1
|
Cash DM, Morgan KE, O'Connor A, Veale TD, Malone IB, Poole T, Benzinger TL, Gordon BA, Ibanez L, Li Y, Llibre-Guerra JJ, McDade E, Wang G, Chhatwal JP, Day GS, Huey E, Jucker M, Levin J, Niimi Y, Noble JM, Roh JH, Sánchez-Valle R, Schofield PR, Bateman RJ, Frost C, Fox NC. Sample size estimates for biomarker-based outcome measures in clinical trials in autosomal dominant Alzheimer's disease. J Prev Alzheimers Dis 2025:100133. [PMID: 40118731 DOI: 10.1016/j.tjpad.2025.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION Alzheimer disease (AD)-modifying therapies are approved for treatment of early-symptomatic AD. Autosomal dominant AD (ADAD) provides a unique opportunity to test therapies in presymptomatic individuals. METHODS Using data from the Dominantly Inherited Alzheimer Network (DIAN), sample sizes for clinical trials were estimated for various cognitive, imaging, and CSF outcomes. Sample sizes were computed for detecting a reduction of either absolute levels of AD-related pathology (amyloid, tau) or change over time in neurodegeneration (atrophy, hypometabolism, cognitive change). RESULTS Biomarkers measuring amyloid and tau pathology had required sample sizes below 200 participants per arm (examples CSF Aβ42/40: 47[95 %CI 25,104], cortical PIB 49[28,99], CSF p-tau181 74[48,125]) for a four-year trial in presymptomatic individuals (CDR=0) to have 80 % power (5 % statistical significance) to detect a 25 % reduction in absolute levels of pathology, allowing 40 % dropout. For cognitive, MRI, and FDG, it was more appropriate to detect a 50 % reduction in rate of change. Sample sizes ranged from 250 to 900 (examples hippocampal volume: 338[131,2096], cognitive composite: 326[157,1074]). MRI, FDG and cognitive outcomes had lower sample sizes when including indivduals with mild impairment (CDR=0.5 and 1) as well as presymptomatic individuals (CDR=0). DISCUSSION Despite the rarity of ADAD, presymptomatic clinical trials with feasible sample sizes given the number of cases appear possible.
Collapse
Affiliation(s)
- David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK; UK Dementia Research Institute, 6th Floor, Maple House, Tottenham Court Road, London W1T 7NF, UK.
| | - Katy E Morgan
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Antoinette O'Connor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Thomas D Veale
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Ian B Malone
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Teresa Poole
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Tammie Ls Benzinger
- Department of Radiology. Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Brian A Gordon
- Department of Radiology. Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., Suite 200, St. Louis, MO 63108 USA
| | - Laura Ibanez
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA; Department of Psychiarty, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Yan Li
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Jorge J Llibre-Guerra
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Guoqiao Wang
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Jasmeer P Chhatwal
- Brigham and Women's Hospital, Massachusetts General Hospital; Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA
| | - Edward Huey
- Alpert Medical School of Brown University, Department of Psychiatry and Human Behavior, 222 Richmond St., Providence, RI 02903, USA
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller Strasse 27, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, 72076 Tübingen, Germany
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, Marchioninistr. 15 D-81377, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Yoshiki Niimi
- Unit for early and exploratory clinical development, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, GH Sergievksy Center, Columbia University, 710W 168th St #3, New York, NY 10032, USA
| | - Jee Hoon Roh
- Departments of Neurology and Physiology, Korea University Anam Hospital, Korea University College of Medicine, 73 goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic Of Korea
| | - Racquel Sánchez-Valle
- Alzheimer's disease and other cognitive disorders group. Hospital Clínic de Barcelona. FRCB-IDIBAPS. University of Barcelona, Carrer de Villarroel, 170, L'Eixample, 08036 Barcelona, Spain
| | - Peter R Schofield
- Neuroscience Research Australia, Margarete Ainsworth Building Barker Street, Randwick NSW 2031 Australia; School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW 2052 Australia
| | - Randall J Bateman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., Suite 200, St. Louis, MO 63108 USA; Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA; Hope Center for Neurological Disorders, Washington University in St Louis, 4370 Duncan Ave., St. Louis, MO 63110, USA
| | - Chris Frost
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK; UK Dementia Research Institute, 6th Floor, Maple House, Tottenham Court Road, London W1T 7NF, UK
| |
Collapse
|
2
|
Cash DM, Morgan KE, O’Connor A, Veale TD, Malone IB, Poole T, Benzinger TLS, Gordon BA, Ibanez L, Li Y, Llibre-Guerra JJ, McDade E, Wang G, Chhatwal JP, Day GS, Huey E, Jucker M, Levin J, Niimi Y, Noble JM, Roh JH, Sánchez-Valle R, Schofield PR, Bateman RJ, Frost C, Fox NC. Sample size estimates for biomarker-based outcome measures in clinical trials in autosomal dominant Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.11.12.24316919. [PMID: 39606328 PMCID: PMC11601746 DOI: 10.1101/2024.11.12.24316919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
INTRODUCTION Alzheimer disease (AD)-modifying therapies are approved for treatment of early-symptomatic AD. Autosomal dominant AD (ADAD) provides a unique opportunity to test therapies in presymptomatic individuals. METHODS Using data from the Dominantly Inherited Alzheimer Network (DIAN), sample sizes for clinical trials were estimated for various cognitive, imaging, and CSF outcomes. Sample sizes were computed for detecting a reduction of either absolute levels of AD-related pathology (amyloid, tau) or change over time in neurodegeneration (atrophy, hypometabolism, cognitive change). RESULTS Biomarkers measuring amyloid and tau pathology had required sample sizes below 200 participants per arm (examples CSF Aβ42/40: 47[95%CI 25,104], cortical PIB 49[28,99], CSF p-tau181 74[48,125]) for a four-year trial in presymptomatic individuals (CDR=0) to have 80% power (5% statistical significance) to detect a 25% reduction in absolute levels of pathology, allowing 40% dropout. For cognitive, MRI, and FDG, it was more appropriate to detect a 50% reduction in rate of change. Sample sizes ranged from 250-900 (examples hippocampal volume: 338[131,2096], cognitive composite: 326[157,1074]). MRI, FDG and cognitive outcomes had lower sample sizes when including indivduals with mild impairment (CDR=0.5 and 1) as well as presymptomatic individuals (CDR=0). DISCUSSION Despite the rarity of ADAD, presymptomatic clinical trials with feasible sample sizes given the number of cases appear possible.
