65 years or older. RECENT FINDINGS An estimated 5.7 million Americans are living with AD dementia, with the number of affected individuals growing rapidly because of an aging population. Vascular risk factors, sleep disorders, and traumatic brain injury are associated with an increased risk of AD, while increased cognitive and physical activity throughout the lifespan reduce the risk of disease. The primary genetic risk factor for late-onset AD is the apolipoprotein E (APOE) ε4 allele. AD typically presents with early and prominent episodic memory loss, although this clinical syndrome is neither sensitive nor specific for underlying AD neuropathology. Emerging CSF and imaging biomarkers can now detect the key neuropathologic features of the disease (amyloid plaques, neurofibrillary tangles, and neurodegeneration) in living people, allowing for characterization of patients based on biological measures. A comprehensive treatment plan for AD includes use of symptomatic medications, optimal treatment of comorbid conditions and neuropsychiatric symptoms, counseling about safety and future planning, and referrals to community resources. SUMMARY AD is very common in older neurologic patients. Neurologists should set the standard for the diagnosis and care of patients with AD and should be familiar with emerging biomarkers that have transformed AD research and are primed to enter the clinical arena.
Collapse
|
36
|
Genetic screening in early-onset Alzheimer's disease identified three novel presenilin mutations. Neurobiol Aging 2019; 86:201.e9-201.e14. [PMID: 30797548 DOI: 10.1016/j.neurobiolaging.2019.01.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 01/09/2019] [Accepted: 01/21/2019] [Indexed: 12/23/2022]
Abstract
Mutations in presenilin 1 (PSEN1), presenilin 2 (PSEN2), and amyloid precursor protein (APP) are major genetic causes of early-onset Alzheimer's disease (EOAD). Clinical heterogeneity is frequently observed in patients with PSEN1 and PSEN2 mutations. Using whole exome sequencing, we screened a Dutch cohort of 68 patients with EOAD for rare variants in Mendelian Alzheimer's disease, frontotemporal dementia, and prion disease genes. We identified 3 PSEN1 and 2 PSEN2 variants. Three variants, 1 in PSEN1 (p.H21Profs*2) and both PSEN2 (p.A415S and p.M174I), were novel and absent in control exomes. These novel variants can be classified as probable pathogenic, except for PSEN1 (p.H21Profs*2) in which the pathogenicity is uncertain. The initial clinical symptoms between mutation carriers varied from behavioral problems to memory impairment. Our findings extend the mutation spectrum of EOAD and underline the clinical heterogeneity among PSEN1 and PSEN2 mutation carriers. Screening for Alzheimer's disease-causing genes is indicated in presenile dementia with an overlapping clinical diagnosis.
Collapse
|
37
|
Shaw LM, Arias J, Blennow K, Galasko D, Molinuevo JL, Salloway S, Schindler S, Carrillo MC, Hendrix JA, Ross A, Illes J, Ramus C, Fifer S. Appropriate use criteria for lumbar puncture and cerebrospinal fluid testing in the diagnosis of Alzheimer's disease. Alzheimers Dement 2018; 14:1505-1521. [PMID: 30316776 PMCID: PMC10013957 DOI: 10.1016/j.jalz.2018.07.220] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/17/2018] [Accepted: 07/31/2018] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The Alzheimer's Association convened a multidisciplinary workgroup to develop appropriate use criteria to guide the safe and optimal use of the lumbar puncture procedure and cerebrospinal fluid (CSF) testing for Alzheimer's disease pathology detection in the diagnostic process. METHODS The workgroup, experienced in the ethical use of lumbar puncture and CSF analysis, developed key research questions to guide the systematic review of the evidence and developed clinical indications commonly encountered in clinical practice based on key patient groups in whom the use of lumbar puncture and CSF may be considered as part of the diagnostic process. Based on their expertise and interpretation of the evidence from systematic review, members rated each indication as appropriate or inappropriate. RESULTS The workgroup finalized 14 indications, rating 6 appropriate and 8 inappropriate. DISCUSSION In anticipation of the emergence of more reliable CSF analysis platforms, the manuscript offers important guidance to health-care practitioners and suggestions for implementation and future research.
Collapse
Affiliation(s)
- Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Jalayne Arias
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenberg, Molndal, Sweden
| | - Douglas Galasko
- Department of Neuroscience, University of California, San Diego, CA, USA
| | | | - Stephen Salloway
- Butler Hospital Memory and Aging Program, The Warren Alpert Medical School of Brown University, Brown University, Providence, RI, USA
| | | | | | | | - April Ross
- Alzheimer's Association, Chicago, IL, USA
| | | | | | | |
Collapse
|
38
|
Recent trends in analytical approaches for detecting neurotransmitters in Alzheimer's disease. Trends Analyt Chem 2018. [DOI: 10.1016/j.trac.2018.05.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
39
|
Handels RLH, Wimo A, Dodel R, Kramberger MG, Visser PJ, Molinuevo JL, Verhey FRJ, Winblad B. Cost-Utility of Using Alzheimer's Disease Biomarkers in Cerebrospinal Fluid to Predict Progression from Mild Cognitive Impairment to Dementia. J Alzheimers Dis 2018; 60:1477-1487. [PMID: 29081416 DOI: 10.3233/jad-170324] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Diagnostic research criteria for Alzheimer's disease support the use of biomarkers in the cerebrospinal fluid (CSF) to improve the accuracy of the prognosis regarding progression to dementia for people with mild cognitive impairment (MCI). OBJECTIVE The aim of this study was to estimate the potential incremental cost-effectiveness ratio of adding CSF biomarker testing to the standard diagnostic workup to determine the prognosis for patients with MCI. METHODS In an early technology assessment, a mathematical simulation model was built, using available evidence on added prognostic value as well as expert opinion to estimate the incremental costs and quality-adjusted life years (QALYs) of 20,000 virtual MCI patients with (intervention strategy) and without (control strategy) relying on CSF, from a health-care sector perspective and with a 5-year time horizon. RESULTS Adding the CSF test improved the accuracy of prognosis by 11%. This resulted in an average QALY gain of 0.046 and € 432 additional costs per patient, representing an incremental cost-effectiveness ratio of € 9,416. CONCLUSION The results show the potential of CSF biomarkers in current practice from a health-economics perspective. This result was, however, marked by a high degree of uncertainty, and empirical research is required into the impact of a prognosis on worrying, false-positive/negative prognosis, and stigmatization.