Collapse
Affiliation(s)
- David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
- UK Dementia Research Institute, 6th Floor, Maple House, Tottenham Court Road, London W1T 7NF, UK
| | - Katy E Morgan
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Antoinette O’Connor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Thomas D Veale
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Ian B Malone
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Teresa Poole
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Tammie LS Benzinger
- Department of Radiology. Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Brian A Gordon
- Department of Radiology. Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., Suite 200, St. Louis, MO 63108 USA
| | - Laura Ibanez
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
- Department of Psychiarty, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Yan Li
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Jorge J. Llibre-Guerra
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Guoqiao Wang
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Jasmeer P Chhatwal
- Brigham and Women’s Hospital; Massachusetts General Hospital; Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA
| | - Edward Huey
- Alpert Medical School of Brown University, Department of Psychiatry and Human Behavior, 222 Richmond St., Providence, RI 02903, USA
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, 72076 Tübingen, Germany
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, Marchioninistr. 15 D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17,81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Yoshiki Niimi
- Unit for early and exploratory clinical development, The UniVersity of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, GH Sergievksy Center, Columbia University, 710 W 168th St #3, New York, NY 10032, USA
| | - Jee Hoon Roh
- Departments of Neurology and Physiology, Korea University Anam Hospital, Korea University College of Medicine, 73 goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic Of Korea
| | - Racquel Sánchez-Valle
- Alzheimer’s disease and other cognitive disorders group. Hospital Clínic de Barcelona. FRCB-IDIBAPS. University of Barcelona, Carrer de Villarroel, 170, L’Eixample, 08036 Barcelona, Spain
| | - Peter R Schofield
- Neuroscience Research Australia, Margarete Ainsworth Building Barker Street, Randwick NSW 2031 Australia
- School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW 2052 Australia
| | - Randall J Bateman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., Suite 200, St. Louis, MO 63108 USA
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
- Hope Center for Neurological Disorders, Washington University in St Louis, 4370 Duncan Ave., St. Louis, MO 63110, USA
| | - Chris Frost
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
- UK Dementia Research Institute, 6th Floor, Maple House, Tottenham Court Road, London W1T 7NF, UK
| | | |
Collapse
|
3
|
Pezzoli S, Giorgio J, Chen X, Ward TJ, Harrison TM, Jagust WJ. Cognitive aging outcomes are related to both tau pathology and maintenance of cingulate cortex structure. Alzheimers Dement 2025; 21:e14515. [PMID: 39807642 PMCID: PMC11848174 DOI: 10.1002/alz.14515] [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: 08/12/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Successful cognitive aging is related to both maintaining brain structure and avoiding Alzheimer's disease (AD) pathology, but how these factors interplay is unclear. METHODS A total of 109 cognitively normal older adults (70+ years old) underwent amyloid beta (Aβ) and tau positron emission tomography (PET) imaging, structural magnetic resonance imaging (MRI), and cognitive testing. Cognitive aging was quantified using the cognitive age gap (CAG), subtracting chronological age from predicted cognitive age. RESULTS Lower CAG (younger cognitive age) was related to slower decline in episodic memory, multi-domain cognition, and atrophy of the midcingulate cortex (MCC). Lower entorhinal cortical tau was linked to slower decline in episodic memory, multi-domain cognition, and hippocampal atrophy. DISCUSSION These results suggest that aging outcomes may be influenced by two independent pathways: one associated with tau accumulation, affecting primarily memory and hippocampal atrophy, and another involving tau-independent structural preservation of the MCC, benefiting multi-domain cognition over time. HIGHLIGHTS Younger cognitive age (lower cognitive age gap [CAG]) is related to slower cognitive decline. Lower CAG is linked to slower midcingulate cortex (MCC) atrophy. Reduced tau in the entorhinal cortex is related to less hippocampal atrophy and cognitive decline. Structural preservation of the MCC benefits multi-domain cognition over time. Two independent pathways influence cognitive aging: tau accumulation and MCC preservation.
Collapse
Affiliation(s)
- Stefania Pezzoli
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Joseph Giorgio
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
- School of Psychological Sciences, College of EngineeringScience and the EnvironmentUniversity of NewcastleNewcastleNew South WalesAustralia
| | - Xi Chen
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
- Department of PsychologyStony Brook UniversityStony BrookNew YorkUSA
| | - Tyler J. Ward
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | | | - William J. Jagust
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| |
Collapse
|
4
|
Barisano G, Iv M, Choupan J, Hayden-Gephart M. Robust, fully-automated assessment of cerebral perivascular spaces and white matter lesions: a multicentre MRI longitudinal study of their evolution and association with risk of dementia and accelerated brain atrophy. EBioMedicine 2025; 111:105523. [PMID: 39721217 PMCID: PMC11732520 DOI: 10.1016/j.ebiom.2024.105523] [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: 07/29/2024] [Revised: 11/03/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Perivascular spaces (PVS) on brain MRI are surrogates for small parenchymal blood vessels and their perivascular compartment, and may relate to brain health. However, it is unknown whether PVS can predict dementia risk and brain atrophy trajectories in participants without dementia, as longitudinal studies on PVS are scarce and current methods for PVS assessment lack robustness and inter-scanner reproducibility. METHODS We developed a robust algorithm to automatically assess PVS count and size on clinical MRI, and investigated 1) their relationship with dementia risk and brain atrophy in participants without dementia, 2) their longitudinal evolution, and 3) their potential use as a screening tool in simulated clinical trials. We analysed 46,478 clinical measurements of cognitive functioning and 20,845 brain MRI scans from 10,004 participants (71.1 ± 9.7 years-old, 56.6% women) from three publicly available observational studies on ageing and dementia (the Alzheimer's Disease Neuroimaging Initiative, the National Alzheimer's Coordinating Centre database, and the Open Access Series of Imaging Studies). Clinical and MRI data collected between 2004 and 2022 were analysed with consistent methods, controlling for confounding factors, and combined using mixed-effects models. FINDINGS Our fully-automated method for PVS assessment showed excellent inter-scanner reproducibility (intraclass correlation coefficients >0.8). Fewer PVS and larger PVS diameter at baseline predicted higher dementia risk and accelerated brain atrophy. Longitudinal trajectories of PVS markers differed significantly in participants without dementia who converted to dementia compared with non-converters. In simulated placebo-controlled trials for treatments targeting cognitive decline, screening out participants at low risk of dementia based on our PVS markers enhanced the power of the trial independently of Alzheimer's disease biomarkers. INTERPRETATION These robust cerebrovascular markers predict dementia risk and brain atrophy and may improve risk-stratification of patients, potentially reducing cost and increasing throughput of clinical trials to combat dementia. FUNDING US National Institutes of Health.
Collapse
Affiliation(s)
| | - Michael Iv
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Jeiran Choupan
- Laboratory of Neuro Imaging, University of Southern California, Los Angeles, CA, USA; NeuroScope Inc., New York, NY, USA
| | | |
Collapse
|
5
|
De Meyer S, Blujdea ER, Schaeverbeke JM, Adamczuk K, Vandenberghe R, Poesen K, Teunissen CE. Serum biomarkers as prognostic markers for Alzheimer's disease in a clinical setting. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70071. [PMID: 39886322 PMCID: PMC11780118 DOI: 10.1002/dad2.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 02/01/2025]
Abstract
INTRODUCTION Blood-based glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL), and phosphorylated tau (pTau) have shown promising prognostic potential in Alzheimer's disease (AD), but their applicability in clinical settings where comorbidities are prevalent remains uncertain. METHODS Simoa assays quantified GFAP, NfL, and pTau181 in retrospectively retrieved prediagnostic serum samples from 102 AD patients and 21 non-AD controls. RESULTS Higher serum GFAP levels predicted earlier clinical presentation and faster subsequent Mini-Mental State Examination decline in AD patients. Serum NfL levels were increased in patients with arterial hypertension (AHT), kidney dysfunction, and a history of stroke and only demonstrated predictive value for time to clinical AD presentation after adjustment for these comorbidities. Serum pTau181 instability during long-term storage at -20°C prevented its prognostic evaluation in retrospectively retrieved serum samples. DISCUSSION Serum GFAP is a robust prognostic marker for AD progression, whereas NfL is impacted by various comorbidities, which complicates the interpretation of its prognostic value. Highlights Serum GFAP levels predict time to clinical AD presentation.Serum NfL levels are increased by hypertension, kidney disease, and stroke history.Prognostic value of serum NfL in AD is only evident after comorbidity correction.Serum levels of GFAP, but not NfL, increase over time within prediagnostic AD stages.