Collapse
Affiliation(s)
- Ron L H Handels
- Department of Psychiatry and Neuropsychology, Alzheimer Centre Limburg, School for Mental Health and Neurosciences, Maastricht University, Maastricht, The Netherlands.,Department of Neurobiology, Care Science and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institute, Huddinge, Sweden
| | - Anders Wimo
- Department of Neurobiology, Care Science and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institute, Huddinge, Sweden
| | - Richard Dodel
- Department of NeuroGeriatrics, University Duisburg-Essen, Essen, Germany
| | - Milica G Kramberger
- Department of Neurobiology, Care Science and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institute, Huddinge, Sweden.,Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centre Limburg, School for Mental Health and Neurosciences, Maastricht University, Maastricht, The Netherlands.,Department of Neurology and Alzheimer Centre, VU University Medical Centre, Amsterdam, The Netherlands
| | - José Luis Molinuevo
- Alzheimer Disease and Other Cognitive Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain.,Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology, Alzheimer Centre Limburg, School for Mental Health and Neurosciences, Maastricht University, Maastricht, The Netherlands
| | - Bengt Winblad
- Department of Neurobiology, Care Science and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institute, Huddinge, Sweden
| |
Collapse
|
40
|
Bos I, Vos S, Vandenberghe R, Scheltens P, Engelborghs S, Frisoni G, Molinuevo JL, Wallin A, Lleó A, Popp J, Martinez-Lage P, Baird A, Dobson R, Legido-Quigley C, Sleegers K, Van Broeckhoven C, Bertram L, ten Kate M, Barkhof F, Zetterberg H, Lovestone S, Streffer J, Visser PJ. The EMIF-AD Multimodal Biomarker Discovery study: design, methods and cohort characteristics. Alzheimers Res Ther 2018; 10:64. [PMID: 29980228 PMCID: PMC6035398 DOI: 10.1186/s13195-018-0396-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 06/08/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND There is an urgent need for novel, noninvasive biomarkers to diagnose Alzheimer's disease (AD) in the predementia stages and to predict the rate of decline. Therefore, we set up the European Medical Information Framework for Alzheimer's Disease Multimodal Biomarker Discovery (EMIF-AD MBD) study. In this report we describe the design of the study, the methods used and the characteristics of the participants. METHODS Participants were selected from existing prospective multicenter and single-center European studies. Inclusion criteria were having normal cognition (NC) or a diagnosis of mild cognitive impairment (MCI) or AD-type dementia at baseline, age above 50 years, known amyloid-beta (Aβ) status, availability of cognitive test results and at least two of the following materials: plasma, DNA, magnetic resonance imaging (MRI) or cerebrospinal fluid (CSF). Targeted and untargeted metabolomic and proteomic analyses were performed in plasma, and targeted and untargeted proteomics were performed in CSF. Genome-wide SNP genotyping, next-generation sequencing and methylation profiling were conducted in DNA. Visual rating and volumetric measures were assessed on MRI. Baseline characteristics were analyzed using ANOVA or chi-square, rate of decline analyzed by linear mixed modeling. RESULTS We included 1221 individuals (NC n = 492, MCI n = 527, AD-type dementia n = 202) with a mean age of 67.9 (SD 8.3) years. The percentage Aβ+ was 26% in the NC, 58% in the MCI, and 87% in the AD-type dementia groups. Plasma samples were available for 1189 (97%) subjects, DNA samples for 929 (76%) subjects, MRI scans for 862 (71%) subjects and CSF samples for 767 (63%) subjects. For 759 (62%) individuals, clinical follow-up data were available. In each diagnostic group, the APOE ε4 allele was more frequent amongst Aβ+ individuals (p < 0.001). Only in MCI was there a difference in baseline Mini Mental State Examination (MMSE) score between the A groups (p < 0.001). Aβ+ had a faster rate of decline on the MMSE during follow-up in the NC (p < 0.001) and MCI (p < 0.001) groups. CONCLUSIONS The characteristics of this large cohort of elderly subjects at various cognitive stages confirm the central roles of Aβ and APOE ε4 in AD pathogenesis. The results of the multimodal analyses will provide new insights into underlying mechanisms and facilitate the discovery of new diagnostic and prognostic AD biomarkers. All researchers can apply for access to the EMIF-AD MBD data by submitting a research proposal via the EMIF-AD Catalog.