Collapse
Affiliation(s)
- Steffi De Meyer
- Laboratory for Molecular Neurobiomarker ResearchDepartment of NeurosciencesLeuven Brain Institute, KU LeuvenLeuvenBelgium
- Laboratory for Cognitive NeurologyDepartment of NeurosciencesLeuven Brain Institute, KU LeuvenLeuvenBelgium
| | - Elena R. Blujdea
- Neurochemistry LaboratoryDepartment of Laboratory MedicineAmsterdam UMCAmsterdamThe Netherlands
| | - Jolien M. Schaeverbeke
- Laboratory for Cognitive NeurologyDepartment of NeurosciencesLeuven Brain Institute, KU LeuvenLeuvenBelgium
- Laboratory for NeuropathologyDepartment of Imaging and PathologyLeuven Brain Institute, KU LeuvenLeuvenBelgium
| | - Katarzyna Adamczuk
- Laboratory for Cognitive NeurologyDepartment of NeurosciencesLeuven Brain Institute, KU LeuvenLeuvenBelgium
| | - Rik Vandenberghe
- Laboratory for Cognitive NeurologyDepartment of NeurosciencesLeuven Brain Institute, KU LeuvenLeuvenBelgium
- Neurology DepartmentUZ LeuvenLeuvenBelgium
| | - Koen Poesen
- Laboratory for Molecular Neurobiomarker ResearchDepartment of NeurosciencesLeuven Brain Institute, KU LeuvenLeuvenBelgium
- Laboratory Medicine DepartmentUZ LeuvenLeuvenBelgium
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Laboratory MedicineAmsterdam UMCAmsterdamThe Netherlands
| |
Collapse
|
6
|
Hartz SM, Schindler SE, Streitz ML, Moulder KL, Mozersky J, Wang G, Xiong C, Morris JC. Assessing the clinical meaningfulness of slowing CDR-SB progression with disease-modifying therapies for Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2025; 11:e70033. [PMID: 39949872 PMCID: PMC11822626 DOI: 10.1002/trc2.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 02/16/2025]
Abstract
INTRODUCTION For many patients and caregivers, a major goal of disease-modifying treatments (DMTs) for Alzheimer's disease (AD) dementia is to extend independence in instrumental and basic activities of daily living (IADLs and BADLs). The goal of this study was to estimate the effect of treatments on the time remaining independent in IADLs and BADLs. METHODS Participants at the Knight Alzheimer Disease Research Center (Knight ADRC) who met eligibility criteria for recent DMT trials were studied: age ≥60 years at baseline, clinical diagnosis of very mild or mild AD dementia (global Clinical Dementia Rating [CDR] score 0.5 or 1), biomarker confirmation of amyloid pathology, and at least one follow-up CDR assessment within 5 years. For IADLs, a subset of the Functional Assessment Questionnaire (FAQ) was examined that rated the degree of independence in the following: paying bills, driving, remembering medications and appointments, and preparing meals. For BADLs, the Personal Care domain of the CDR was used. Mixed-effects logistic and ordinal regression models were used to examine the relationship between CDR Sum of Boxes (CDR-SB) and the individual functional outcomes and their components. The change in CDR-SB over time was estimated with linear mixed-effects models. RESULTS A total of 282 participants were followed for an average of 2.9 years (standard deviation [SD] 1.3 years). For 50% of individuals, loss of independence in IADLs occurred at CDR-SB >4.5 and in BADLs at CDR-SB >11.5. For individuals with a baseline CDR-SB = 2, treatment with lecanemab would extend independence in IADLs for 10 months (95% confidence interval [CI] 4-18 months) and treatment with donanemab in the low/medium tau group would extend independence in IADLs by 13 months (95% CI 6-24 months). DISCUSSION Independence in ADLs can be related to CDR-SB and used to demonstrate the effect of AD treatments in extending the time of independent function, a meaningful outcome for patients and their families. Highlights We estimated time to loss of independence for people with AD dementiaEstimating time to loss of independence can help with clinical decision-makingDisease-modifying treatments for AD dementia can extend independence.
Collapse
Affiliation(s)
- Sarah M. Hartz
- Department of PsychiatryWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - Suzanne E. Schindler
- Department of NeurologyWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - Marissa L. Streitz
- Department of NeurologyWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - Krista L. Moulder
- Department of NeurologyWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - Jessica Mozersky
- Department of MedicineWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - Guoqiao Wang
- Department of NeurologyWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
- Division of BiostatisticsWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - Chengjie Xiong
- Division of BiostatisticsWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| | - John C. Morris
- Department of NeurologyWashington University School of Medicine in St. LouisSt. LouisMissouriUSA
| |
Collapse
|
7
|
Parul, Singh A, Shukla S. Novel techniques for early diagnosis and monitoring of Alzheimer's disease. Expert Rev Neurother 2025; 25:29-42. [PMID: 39435792 DOI: 10.1080/14737175.2024.2415985] [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: 05/30/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common neurodegenerative disorder, which is characterized by a progressive loss of cognitive functions. The high prevalence, chronicity, and multimorbidity are very common in AD, which significantly impair the quality of life and functioning of patients. Early detection and accurate diagnosis of Alzheimer's disease (AD) can stop the illness from progressing thereby postponing its symptoms. Therefore, for the early diagnosis and monitoring of AD, more sensitive, noninvasive, straightforward, and affordable screening tools are needed. AREAS COVERED This review summarizes the importance of early detection methods and novel techniques for Alzheimer's disease diagnosis that can be used by healthcare professionals. EXPERT OPINION Early diagnosis assists the patient and caregivers to understand the problem establishing reasonable goals and making future plans together. Early diagnosis techniques not only help in monitoring disease progression but also provide crucial information for the development of novel therapeutic targets. Researchers can plan to potentially alleviate symptoms or slow down the progression of Alzheimer's disease by identifying early molecular changes and targeting altered pathways.
Collapse
Affiliation(s)
- Parul
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Animesh Singh
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shubha Shukla
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
8
|
Weinstein AM, Fang F, Chang CCH, Cohen A, Lopresti BJ, Laymon CM, Nadkarni NK, Aizenstein HJ, Villemagne VL, Kamboh MI, Shaaban CE, Gogniat MA, Wu M, Karikari TK, Ganguli M, Snitz BE. Multimodal neuroimaging biomarkers and subtle cognitive decline in a population-based cohort without dementia. J Alzheimers Dis 2025; 103:570-581. [PMID: 39702989 PMCID: PMC11798718 DOI: 10.1177/13872877241303926] [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] [Indexed: 12/21/2024]
Abstract
BACKGROUND The relationship between subtle cognitive decline and Alzheimer's disease (AD) pathology as measured by biomarkers in settings outside of specialty memory clinics is not well characterized. OBJECTIVE To investigate how subtle longitudinal cognitive decline relates to neuroimaging biomarkers in individuals drawn from a population-based study in an economically depressed, small-town area in southwestern Pennsylvania, USA. METHODS A subset of participants without dementia (N = 115, age 76.53 years ± 6.25) from the Monongahela Youghiogheny Healthy Aging Team (MYHAT) study completed neuroimaging including magnetic resonance imaging (MRI) measures of AD-signature region cortical thickness and white matter hyperintensities (WMH), Pittsburgh compound B (PiB)-positron emission tomography (PET) for amyloid-β (Aβ) deposition, and [18F]AV-1451-PET for tau deposition. Neuropsychological evaluations were completed at multiple timepoints up to 11 years prior to neuroimaging. Aβ positivity was determined using a regional approach. We used linear mixed models to examine neuroimaging biomarker associations with retrospective cognitive slopes in five domains and a global cognitive composite. RESULTS Among Aβ(+) participants (38%), there were associations between (i) tau Braak III/IV and language decline (p < 0.05), (ii) cortical thickness and both memory decline (p < 0.001) and global cognitive decline (p < 0.01), and (iii) WMH and decline in executive function (p < 0.05) and global cognition (p < 0.05). Among Aβ(-) participants, there was an association between tau Braak III/IV and decline on tests of attention/psychomotor speed (p < 0.05). CONCLUSIONS These findings confirm an Aβ-dependent early AD biomarker pathway, and suggest a possible Aβ-independent, non-AD process underlying subtle cognitive decline in a population-based sample of older adults without dementia.
Collapse
Affiliation(s)
- Andrea M Weinstein
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
| | - Fang Fang
- Research & Infrastructure Service Enterprise (RISE), Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, 23501 USA
| | - Chung-Chou H Chang
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261 USA
| | - Ann Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
| | - Charles M Laymon
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
- Department of Bioengineering, University of Pittsburgh School of Engineering, Pittsburgh, PA, 15260 USA
| | - Neelesh K Nadkarni
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh PA, 15213 USA
| | - Howard J Aizenstein
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
- Department of Bioengineering, University of Pittsburgh School of Engineering, Pittsburgh, PA, 15260 USA
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, 15261 USA
| | - C. Elizabeth Shaaban
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, 15261 USA
| | - Marissa A. Gogniat
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh PA, 15213 USA
| | - Minjie Wu
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
| | - Mary Ganguli
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213 USA
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, 15261 USA
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh PA, 15213 USA
| |
Collapse
|
9
|
Aljuhani M, Ashraf A, Edison P. Evaluating clinical meaningfulness of anti-β-amyloid therapies amidst amyloid-related imaging abnormalities concern in Alzheimer's disease. Brain Commun 2024; 6:fcae435. [PMID: 39703326 PMCID: PMC11656198 DOI: 10.1093/braincomms/fcae435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/28/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024] Open
Abstract
Alzheimer's disease is the most prevalent form of dementia in the elderly, which is clinically characterized by a gradual and progressive deterioration of cognitive functions. The central and early role of β-amyloid in the pathogenesis of Alzheimer's disease is supported by a plethora of studies including genetic analyses, biomarker research and genome-wide association studies in both familial (early-onset) and sporadic (late-onset) forms of Alzheimer's. Monoclonal antibodies directed against β-amyloid demonstrate slowing of the clinical deterioration of patients with early Alzheimer's disease. Aducanumab, lecanemab and donanemab clinical trials showed slowing of Alzheimer's disease progression on composite scores by 25-40% based on the measure used. Anti-β-amyloid antibodies can cause side effects of bleeding and swelling in the brain, called amyloid-related imaging abnormalities. Amyloid-related imaging abnormalities typically occur early in treatment and are often asymptomatic, and though in rare cases, they can lead to serious or life-threatening events. The aim of this review is to evaluate the clinical meaningfulness of anti-β-amyloid therapies amidst amyloid-related imaging abnormalities concern in Alzheimer's disease.
Collapse
Affiliation(s)
- Manal Aljuhani
- Radiological Science and Medical Imaging Department, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Azhaar Ashraf
- Division of Neurology, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Paul Edison
- Division of Neurology, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London W12 0NN, UK
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Wales CF24 4HQ, UK
| |
Collapse
|
10
|
Park C, Joo G, Roh M, Shin S, Yum S, Yeo NY, Park SW, Jang JW, Im H. Predicting the Progression of Mild Cognitive Impairment to Alzheimer's Dementia Using Recurrent Neural Networks With a Series of Neuropsychological Tests. J Clin Neurol 2024; 20:478-486. [PMID: 39227330 PMCID: PMC11372213 DOI: 10.3988/jcn.2023.0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 04/30/2024] [Accepted: 05/24/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND AND PURPOSE The prevalence of Alzheimer's dementia (AD) is increasing as populations age, causing immense suffering for patients, families, and communities. Unfortunately, no treatments for this neurodegenerative disease have been established. Predicting AD is therefore becoming more important, because early diagnosis is the best way to prevent its onset and delay its progression. METHODS Mild cognitive impairment (MCI) is the stage between normal cognition and AD, with large variations in its progression. The disease can be effectively managed by accurately predicting the probability of MCI progressing to AD over several years. In this study we used the Alzheimer's Disease Neuroimaging Initiative dataset to predict the progression of MCI to AD over a 3-year period from baseline. We developed and compared various recurrent neural network (RNN) models to determine the predictive effectiveness of four neuropsychological (NP) tests and magnetic resonance imaging (MRI) data at baseline. RESULTS The experimental results confirmed that the Preclinical Alzheimer's Cognitive Composite score was the most effective of the four NP tests, and that the prediction performance of the NP tests improved over time. Moreover, the gated recurrent unit model exhibited the best performance among the prediction models, with an average area under the receiver operating characteristic curve of 0.916. CONCLUSIONS Timely prediction of progression from MCI to AD can be achieved using a series of NP test results and an RNN, both with and without using the baseline MRI data.
Collapse
Affiliation(s)
- Chaeyoon Park
- Graduate School of Data Science, Kangwon National University, Chuncheon, Korea
| | - Gihun Joo
- Interdisciplinary Graduate Program in Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea
| | - Minji Roh
- Interdisciplinary Graduate Program in Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea
| | - Seunghun Shin
- Interdisciplinary Graduate Program in Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea
| | - Sujin Yum
- Interdisciplinary Graduate Program in Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea
- Department of Neurology, Kangwon National University Hospital, Chuncheon, Korea
| | - Na Young Yeo
- Interdisciplinary Graduate Program in Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea
- Department of Neurology, Kangwon National University Hospital, Chuncheon, Korea
| | - Sang Won Park
- Department of Neurology, Kangwon National University Hospital, Chuncheon, Korea
- Department of Medical Informatics, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Jae-Won Jang
- Interdisciplinary Graduate Program in Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea
- Department of Neurology, Kangwon National University Hospital, Chuncheon, Korea
- Department of Medical Informatics, School of Medicine, Kangwon National University, Chuncheon, Korea
- Department of Convergence Security, Kangwon National University, Chuncheon, Korea.
| | - Hyeonseung Im
- Graduate School of Data Science, Kangwon National University, Chuncheon, Korea
- Interdisciplinary Graduate Program in Medical Bigdata Convergence, Kangwon National University, Chuncheon, Korea
- Department of Convergence Security, Kangwon National University, Chuncheon, Korea
- Department of Computer Science and Engineering, Kangwon National University, Chuncheon, Korea.
| |
Collapse
|
11
|
Abdul Manap AS, Almadodi R, Sultana S, Sebastian MG, Kavani KS, Lyenouq VE, Shankar A. Alzheimer's disease: a review on the current trends of the effective diagnosis and therapeutics. Front Aging Neurosci 2024; 16:1429211. [PMID: 39185459 PMCID: PMC11341404 DOI: 10.3389/fnagi.2024.1429211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
The most prevalent cause of dementia is Alzheimer's disease. Cognitive decline and accelerating memory loss characterize it. Alzheimer's disease advances sequentially, starting with preclinical stages, followed by mild cognitive and/or behavioral impairment, and ultimately leading to Alzheimer's disease dementia. In recent years, healthcare providers have been advised to make an earlier diagnosis of Alzheimer's, prior to individuals developing Alzheimer's disease dementia. Regrettably, the identification of early-stage Alzheimer's disease in clinical settings can be arduous due to the tendency of patients and healthcare providers to disregard symptoms as typical signs of aging. Therefore, accurate and prompt diagnosis of Alzheimer's disease is essential in order to facilitate the development of disease-modifying and secondary preventive therapies prior to the onset of symptoms. There has been a notable shift in the goal of the diagnosis process, transitioning from merely confirming the presence of symptomatic AD to recognizing the illness in its early, asymptomatic phases. Understanding the evolution of disease-modifying therapies and putting effective diagnostic and therapeutic management into practice requires an understanding of this concept. The outcomes of this study will enhance in-depth knowledge of the current status of Alzheimer's disease's diagnosis and treatment, justifying the necessity for the quest for potential novel biomarkers that can contribute to determining the stage of the disease, particularly in its earliest stages. Interestingly, latest clinical trial status on pharmacological agents, the nonpharmacological treatments such as behavior modification, exercise, and cognitive training as well as alternative approach on phytochemicals as neuroprotective agents have been covered in detailed.