Collapse
Affiliation(s)
- Isabelle Bos
- Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Universiteitssingel 40, Box 34, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Stephanie Vos
- Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Rik Vandenberghe
- University Hospital Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Philip Scheltens
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Giovanni Frisoni
- University of Geneva, Geneva, Switzerland
- IRCCS Instituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - José Luis Molinuevo
- Alzheimer’s Disease & Other Cognitive Disorders Unit, Hospital Clínic—IDIBAPS, Barcelona, Spain
- Barcelona Beta Brain Research Center, Fundació Pasqual Maragall, Barcelona, Spain
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Moelndal, Sweden
| | - Alberto Lleó
- Hospital de la Santa Creu i Sant Pau, Barcelona, 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
| | - Pablo Martinez-Lage
- Center for Research and Advanced Therapies, CITA—Alzheimer Foundation, San Sebastian, Spain
| | | | - Richard Dobson
- King’s College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Farr Institute of Health Informatics Research, UCL Institute of Health Informatics, University College London, London, UK
- NIHR University College London Hospitals Biomedical Research Centre, London, UK
| | | | - Kristel Sleegers
- Neurodegenerative Brain Diseases Group, VIB—Department of Molecular Genetics, Antwerp, Belgium
- Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, VIB—Department of Molecular Genetics, Antwerp, Belgium
- Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
- School of Public Health, Imperial College London, London, UK
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Mara ten Kate
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute, London, UK
| | | | - Johannes Streffer
- Experimental Medicine, Janssen Pharmaceutical Companies, Beerse, Belgium
| | - Pieter Jelle Visser
- Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
41
|
Conwell K, von Reutern B, Richter N, Kukolja J, Fink GR, Onur OA. Test-retest variability of resting-state networks in healthy aging and prodromal Alzheimer's disease. NEUROIMAGE-CLINICAL 2018; 19:948-962. [PMID: 30003032 PMCID: PMC6039839 DOI: 10.1016/j.nicl.2018.06.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 12/03/2022]
Abstract
In recent years, changes in resting-state networks (RSN), identified by functional magnetic resonance imaging (fMRI), have gained increasing attention as potential biomarkers and trackers of neurological disorders such as Alzheimer's disease (AD). Intersession reliability of RSN is fundamental to this approach. In this study, we investigated the test-retest reliability of three memory related RSN (i.e., the default mode, salience, and executive control network) in 15 young, 15 healthy seniors (HS), and 15 subjects affected by mild cognitive impairment (MCI) with positive biomarkers suggestive of incipient AD (6 females each). FMRI was conducted on three separate occasions. Independent Component Analysis decomposed the resting-state data into RSNs. Comparisons of variation in functional connectivity between groups were made applying different thresholds in an explorative approach. Intersession test-retest reliability was evaluated by intraclass correlation coefficient (ICC) comparisons. To assess the effect of gray matter volume loss, motion, cerebrospinal fluid based biomarkers and the time gap between sessions on intersession variation, the former four were correlated separately with the latter. Data showed that i) young subjects ICCs (relative to HS/MCI-subjects) had higher intersession reliability, ii) stringent statistical thresholds need to be applied to prevent false-positives, iii) both HS and MCI-subjects (relative to young) showed significantly more clusters of intersession variation in all three RSN, iv) while intersession variation was highly correlated with head motion, it was also correlated with biomarkers (especially phospho-tau), the time gap between sessions and local GMV. Results indicate that time gaps between sessions should be kept constant and that head motion must be taken into account when using RSN to assess aging and neurodegeneration. In patients with prodromal AD, re-test reliability may be increased by accouting for overall disease burden by including biomarkers of neuronal injury (especially phospho-tau) in statistical analyses. Local atrophy however, does not seem to play a major role in regards to reliability, but should be used as covariate depending on the research question. Intersession reliability of resting state networks is highest in young subjects. Test-Retest Variability increases with aging and in MCI. Motion and csf-biomarkers correlate with increased variability. Motion and biomarkers should be included as confounders in the statistical models. Stringent statistical thresholds should be applied to prevent type I-errors.
Collapse
Affiliation(s)
- K Conwell
- Department of Neurology, University Hospital of Cologne, Cologne 50937, Germany; Department of General, Abdominal, Endocrine and Minimally Invasive Surgery, Academic Hospital Bogenhausen, 81925 Munich, Germany
| | - B von Reutern
- Department of Neurology, University Hospital of Cologne, Cologne 50937, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre, Jülich 52428, Germany
| | - N Richter
- Department of Neurology, University Hospital of Cologne, Cologne 50937, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre, Jülich 52428, Germany
| | - J Kukolja
- Department of Neurology, University Hospital of Cologne, Cologne 50937, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre, Jülich 52428, Germany
| | - G R Fink
- Department of Neurology, University Hospital of Cologne, Cologne 50937, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre, Jülich 52428, Germany
| | - O A Onur
- Department of Neurology, University Hospital of Cologne, Cologne 50937, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre, Jülich 52428, Germany.