Collapse
Affiliation(s)
- Aimi Syamima Abdul Manap
- Department of Biomedical Science, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Reema Almadodi
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | - Shirin Sultana
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | | | | | - Vanessa Elle Lyenouq
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| | - Aravind Shankar
- Faculty of Pharmacy and Biomedical Sciences, MAHSA University, Selangor, Malaysia
| |
Collapse
|
12
|
Hartz SM, Schindler SE, Streitz ML, Moulder KL, Mozersky J, Wang G, Xiong C, Morris JC. Assessing the clinical meaningfulness of slowing CDR-SB progression with disease-modifying therapies for Alzheimer disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.16.24310511. [PMID: 39108536 PMCID: PMC11302622 DOI: 10.1101/2024.07.16.24310511] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
INTRODUCTION For many patients and caregivers, a major goal of disease-modifying treatments (DMT) for Alzheimer disease (AD) dementia is to extend independence in instrumental and basic activities of daily living (IADLs and BADLs). The goal of this study was to estimate the effect of treatments on the time remaining independent in IADLs and BADLs. METHODS Participants at the Knight Alzheimer Disease Research Center were selected who were potentially eligible for recent DMT trials: age ≥ 60 years at baseline, clinical diagnosis of very mild or mild AD dementia (global Clinical Dementia Rating® (CDR®) score 0.5 or 1), biomarker confirmation of amyloid pathology, and at least one follow-up CDR assessment within 5 years. For IADLs, a subset of the Functional Assessment Questionnaire (FAQ) was examined that rated the degree of independence in the following: paying bills, driving, remembering medications and appointments, and preparing meals. For BADLs, the Personal Care domain of the CDR was used. Mixed-effects logistic and ordinal regression models were used to examine the relationship between CDR Sum Boxes (CDR-SB) and the individual functional outcomes and their components. The change in CDR-SB over time was estimated with linear mixed effects models. RESULTS 282 participants were followed for an average of 2.9 years (SD 1.3 years). For 50% of individuals, loss of independence in IADLs occurred at CDR-SB>4.5 and in BADLs at CDR-SB>11.5. For individuals with a baseline CDR-SB=2, treatment with lecanemab would extend independence in IADLs for 10 months (95% CI 4-18 months) and treatment with donanemab in the low/medium tau group would extend independence in IADLs by 13 months (95% CI 6-24 months). DISCUSSION Independence in ADLs can be related to CDR-SB and used to demonstrate the effect of AD treatments in extending the time of independent function, a meaningful outcome for patients and their families.
Collapse
|
13
|
Bhalala OG, Thompson J, Watson R, Yassi N. Contextualising the benefits and risks of anti-amyloid therapy for patients with Alzheimer disease and their care team. Med J Aust 2024; 221:78-82. [PMID: 38894659 DOI: 10.5694/mja2.52359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/20/2024] [Indexed: 06/21/2024]
Affiliation(s)
- Oneil G Bhalala
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
- Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC
| | - Jane Thompson
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
| | - Rosie Watson
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
- Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC
| | - Nawaf Yassi
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
- Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC
| |
Collapse
|
14
|
Wang W, Huang J, Qian S, Zheng Y, Yu X, Jiang T, Ai R, Hou J, Ma E, Cai J, He H, Wang X, Xie C. Amyloid-β but not tau accumulation is strongly associated with longitudinal cognitive decline. CNS Neurosci Ther 2024; 30:e14860. [PMID: 39014268 PMCID: PMC11251873 DOI: 10.1111/cns.14860] [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: 12/13/2023] [Revised: 06/11/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) pathology is featured by the extracellular accumulation of amyloid-β (Aβ) plaques and intracellular tau neurofibrillary tangles in the brain. We studied whether Aβ and tau accumulation are independently associated with future cognitive decline in the AD continuum. METHODS Data were acquired from the Alzheimer's Disease Neuroimaging Initiative (ADNI) public database. A total of 1272 participants were selected based on the availability of Aβ-PET and CSF tau at baseline and of those 777 participants with follow-up visits. RESULTS We found that Aβ-PET and CSF tau pathology were related to cognitive decline across the AD clinical spectrum, both as potential predictors for dementia progression. Among them, Aβ-PET (A + T- subjects) is an independent reliable predictor of longitudinal cognitive decline in terms of ADAS-13, ADNI-MEM, and MMSE scores rather than tau pathology (A - T+ subjects), indicating tau accumulation is not closely correlated with future cognitive impairment without being driven by Aβ deposition. Of note, a high percentage of APOE ε4 carriers with Aβ pathology (A+) develop poor memory and learning capacity. Interestingly, this condition is not recurrence in terms of the ADNI-MEM domain when adding APOE ε4 status. Finally, the levels of Aβ-PET SUVR related to glucose hypometabolism more strongly in subjects with A + T- than A - T+ both happen at baseline and longitudinal changes. CONCLUSIONS In conclusion, Aβ-PET alone without tau pathology (A + T-) measure is an independent reliable predictor of longitudinal cognitive decline but may nonetheless forecast different status of dementia progression. However, tau accumulation alone without Aβ pathology background (A - T+) was not enough to be an independent predictor of cognitive worsening.
Collapse
Affiliation(s)
- Wenwen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated HospitalYuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jiani Huang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Shuangjie Qian
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yi Zheng
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xinyue Yu
- Alberta InstituteWenzhou Medical UniversityWenzhouZhejiangChina
| | - Tao Jiang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ruixue Ai
- Department of Clinical Molecular Biology, Akershus University HospitalUniversity of OsloLørenskogNorway
| | - Jialong Hou
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Enzi Ma
- Department of NeurologyTraditional Chinese and Western Medicine Hospital of WenzhouWenzhouZhejiangChina
| | - Jinlai Cai
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Haijun He
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - XinShi Wang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Chenglong Xie
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Oujiang LaboratoryWenzhouZhejiangChina
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
- Department of Geriatrics, Geriatric Medical CenterThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
15
|
Bloomberg M, Muniz-Terrera G, Brocklebank L, Steptoe A. Healthy lifestyle and cognitive decline in middle-aged and older adults residing in 14 European countries. Nat Commun 2024; 15:5003. [PMID: 38937442 PMCID: PMC11211489 DOI: 10.1038/s41467-024-49262-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/30/2024] [Indexed: 06/29/2024] Open
Abstract
Studies examining lifestyle and cognitive decline often use healthy lifestyle indices, making it difficult to understand implications for interventions. We examined associations of 16 lifestyles with cognitive decline. Data from 32,033 cognitively-healthy adults aged 50-104 years participating in prospective cohort studies of aging from 14 European countries were used to examine associations of lifestyle with memory and fluency decline over 10 years. The reference lifestyle comprised not smoking, no-to-moderate alcohol consumption, weekly moderate-plus-vigorous physical activity, and weekly social contact. We found that memory and fluency decline was generally similar for non-smoking lifestyles. By contrast, memory scores declined up to 0.17 standard deviations (95% confidence interval= 0.08 - 0.27) and fluency scores up to 0.16 standard deviations (0.07 - 0.25) more over 10 years for those reporting smoking lifestyles compared with the reference lifestyle. We thus show that differences in cognitive decline between lifestyles were primarily dependent on smoking status.