| |
Collapse
|
42
|
Lewczuk P, Riederer P, O’Bryant SE, Verbeek MM, Dubois B, Visser PJ, Jellinger KA, Engelborghs S, Ramirez A, Parnetti L, Jack CR, Teunissen CE, Hampel H, Lleó A, Jessen F, Glodzik L, de Leon MJ, Fagan AM, Molinuevo JL, Jansen WJ, Winblad B, Shaw LM, Andreasson U, Otto M, Mollenhauer B, Wiltfang J, Turner MR, Zerr I, Handels R, Thompson AG, Johansson G, Ermann N, Trojanowski JQ, Karaca I, Wagner H, Oeckl P, van Waalwijk van Doorn L, Bjerke M, Kapogiannis D, Kuiperij HB, Farotti L, Li Y, Gordon BA, Epelbaum S, Vos SJB, Klijn CJM, Van Nostrand WE, Minguillon C, Schmitz M, Gallo C, Mato AL, Thibaut F, Lista S, Alcolea D, Zetterberg H, Blennow K, Kornhuber J, Riederer P, Gallo C, Kapogiannis D, Mato AL, Thibaut F. Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: An update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of Societies of Biological Psychiatry. World J Biol Psychiatry 2018; 19:244-328. [PMID: 29076399 PMCID: PMC5916324 DOI: 10.1080/15622975.2017.1375556] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the 12 years since the publication of the first Consensus Paper of the WFSBP on biomarkers of neurodegenerative dementias, enormous advancement has taken place in the field, and the Task Force takes now the opportunity to extend and update the original paper. New concepts of Alzheimer's disease (AD) and the conceptual interactions between AD and dementia due to AD were developed, resulting in two sets for diagnostic/research criteria. Procedures for pre-analytical sample handling, biobanking, analyses and post-analytical interpretation of the results were intensively studied and optimised. A global quality control project was introduced to evaluate and monitor the inter-centre variability in measurements with the goal of harmonisation of results. Contexts of use and how to approach candidate biomarkers in biological specimens other than cerebrospinal fluid (CSF), e.g. blood, were precisely defined. Important development was achieved in neuroimaging techniques, including studies comparing amyloid-β positron emission tomography results to fluid-based modalities. Similarly, development in research laboratory technologies, such as ultra-sensitive methods, raises our hopes to further improve analytical and diagnostic accuracy of classic and novel candidate biomarkers. Synergistically, advancement in clinical trials of anti-dementia therapies energises and motivates the efforts to find and optimise the most reliable early diagnostic modalities. Finally, the first studies were published addressing the potential of cost-effectiveness of the biomarkers-based diagnosis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, and Department of Biochemical Diagnostics, University Hospital of Białystok, Białystok, Poland
| | - Peter Riederer
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Sid E. O’Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Marcel M. Verbeek
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Salpêtrièrie Hospital, INSERM UMR-S 975 (ICM), Paris 6 University, Paris, France
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
- Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience VU University Medical Centre, Amsterdam, The Netherlands
| | | | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Alfredo Ramirez
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Lucilla Parnetti
- Section of Neurology, Center for Memory Disturbances, Lab of Clinical Neurochemistry, University of Perugia, Perugia, Italy
| | | | - Charlotte E. Teunissen
- Neurochemistry Lab and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Spain
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Germany
| | - Lidia Glodzik
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Mony J. de Leon
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Anne M. Fagan
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - José Luis Molinuevo
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Willemijn J. Jansen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Bengt Winblad
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ulf Andreasson
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel and University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
| | - Jens Wiltfang
- Department of Psychiatry & Psychotherapy, University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Martin R. Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Inga Zerr
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Clinical Dementia Centre, Department of Neurology, University Medical School, Göttingen, Germany
| | - Ron Handels
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | | | - Gunilla Johansson
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Natalia Ermann
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilker Karaca
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Holger Wagner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Linda van Waalwijk van Doorn
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), Baltimore, MD, USA
| | - H. Bea Kuiperij
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Lucia Farotti
- Section of Neurology, Center for Memory Disturbances, Lab of Clinical Neurochemistry, University of Perugia, Perugia, Italy
| | - Yi Li
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Brian A. Gordon
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stéphane Epelbaum
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Salpêtrièrie Hospital, INSERM UMR-S 975 (ICM), Paris 6 University, Paris, France
| | - Stephanie J. B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Catharina J. M. Klijn
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | | | - Carolina Minguillon
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Matthias Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Clinical Dementia Centre, Department of Neurology, University Medical School, Göttingen, Germany
| | - Carla Gallo
- Departamento de Ciencias Celulares y Moleculares/Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Andrea Lopez Mato
- Chair of Psychoneuroimmunoendocrinology, Maimonides University, Buenos Aires, Argentina
| | - Florence Thibaut
- Department of Psychiatry, University Hospital Cochin-Site Tarnier 89 rue d’Assas, INSERM 894, Faculty of Medicine Paris Descartes, Paris, France
| | - Simone Lista
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Muguruma Y, Tsutsui H, Noda T, Akatsu H, Inoue K. Widely targeted metabolomics of Alzheimer's disease postmortem cerebrospinal fluid based on 9-fluorenylmethyl chloroformate derivatized ultra-high performance liquid chromatography tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1091:53-66. [PMID: 29852382 DOI: 10.1016/j.jchromb.2018.05.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/10/2018] [Accepted: 05/21/2018] [Indexed: 12/22/2022]
Abstract
Confirmed biomarkers of postmortem cerebrospinal fluid (pCSF) are used to differentiate between Alzheimer's disease (AD) patients and healthy seniors with high diagnostic accuracy. However, the extent to which the performance of specific metabolic profiling facilitates reliable estimations of the concentrations of the different pCSF biomarkers and their ratios remains unclear. The interpretation of the lower levels of molecules of metabolic profiling and their concentration ratios in pCSF related to brain disorders could facilitate an unchallenging detection of peripheral biomarkers of AD stages and other dementia types. In this study, we proposed the use of widely targeted metabolomics for pCSF metabolic profiling using 9-fluorenylmethyl chloroformate- (FMOC) derivatized ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) to evaluate the diversity of 97 amine-mediated metabolic patterns and pathways from confirmed diagnosis based on AD brain pathology. Our results identified the metabolites that contributed toward and mutually influenced the principal component analysis plot with integrated analytes. Furthermore, the AD group showed a significant variation in several analyte concentration levels compared to those of control subjects. These trends of the concentration levels expressed by the amine metabolic pathways indicated the decreased activity of polyamine and tryptophan-kynurenine (Trp-Kyn) metabolisms. Moreover, increased metabolites such as methionine sulfoxide, 3-methoxy-anthranilate, cadaverine, guanine, and histamine were observed by widely targeted metabolomics of pCSF from the AD subjects. According to their metabolic pathway analysis using FMOC-derivatized UHPLC-MS/MS assay, we supposed that the involvement of polyamine and Trp-Kyn metabolisms was observed in the pCSF samples.