Collapse
Affiliation(s)
- Mikaela Bloomberg
- Department of Behavioural Science and Health, University College London, London, UK.
| | | | - Laura Brocklebank
- Department of Behavioural Science and Health, University College London, London, UK
| | - Andrew Steptoe
- Department of Behavioural Science and Health, University College London, London, UK
| |
Collapse
|
16
|
Chen A, Shea D, Daggett V. Performance of SOBA-AD blood test in discriminating Alzheimer's disease patients from cognitively unimpaired controls in two independent cohorts. Sci Rep 2024; 14:7946. [PMID: 38575622 PMCID: PMC10995183 DOI: 10.1038/s41598-024-57107-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
Amyloid-beta (Aβ) toxic oligomers are critical early players in the molecular pathology of Alzheimer's disease (AD). We have developed a Soluble Oligomer Binding Assay (SOBA-AD) for detection of these Aβ oligomers that contain α-sheet secondary structure that discriminates plasma samples from patients on the AD continuum from non-AD controls. We tested 265 plasma samples from two independent cohorts to investigate the performance of SOBA-AD. Testing was performed at two different sites, with different personnel, reagents, and instrumentation. Across two cohorts, SOBA-AD discriminated AD patients from cognitively unimpaired (CU) subjects with 100% sensitivity, > 95% specificity, and > 98% area under the curve (AUC) (95% CI 0.95-1.00). A SOBA-AD positive readout, reflecting α-sheet toxic oligomer burden, was found in AD patients, and not in controls, providing separation of the two populations, aside from 5 SOBA-AD positive controls. Based on an earlier SOBA-AD study, the Aβ oligomers detected in these CU subjects may represent preclinical cases of AD. The results presented here support the value of SOBA-AD as a promising blood-based tool for the detection and confirmation of AD.
Collapse
Affiliation(s)
- Amy Chen
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA
| | - Dylan Shea
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA
- University of Washington, Box 355610, Seattle, WA, 98195-5610, USA
| | - Valerie Daggett
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA.
- University of Washington, Box 355610, Seattle, WA, 98195-5610, USA.
| |
Collapse
|
17
|
Klein EG, Schroeder K, Wessels AM, Phipps A, Japha M, Schilling T, Zimmer JA. How donanemab data address the coverage with evidence development questions. Alzheimers Dement 2024; 20:3127-3140. [PMID: 38323738 PMCID: PMC11032520 DOI: 10.1002/alz.13700] [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: 10/12/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 02/08/2024]
Abstract
The Centers for Medicare & Medicaid Services (CMS) established a class-based National Coverage Determination (NCD) for monoclonal antibodies directed against amyloid for Alzheimer's disease (AD) with patient access through Coverage with Evidence Development (CED) based on three questions. This review, focused on donanemab, answers each of these CED questions with quality evidence. TRAILBLAZER-ALZ registration trials are presented with supporting literature and real-world data to answer CED questions for donanemab. TRAILBLAZER-ALZ registration trials demonstrated that donanemab significantly slowed cognitive and functional decline in amyloid-positive early symptomatic AD participants, and lowered their risk of disease progression while key safety risks occurred primarily within the first 6 months and then declined. Donanemab meaningfully improved health outcomes with a manageable safety profile in an early symptomatic AD population, representative of Medicare populations across diverse practice settings. The donanemab data provide the necessary level of evidence for CMS to open a reconsideration of their NCD. HIGHLIGHTS: Donanemab meaningfully improved outcomes in trial participants with early symptomatic Alzheimer's disease. Comorbidities in trial participants were consistent with the Medicare population. Co-medications in trial participants were consistent with the Medicare population. Risks associated with treatment tended to occur in the first 6 months. Risks of amyloid-related imaging abnormalities were managed with careful observation and magnetic resonance imaging monitoring.
Collapse
Affiliation(s)
- Eric G. Klein
- Global Medical Affairs, Eli Lilly and CompanyLilly Corporate CenterIndianapolisIndianaUSA
| | - Krista Schroeder
- Research and Development, Eli Lilly and CompanyLilly Corporate CenterIndianapolisIndianaUSA
| | - Alette M. Wessels
- Research and Development, Eli Lilly and CompanyLilly Corporate CenterIndianapolisIndianaUSA
| | - Adam Phipps
- Lilly Value and Access, Eli Lilly and CompanyLilly Corporate CenterIndianapolisIndianaUSA
| | - Maureen Japha
- Corporate Affairs, Eli Lilly and CompanyLilly Corporate CenterIndianapolisIndianaUSA
| | - Traci Schilling
- Global Medical Affairs, Eli Lilly and CompanyLilly Corporate CenterIndianapolisIndianaUSA
| | - Jennifer A. Zimmer
- Research and Development, Eli Lilly and CompanyLilly Corporate CenterIndianapolisIndianaUSA
| |
Collapse
|
18
|
Lista S, Mapstone M, Caraci F, Emanuele E, López-Ortiz S, Martín-Hernández J, Triaca V, Imbimbo C, Gabelle A, Mielke MM, Nisticò R, Santos-Lozano A, Imbimbo BP. A critical appraisal of blood-based biomarkers for Alzheimer's disease. Ageing Res Rev 2024; 96:102290. [PMID: 38580173 DOI: 10.1016/j.arr.2024.102290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/18/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
Biomarkers that predict the clinical onset of Alzheimer's disease (AD) enable the identification of individuals in the early, preclinical stages of the disease. Detecting AD at this point may allow for more effective therapeutic interventions and optimized enrollment for clinical trials of novel drugs. The current biological diagnosis of AD is based on the AT(N) classification system with the measurement of brain deposition of amyloid-β (Aβ) ("A"), tau pathology ("T"), and neurodegeneration ("N"). Diagnostic cut-offs for Aβ1-42, the Aβ1-42/Aβ1-40 ratio, tau and hyperphosphorylated-tau concentrations in cerebrospinal fluid have been defined and may support AD clinical diagnosis. Blood-based biomarkers of the AT(N) categories have been described in the AD continuum. Cross-sectional and longitudinal studies have shown that the combination of blood biomarkers tracking neuroaxonal injury (neurofilament light chain) and neuroinflammatory pathways (glial fibrillary acidic protein) enhance sensitivity and specificity of AD clinical diagnosis and improve the prediction of AD onset. However, no international accepted cut-offs have been identified for these blood biomarkers. A kit for blood Aβ1-42/Aβ1-40 is commercially available in the U.S.; however, it does not provide a diagnosis, but simply estimates the risk of developing AD. Although blood-based AD biomarkers have a great potential in the diagnostic work-up of AD, they are not ready for the routine clinical use.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Mark Mapstone
- Department of Neurology, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA.
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina 94018, Italy.
| | | | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome 00015, Italy.
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy.
| | - Audrey Gabelle
- Memory Resources and Research Center, Montpellier University of Excellence i-site, Montpellier 34295, France.
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA.
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome 00133, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome 00143, Italy.
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain; Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid 28041, Spain.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma 43122, Italy.
| |
Collapse
|
19
|
De Strooper B, Karran E. New precision medicine avenues to the prevention of Alzheimer's disease from insights into the structure and function of γ-secretases. EMBO J 2024; 43:887-903. [PMID: 38396302 PMCID: PMC10943082 DOI: 10.1038/s44318-024-00057-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/20/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Two phase-III clinical trials with anti-amyloid peptide antibodies have met their primary goal, i.e. slowing of Alzheimer's disease (AD) progression. However, antibody therapy may not be the optimal therapeutic modality for AD prevention, as we will discuss in the context of the earlier small molecules described as "γ-secretase modulators" (GSM). We review here the structure, function, and pathobiology of γ-secretases, with a focus on how mutations in presenilin genes result in early-onset AD. Significant progress has been made in generating compounds that act in a manner opposite to pathogenic presenilin mutations: they stabilize the proteinase-substrate complex, thereby increasing the processivity of substrate cleavage and altering the size spectrum of Aβ peptides produced. We propose the term "γ-secretase allosteric stabilizers" (GSAS) to distinguish these compounds from the rather heterogenous class of GSM. The GSAS represent, in theory, a precision medicine approach to the prevention of amyloid deposition, as they specifically target a discrete aspect in a complex cell biological signalling mechanism that initiates the pathological processes leading to Alzheimer's disease.