Collapse
Affiliation(s)
- Yoshio Muguruma
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
| | - Haruhito Tsutsui
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan; ONO Pharmaceutical Co., Ltd, 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - Takumi Noda
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan; ONO Pharmaceutical Co., Ltd, 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - Hiroyasu Akatsu
- Department of Medicine for Aging Place, Community Health Care/Community-Based Medical Education, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-0001, Japan; Department of Neuropathology, Choju Medical Institute, Fukushimura Hospital, Toyohashi 441-8124, Japan
| | - Koichi Inoue
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan.
| |
Collapse
|
44
|
van der Flier WM, Scheltens P. Amsterdam Dementia Cohort: Performing Research to Optimize Care. J Alzheimers Dis 2018; 62:1091-1111. [PMID: 29562540 PMCID: PMC5870023 DOI: 10.3233/jad-170850] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2017] [Indexed: 01/01/2023]
Abstract
The Alzheimer center of the VU University Medical Center opened in 2000 and was initiated to combine both patient care and research. Together, to date, all patients forming the Amsterdam Dementia Cohort number almost 6,000 individuals. In this cohort profile, we provide an overview of the results produced based on the Amsterdam Dementia Cohort. We describe the main results over the years in each of these research lines: 1) early diagnosis, 2) heterogeneity, and 3) vascular factors. Among the most important research efforts that have also impacted patients' lives and/or the research field, we count the development of novel, easy to use diagnostic measures such as visual rating scales for MRI and the Amsterdam IADL Questionnaire, insight in different subgroups of AD, and findings on incidence and clinical sequelae of microbleeds. Finally, we describe in the outlook how our research endeavors have improved the lives of our patients.
Collapse
Affiliation(s)
- Wiesje M. van der Flier
- Department of Neurology, Alzheimer Center, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center, VU University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
45
|
van Maurik IS, Zwan MD, Tijms BM, Bouwman FH, Teunissen CE, Scheltens P, Wattjes MP, Barkhof F, Berkhof J, van der Flier WM. Interpreting Biomarker Results in Individual Patients With Mild Cognitive Impairment in the Alzheimer's Biomarkers in Daily Practice (ABIDE) Project. JAMA Neurol 2017; 74:1481-1491. [PMID: 29049480 PMCID: PMC5822193 DOI: 10.1001/jamaneurol.2017.2712] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/24/2017] [Indexed: 12/12/2022]
Abstract
Importance Biomarkers do not determine conversion to Alzheimer disease (AD) perfectly, and criteria do not specify how to take patient characteristics into account. Consequently, biomarker use may be challenging for clinicians, especially in patients with mild cognitive impairment (MCI). Objective To construct biomarker-based prognostic models that enable determination of future AD dementia in patients with MCI. Design, Setting, and Participants This study is part of the Alzheimer's Biomarkers in Daily Practice (ABIDE) project. A total of 525 patients with MCI from the Amsterdam Dementia Cohort (longitudinal cohort, tertiary referral center) were studied. All patients had their baseline visit to a memory clinic from September 1, 1997, through August 31, 2014. Prognostic models were constructed by Cox proportional hazards regression with patient characteristics (age, sex, and Mini-Mental State Examination [MMSE] score), magnetic resonance imaging (MRI) biomarkers (hippocampal volume, normalized whole-brain volume), cerebrospinal fluid (CSF) biomarkers (amyloid-β1-42, tau), and combined biomarkers. Data were analyzed from November 1, 2015, to October 1, 2016. Main Outcomes and Measures Clinical end points were AD dementia and any type of dementia after 1 and 3 years. Results Of the 525 patients, 210 (40.0%) were female, and the mean (SD) age was 67.3 (8.4) years. On the basis of age, sex, and MMSE score only, the 3-year progression risk to AD dementia ranged from 26% (95% CI, 19%-34%) in younger men with MMSE scores of 29 to 76% (95% CI, 65%-84%) in older women with MMSE scores of 24 (1-year risk: 6% [95% CI, 4%-9%] to 24% [95% CI, 18%-32%]). Three- and 1-year progression risks were 86% (95% CI, 71%-95%) and 27% (95% CI, 17%-41%) when MRI results were abnormal, 82% (95% CI, 73%-89%) and 26% (95% CI, 20%-33%) when CSF test results were abnormal, and 89% (95% CI, 79%-95%) and 26% (95% CI, 18%-36%) when the results of both tests were abnormal. Conversely, 3- and 1-year progression risks were 18% (95% CI, 13%-27%) and 3% (95% CI, 2%-5%) after normal MRI results, 6% (95% CI, 3%-9%) and 1% (95% CI, 0.5%-2%) after normal CSF test results, and 4% (95% CI, 2%-7%) and 0.5% (95% CI, 0.2%-1%) after combined normal MRI and CSF test results. The prognostic value of models determining any type of dementia were in the same order of magnitude although somewhat lower. External validation in Alzheimer's Disease Neuroimaging Initiative 2 showed that our models were highly robust. Conclusions and Relevance This study provides biomarker-based prognostic models that may help determine AD dementia and any type of dementia in patients with MCI at the individual level. This finding supports clinical decision making and application of biomarkers in daily practice.