Collapse
Affiliation(s)
- Bart De Strooper
- Dementia Research Institute, Institute of Neurology, University College London, at the Francis Crick Institute, London, NW1 AT, UK.
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain & Disease Research, and Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium.
| | - Eric Karran
- Cambridge Research Center, AbbVie, Inc., Cambridge, MA, USA
| |
Collapse
|
20
|
Abiose O, Rutledge J, Moran‐Losada P, Belloy ME, Wilson EN, He Z, Trelle AN, Channappa D, Romero A, Park J, Yutsis MV, Sha SJ, Andreasson KI, Poston KL, Henderson VW, Wagner AD, Wyss‐Coray T, Mormino EC. Post-translational modifications linked to preclinical Alzheimer's disease-related pathological and cognitive changes. Alzheimers Dement 2024; 20:1851-1867. [PMID: 38146099 PMCID: PMC10984434 DOI: 10.1002/alz.13576] [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: 08/01/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION In this study, we leverage proteomic techniques to identify communities of proteins underlying Alzheimer's disease (AD) risk among clinically unimpaired (CU) older adults. METHODS We constructed a protein co-expression network using 3869 cerebrospinal fluid (CSF) proteins quantified by SomaLogic, Inc., in a cohort of participants along the AD clinical spectrum. We then replicated this network in an independent cohort of CU older adults and related these modules to clinically-relevant outcomes. RESULTS We discovered modules enriched for phosphorylation and ubiquitination that were associated with abnormal amyloid status, as well as p-tau181 (M4: β = 2.44, p < 0.001, M7: β = 2.57, p < 0.001) and executive function performance (M4: β = -2.00, p = 0.005, M7: β = -2.39, p < 0.001). DISCUSSION In leveraging CSF proteomic data from individuals spanning the clinical spectrum of AD, we highlight the importance of post-translational modifications for early cognitive and pathological changes.
Collapse
Affiliation(s)
- Olamide Abiose
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
| | - Jarod Rutledge
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
- Department of GeneticsStanford UniversityStanfordCaliforniaUSA
| | - Patricia Moran‐Losada
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Michael E. Belloy
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Edward N. Wilson
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
| | - Zihuai He
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Center for Biomedical Informatics ResearchStanford University School of MedicineStanfordCaliforniaUSA
| | - Alexandra N. Trelle
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Divya Channappa
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - America Romero
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Jennifer Park
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Maya V. Yutsis
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Sharon J. Sha
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Katrin I. Andreasson
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- Chan Zuckerberg BiohubSan FranciscoCaliforniaUSA
| | - Kathleen L. Poston
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Victor W. Henderson
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Department of Epidemiology & Population HealthStanford University School of MedicineStanfordCaliforniaUSA
| | - Anthony D. Wagner
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- Department of PsychologyStanford UniversityStanfordCaliforniaUSA
| | - Tony Wyss‐Coray
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Elizabeth C. Mormino
- Department of Neurology and Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
21
|
Evans SA, Paitel ER, Bhasin R, Nielson KA. Genetic Risk for Alzheimer's Disease Alters Perceived Executive Dysfunction in Cognitively Healthy Middle-Aged and Older Adults. J Alzheimers Dis Rep 2024; 8:267-279. [PMID: 38405345 PMCID: PMC10894609 DOI: 10.3233/adr-230166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024] Open
Abstract
Background Subjective cognitive complaints (SCC) may be an early indicator of future cognitive decline. However, findings comparing SCC and objective cognitive performance have varied, particularly in the memory domain. Even less well established is the relationship between subjective and objective complaints in non-amnestic domains, such as in executive functioning, despite evidence indicating very early changes in these domains. Moreover, particularly early changes in both amnestic and non-amnestic domains are apparent in those carrying the Apolipoprotein-E ɛ4 allele, a primary genetic risk for Alzheimer's disease (AD). Objective This study investigated the role of the ɛ4 allele in the consistency between subjective and objective executive functioning in 54 healthy, cognitively intact, middle-aged and older adults. Methods Participants (Mage = 64.07, SD = 9.27, range = 48-84; ɛ4+ = 18) completed the Frontal Systems Behavior Scale (FrSBe) Executive Dysfunction Scale (EXECDYS) to measure subjective executive functioning (SEF) and multiple executive functioning tasks, which were condensed into a single factor. Results After accounting for age, depression, and anxiety, objective executive functioning performance significantly predicted SEF. Importantly, ɛ4 moderated this effect. Specifically, those carrying the ɛ4 allele had significantly less accurate self-awareness of their executive functioning compared to ɛ4 non-carriers. Conclusions Utilizing an approach that integrates self-evaluation of executive functioning with objective neurocognitive assessment may help identify the earliest signs of impending cognitive decline, particularly in those with genetic risk for AD. Such an approach could sensitively determine those most prone to future cognitive decline prior to symptom onset, when interventions could be most effective.
Collapse
Affiliation(s)
- Sarah A. Evans
- Department of Psychology, Marquette University, Milwaukee, WI, USA
| | | | - Riya Bhasin
- Department of Psychology, Marquette University, Milwaukee, WI, USA
| | | |
Collapse
|
22
|
Ribba B, Simuni T, Marek K, Siderowf A, Diack C, Pierrillas PB, Monnet A, Ricci B, Nikolcheva T, Pagano G. Modeling of Parkinson's Disease Progression and Implications for Detection of Disease Modification in Treatment Trials. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1225-1235. [PMID: 39058452 PMCID: PMC11380229 DOI: 10.3233/jpd-230446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Background Objectively measuring Parkinson's disease (PD) signs and symptoms over time is critical for the successful development of treatments aimed at halting the disease progression of people with PD. Objective To create a clinical trial simulation tool that characterizes the natural history of PD progression and enables a data-driven design of randomized controlled studies testing potential disease-modifying treatments (DMT) in early-stage PD. Methods Data from the Parkinson's Progression Markers Initiative (PPMI) were analyzed with nonlinear mixed-effect modeling techniques to characterize the progression of MDS-UPDRS part I (non-motor aspects of experiences of daily living), part II (motor aspects of experiences of daily living), and part III (motor signs). A clinical trial simulation tool was built from these disease models and used to predict probability of success as a function of trial design. Results MDS-UPDRS part III progresses approximately 3 times faster than MDS-UPDRS part II and I, with an increase of 3 versus 1 points/year. Higher amounts of symptomatic therapy is associated with slower progression of MDS-UPDRS part II and III. The modeling framework predicts that a DMT effect on MDS-UPDRS part III could precede effect on part II by approximately 2 to 3 years. Conclusions Our clinical trial simulation tool predicted that in a two-year randomized controlled trial, MDS-UPDRS part III could be used to evaluate a potential novel DMT, while part II would require longer trials of a minimum duration of 3 to 5 years underscoring the need for innovative trial design approaches including novel patient-centric measures.