Collapse
Affiliation(s)
- Ingrid S. van Maurik
- Department of Neurology and Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Marissa D. Zwan
- Department of Neurology and Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Betty M. Tijms
- Department of Neurology and Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Femke H. Bouwman
- Department of Neurology and Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Philip Scheltens
- Department of Neurology and Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Mike P. Wattjes
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
- Institutes of Neurology and Healthcare Engineering, University College London, London, England
| | - Johannes Berkhof
- Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Wiesje M. van der Flier
- Department of Neurology and Alzheimer Center, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
46
|
Abstract
Laboratory-specific reference values for cerebrospinal fluid (CSF) Alzheimer disease (AD) biomarkers are necessary. Our objective was to apply well-known CSF biomarkers and redetermine their diagnostic cutoff values for AD in South Korea. CSF samples from matched control subjects (n=71), patients with AD dementia (ADD, n=76), and other neurological disorders with cognitive decline (OND, n=47) were obtained from 6 Korean dementia clinics according to a standardized protocol. CSF biomarker concentrations were measured using enzyme-linked immunosorbent assay. CSF biomarkers differed significantly between the ADD and control groups (P<0.001 for all), and between the ADD and OND groups (P<0.001 for all). The areas under the curve in differentiation of ADD from control subjects were 0.97 for Aβ42, 0.93 for total tau (tTau), 0.86 for pTau, and 0.99 for both tTau/Aβ42 and pTau/Aβ42 ratios. Our revised cutoff value for Aβ42 was higher than our previous one, whereas the values for the Tau proteins were similar. The tTau/Aβ42 ratio had the highest accuracy, 97%. Our findings highlight the usefulness of CSF AD biomarkers in South Korea, and the necessity of continually testing the reliability of cutoff values.
Collapse
|
47
|
Ceccaldi M, Jonveaux T, Verger A, Krolak‐Salmon P, Houzard C, Godefroy O, Shields T, Perrotin A, Gismondi R, Bullich S, Jovalekic A, Raffa N, Pasquier F, Semah F, Dubois B, Habert M, Wallon D, Chastan M, Payoux P, Ceccaldi M, Guedj E, Ceccaldi M, Felician O, Didic M, Gueriot C, Koric L, Kletchkova‐Gantchev R, Guedj E, Godefroy O, Andriuta D, Devendeville A, Dupuis D, Binot I, Barbay M, Meyer M, Moullard V, Magnin E, Chamard L, Haffen S, Morel O, Drouet C, Boulahdour H, Goas P, Querellou‐Lefranc S, Sayette V, Cogez J, Branger P, Agostini D, Manrique A, Rouaud O, Bejot Y, Jacquin‐Piques A, Dygai‐Cochet I, Berriolo‐Riedinger A, Moreaud O, Sauvee M, Crépin CG, Pasquier F, Bombois S, Lebouvier T, Mackowiak‐Cordoliani M, Deramecourt V, Rollin‐Sillaire A, Cassagnaud‐Thuillet P, Chen Y, Semah F, Petyt G, Krolak‐Salmon P, Federico D, Danaila KL, Guilhermet Y, Magnier C, Makaroff Z, Rouch I, Xie J, Roubaud C, Coste M, David K, Sarciron A, Waissi AS, Scheiber C, Houzard C, Gabelle‐Deloustal A, Bennys K, Marelli C, Touati L, Mariano‐Goulart D, Verbizier‐Lonjon D, Jonveaux T, Benetos A, Kearney‐Schwartz A, Perret‐Guillaume C, Verger A, Vercelletto M, Boutoleau‐Bretonniere C, Pouclet‐Courtemanche H, Wagemann N, Pallardy A, Hugon J, Paquet C, et alCeccaldi M, Jonveaux T, Verger A, Krolak‐Salmon P, Houzard C, Godefroy O, Shields T, Perrotin A, Gismondi R, Bullich S, Jovalekic A, Raffa N, Pasquier F, Semah F, Dubois B, Habert M, Wallon D, Chastan M, Payoux P, Ceccaldi M, Guedj E, Ceccaldi M, Felician O, Didic M, Gueriot C, Koric L, Kletchkova‐Gantchev R, Guedj E, Godefroy O, Andriuta D, Devendeville A, Dupuis D, Binot I, Barbay M, Meyer M, Moullard V, Magnin E, Chamard L, Haffen S, Morel O, Drouet C, Boulahdour H, Goas P, Querellou‐Lefranc S, Sayette V, Cogez J, Branger P, Agostini D, Manrique A, Rouaud O, Bejot Y, Jacquin‐Piques A, Dygai‐Cochet I, Berriolo‐Riedinger A, Moreaud O, Sauvee M, Crépin CG, Pasquier F, Bombois S, Lebouvier T, Mackowiak‐Cordoliani M, Deramecourt V, Rollin‐Sillaire A, Cassagnaud‐Thuillet P, Chen Y, Semah F, Petyt G, Krolak‐Salmon P, Federico D, Danaila KL, Guilhermet Y, Magnier C, Makaroff Z, Rouch I, Xie J, Roubaud C, Coste M, David K, Sarciron A, Waissi AS, Scheiber C, Houzard C, Gabelle‐Deloustal A, Bennys K, Marelli C, Touati L, Mariano‐Goulart D, Verbizier‐Lonjon D, Jonveaux T, Benetos A, Kearney‐Schwartz A, Perret‐Guillaume C, Verger A, Vercelletto M, Boutoleau‐Bretonniere C, Pouclet‐Courtemanche H, Wagemann N, Pallardy A, Hugon J, Paquet C, Dumurgier J, Millet P, Queneau M, Dubois B, Epelbaum S, Levy M, Habert M, Novella J, Jaidi Y, Papathanassiou D, Morland D, Belliard S, Salmon A, Lejeune F, Hannequin D, Wallon D, Martinaud O, Zarea A, Chastan M, Pariente J, Thalamas C, Galitzky‐Gerber M, Tricoire Ricard A, Calvas F, Rigal E, Payoux P, Hitzel A, Delrieu J, Ousset P, Lala F, Sastre‐Hengan N, Stephens A, Guedj E. Added value of
18
F‐florbetaben amyloid PET in the diagnostic workup of most complex patients with dementia in France: A naturalistic study. Alzheimers Dement 2017; 14:293-305. [DOI: 10.1016/j.jalz.2017.09.009] [Show More Authors] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/29/2017] [Accepted: 09/06/2017] [Indexed: 11/25/2022]
Affiliation(s)
- Mathieu Ceccaldi
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Thérèse Jonveaux
- Geriatric Department CHRU de Nancy–Hôpital Brabois Vandoeuvre‐les‐Nancy France
| | - Antoine Verger
- INSERM U947 Unité d'Imagerie Adaptative Diagnostique et Interventionnelle Nancy France
| | - Pierre Krolak‐Salmon
- Clinical and Research Memory Center of Lyon Hospices civils de Lyon, Université Claude Bernard Lyon 1 Inserm 1028 Lyon France
| | | | - Olivier Godefroy
- Neurology Department CHU Amiens Picardie–Hôpital Sud Amiens France
| | - Trevor Shields
- Nuclear Medicine Department CHU Amiens Picardie–Hôpital Sud Amiens France
| | - Audrey Perrotin
- Piramal Imaging Clinical Research and Development Berlin Germany
| | | | - Santiago Bullich
- AP‐HP–Hôpital Pitié Salpétrière Memory and Alzheimer Disease Institute IM2A Paris France
| | - Aleksandar Jovalekic
- Laboratoire d'Imagerie Biomédicale Sorbonne Universités, UPMC Univ Paris 06, Inserm U 1146, CNRS UMR 7371 Paris France
| | - Nicola Raffa
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Florence Pasquier
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Franck Semah
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Bruno Dubois
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Marie‐Odile Habert
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - David Wallon
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Mathieu Chastan
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Pierre Payoux
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Mathieu Ceccaldi
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Eric Guedj
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Mathieu Ceccaldi
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Olivier Felician
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Mira Didic
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Claude Gueriot
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Lejla Koric
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Radka Kletchkova‐Gantchev
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Eric Guedj
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Olivier Godefroy
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Daniela Andriuta
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Agnès Devendeville
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Diane Dupuis
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Ingrid Binot
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Mélanie Barbay
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Marc‐Etienne Meyer
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Véronique Moullard
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Eloi Magnin
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Ludivine Chamard
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Sophie Haffen
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Olivier Morel
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Clément Drouet
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Hatem Boulahdour
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Philippe Goas
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Solène Querellou‐Lefranc
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Vincent Sayette
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Julien Cogez
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Pierre Branger
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Denis Agostini
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Alain Manrique
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Olivier Rouaud
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Yannick Bejot
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Agnès Jacquin‐Piques
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Inna Dygai‐Cochet
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Alina Berriolo‐Riedinger
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Olivier Moreaud
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Mathilde Sauvee
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Céline Gallazzani Crépin
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Florence Pasquier
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Stéphanie Bombois
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Thibaud Lebouvier
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Marie‐Anne Mackowiak‐Cordoliani
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Vincent Deramecourt
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Adeline Rollin‐Sillaire
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Pascaline Cassagnaud‐Thuillet
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Yaohua Chen
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Franck Semah
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Grégory Petyt
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Pierre Krolak‐Salmon
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Denis Federico
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Keren Liora Danaila
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Yves Guilhermet
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Christophe Magnier
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Zaza Makaroff
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Isabelle Rouch
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Jing Xie
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Caroline Roubaud
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Marie‐Hélène Coste
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Kenny David
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Alain Sarciron
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Aziza Sediq Waissi
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Christian Scheiber
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Claire Houzard
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Audrey Gabelle‐Deloustal
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Karim Bennys
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Cecilia Marelli
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Lynda Touati
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Denis Mariano‐Goulart
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Delphine Verbizier‐Lonjon
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Thérèse Jonveaux
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Athanase Benetos
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Anna Kearney‐Schwartz
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Christine Perret‐Guillaume
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Antoine Verger
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Martine Vercelletto
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Claire Boutoleau‐Bretonniere
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Hélène Pouclet‐Courtemanche
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Nathalie Wagemann
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Amandine Pallardy
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Jacques Hugon
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Claire Paquet
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Julien Dumurgier
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Pascal Millet
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Mathieu Queneau
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Bruno Dubois
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Stéphane Epelbaum
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Marcel Levy
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | | | - Jean‐Luc Novella
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Yacine Jaidi
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Dimitri Papathanassiou
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | | | - Serge Belliard
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Anne Salmon
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Florence Lejeune
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Didier Hannequin
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - David Wallon
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Olivier Martinaud
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Aline Zarea
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Mathieu Chastan
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | | | - Claire Thalamas
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | | | | | - Fabienne Calvas
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Emilie Rigal
- ToNIC, Toulouse NeuroImaging Center Université de Toulouse, Inserm, UPS Toulouse France
| | - Pierre Payoux
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Anne Hitzel
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Julien Delrieu
- Neurology Department CHU de Rouen–Hôpital Charles Nicolle Rouen France
| | - Pierre‐Jean Ousset
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Françoise Lala
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Nathalie Sastre‐Hengan
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Andrew Stephens
- AP‐HM–Hôpital de la Timone, Neurology and Neuropsychology Department Aix Marseille University, Inserm, INS, Institut de Neurosciences des Systèmes Marseille France
| | - Eric Guedj
- AP‐HM–Hôpital de la Timone, Nuclear Medicine Department Aix‐Marseille University, CERIMED, CNRS, INT, Institut de Neurosciences de la Timone Marseille France
| | | |
Collapse
|
48
|
Engels MMA, Yu M, Stam CJ, Gouw AA, van der Flier WM, Scheltens P, van Straaten ECW, Hillebrand A. Directional information flow in patients with Alzheimer's disease. A source-space resting-state MEG study. Neuroimage Clin 2017; 15:673-681. [PMID: 28702344 PMCID: PMC5486371 DOI: 10.1016/j.nicl.2017.06.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/10/2017] [Accepted: 06/16/2017] [Indexed: 01/24/2023]
Abstract
In a recent magnetoencephalography (MEG) study, we found posterior-to-anterior information flow over the cortex in higher frequency bands in healthy subjects, with a reversed pattern in the theta band. A disruption of information flow may underlie clinical symptoms in Alzheimer's disease (AD). In AD, highly connected regions (hubs) in posterior areas are mostly disrupted. We therefore hypothesized that in AD the information flow from these hub regions would be disturbed. We used resting-state MEG recordings from 27 early-onset AD patients and 26 healthy controls. Using beamformer-based virtual electrodes, we estimated neuronal oscillatory activity for 78 cortical regions of interest (ROIs) and 12 subcortical ROIs of the AAL atlas, and calculated the directed phase transfer entropy (dPTE) as a measure of information flow between these ROIs. Group differences were evaluated using permutation tests and, for the AD group, associations between dPTE and general cognition or CSF biomarkers were determined using Spearman correlation coefficients. We confirmed the previously reported posterior-to-anterior information flow in the higher frequency bands in the healthy controls, and found it to be disturbed in the beta band in AD. Most prominently, the information flow from the precuneus and the visual cortex, towards frontal and subcortical structures, was decreased in AD. These disruptions did not correlate with cognitive impairment or CSF biomarkers. We conclude that AD pathology may affect the flow of information between brain regions, particularly from posterior hub regions, and that changes in the information flow in the beta band indicate an aspect of the pathophysiological process in AD.
Collapse
Affiliation(s)
- M M A Engels
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.
| | - M Yu
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, The Netherlands
| | - C J Stam
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, The Netherlands
| | - A A Gouw
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, The Netherlands
| | - W M van der Flier
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; Department of Epidemiology and Biostatistics, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Ph Scheltens
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - E C W van Straaten
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, The Netherlands
| | - A Hillebrand
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
49
|
Mattsson N, Lönneborg A, Boccardi M, Blennow K, Hansson O. Clinical validity of cerebrospinal fluid Aβ42, tau, and phospho-tau as biomarkers for Alzheimer's disease in the context of a structured 5-phase development framework. Neurobiol Aging 2017; 52:196-213. [DOI: 10.1016/j.neurobiolaging.2016.02.034] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/09/2016] [Accepted: 02/10/2016] [Indexed: 01/01/2023]
|
50
|
Dekker AD, Fortea J, Blesa R, De Deyn PP. Cerebrospinal fluid biomarkers for Alzheimer's disease in Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2017; 8:1-10. [PMID: 28413821 PMCID: PMC5384293 DOI: 10.1016/j.dadm.2017.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Down syndrome (DS), present in nearly six million people, is associated with an extremely high risk to develop Alzheimer's disease (AD). Amyloid-β and tau pathology are omnipresent from age 40 years onward, but clinical symptoms do not appear in all DS individuals. Dementia diagnostics is complex in this population, illustrating the great need for predictive biomarkers. Although blood biomarkers have not yet proven useful, cerebrospinal fluid (CSF) biomarkers (low amyloid-β42, high t-tau, and high p-tau) effectively contribute to AD diagnoses in the general population and are increasingly used in clinical practice. Surprisingly, CSF biomarkers have been barely evaluated in DS. Breaking the taboo on CSF analyses would finally allow for the elucidation of its utility in (differential) diagnoses and staging of disease severity. A sensitive and specific biomarker profile for AD in DS would be of paramount importance to daily care, adaptive caregiving, and specific therapeutic interventions.
Collapse
Affiliation(s)
- Alain D. Dekker
- Department of Neurology and Alzheimer Research Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Juan Fortea
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Down Medical Center, Catalan Down Syndrome Foundation, Barcelona, Spain
| | - Rafael Blesa
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Peter P. De Deyn
- Department of Neurology and Alzheimer Research Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| |
Collapse
|