Collapse
Affiliation(s)
- Benjamin Ribba
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cheikh Diack
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Philippe Bernard Pierrillas
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Annabelle Monnet
- Roche Product Development, F. Hoffmann La Roche Ltd., Basel, Switzerland
| | - Benedicte Ricci
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Tania Nikolcheva
- Roche Product Development, F. Hoffmann La Roche Ltd., Basel, Switzerland
| | - Gennaro Pagano
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
- University of Exeter Medical School, London, UK
| |
Collapse
|
23
|
Evenden D, Prosser A, Michopoulou S, Kipps C. ADCOMS sensitivity versus baseline diagnosis and progression phenotypes. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12540. [PMID: 38406608 PMCID: PMC10885177 DOI: 10.1002/dad2.12540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 02/27/2024]
Abstract
BACKGROUND The Alzheimer's Disease COMposite Score (ADCOMS) is more sensitive in clinical trials than conventional measures when assessing pre-dementia. This study compares ADCOMS trajectories using clustered progression characteristics to better understand different patterns of decline. METHODS Post-baseline ADCOMS values were analyzed for sensitivity using mean-to-standard deviation ratio (MSDR), partitioned by baseline diagnosis, comparing with the original scales upon which ADCOMS is based. Because baseline diagnosis was not a particularly reliable predictor of progression, individuals were also grouped into similar ADCOMS progression trajectories using clustering methods and the MSDR compared for each progression group. RESULTS ADCOMS demonstrated increased sensitivity for clinically important progression groups. ADCOMS did not show statistically significant sensitivity or clinical relevance for the less-severe baseline diagnoses and marginal progression groups. CONCLUSIONS This analysis complements and extends previous work validating the sensitivity of ADCOMS. The large data set permitted evaluation-in a novel approach-by the clustered progression group.
Collapse
Affiliation(s)
- Dave Evenden
- Clinical and Experimental SciencesUniversity of SouthamptonSouthamptonUK
| | - Angus Prosser
- Clinical and Experimental SciencesUniversity of SouthamptonSouthamptonUK
| | - Sofia Michopoulou
- Clinical and Experimental SciencesUniversity of SouthamptonSouthamptonUK
- Imaging PhysicsUniversity Hospital SouthamptonSouthamptonUK
| | - Christopher Kipps
- Clinical and Experimental SciencesUniversity of SouthamptonSouthamptonUK
- Wessex Neurological CentreUniversity Hospital SouthamptonSouthamptonUK
| | | |
Collapse
|
24
|
Maleki SF, Yousefi M, Sobhi N, Jafarizadeh A, Alizadehsani R, Gorriz-Saez JM. Artificial Intelligence in Eye Movements Analysis for Alzheimer's Disease Early Diagnosis. Curr Alzheimer Res 2024; 21:155-165. [PMID: 38840390 DOI: 10.2174/0115672050322607240529075641] [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: 04/18/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
As the world's population ages, Alzheimer's disease is currently the seventh most common cause of death globally; the burden is anticipated to increase, especially among middle-class and elderly persons. Artificial intelligence-based algorithms that work well in hospital environments can be used to identify Alzheimer's disease. A number of databases were searched for English- language articles published up until March 1, 2024, that examined the relationships between artificial intelligence techniques, eye movements, and Alzheimer's disease. A novel non-invasive method called eye movement analysis may be able to reflect cognitive processes and identify anomalies in Alzheimer's disease. Artificial intelligence, particularly deep learning, and machine learning, is required to enhance Alzheimer's disease detection using eye movement data. One sort of deep learning technique that shows promise is convolutional neural networks, which need further data for precise classification. Nonetheless, machine learning models showed a high degree of accuracy in this context. Artificial intelligence-driven eye movement analysis holds promise for enhancing clinical evaluations, enabling tailored treatment, and fostering the development of early and precise Alzheimer's disease diagnosis. A combination of artificial intelligence-based systems and eye movement analysis can provide a window for early and non-invasive diagnosis of Alzheimer's disease. Despite ongoing difficulties with early Alzheimer's disease detection, this presents a novel strategy that may have consequences for clinical evaluations and customized medication to improve early and accurate diagnosis.
Collapse
Affiliation(s)
| | - Milad Yousefi
- Faculty of Mathematics, Statistics, and Computer Sciences, University of Tabriz, Tabriz, Iran
| | - Navid Sobhi
- Nikookari Eye Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Jafarizadeh
- Nikookari Eye Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovation, Deakin University, VIC3216, Australia
| | - Juan Manuel Gorriz-Saez
- Data Science and Computational Intelligence Institute, Universidad de Granada, Granada, Spain
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
Sun MK, Alkon DL. Treating Alzheimer's Disease: Focusing on Neurodegenerative Consequences. J Alzheimers Dis 2024; 101:S263-S274. [PMID: 39422958 DOI: 10.3233/jad-240479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Neurodegenerative disorders involve progressive dysfunction and loss of synapses and neurons and brain atrophy, slowly declining memories and cognitive skills, throughout a long process. Alzheimer's disease (AD), the leading neurodegenerative disorder, suffers from a lack of effective therapeutic drugs. Decades of efforts targeting its pathologic hallmarks, amyloid plaques and neurofibrillary tangles, in clinical trials have produced therapeutics with marginal benefits that lack meaningful clinical improvements in cognition. Delivering meaningful clinical therapeutics to treat or prevent neurodegenerative disorders thus remains a great challenge to scientists and clinicians. Emerging evidence, however, suggests that dysfunction of various synaptogenic signaling pathways participates in the neurodegenerative progression, resulting in deterioration of operation/structure of the synaptic networks involved in cognition. These derailed endogenous signaling pathways and disease processes are potential pharmacological targets for the therapies. Therapeutics with meaningful clinical benefit in cognition may depend on the effectiveness of arresting and reversing the neurodegenerative process through these targets. In essence, promoting neuro-regeneration may represent the only option to recover degenerated synapses and neurons. These potential directions in clinical trials for AD therapeutics with meaningful clinical benefit in cognitive function are summarized and discussed.
Collapse
|
26
|
Gu D, Lv X, Shi C, Zhang T, Liu S, Fan Z, Tu L, Zhang M, Zhang N, Chen L, Wang Z, Wang J, Zhang Y, Li H, Wang L, Zhu J, Zheng Y, Wang H, Yu X. A Stable and Scalable Digital Composite Neurocognitive Test for Early Dementia Screening Based on Machine Learning: Model Development and Validation Study. J Med Internet Res 2023; 25:e49147. [PMID: 38039074 PMCID: PMC10724812 DOI: 10.2196/49147] [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: 05/20/2023] [Revised: 09/30/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Dementia has become a major public health concern due to its heavy disease burden. Mild cognitive impairment (MCI) is a transitional stage between healthy aging and dementia. Early identification of MCI is an essential step in dementia prevention. OBJECTIVE Based on machine learning (ML) methods, this study aimed to develop and validate a stable and scalable panel of cognitive tests for the early detection of MCI and dementia based on the Chinese Neuropsychological Consensus Battery (CNCB) in the Chinese Neuropsychological Normative Project (CN-NORM) cohort. METHODS CN-NORM was a nationwide, multicenter study conducted in China with 871 participants, including an MCI group (n=327, 37.5%), a dementia group (n=186, 21.4%), and a cognitively normal (CN) group (n=358, 41.1%). We used the following 4 algorithms to select candidate variables: the F-score according to the SelectKBest method, the area under the curve (AUC) from logistic regression (LR), P values from the logit method, and backward stepwise elimination. Different models were constructed after considering the administration duration and complexity of combinations of various tests. Receiver operating characteristic curve and AUC metrics were used to evaluate the discriminative ability of the models via stratified sampling cross-validation and LR and support vector classification (SVC) algorithms. This model was further validated in the Alzheimer's Disease Neuroimaging Initiative phase 3 (ADNI-3) cohort (N=743), which included 416 (56%) CN subjects, 237 (31.9%) patients with MCI, and 90 (12.1%) patients with dementia. RESULTS Except for social cognition, all other domains in the CNCB differed between the MCI and CN groups (P<.008). In feature selection results regarding discrimination between the MCI and CN groups, the Hopkins Verbal Learning Test-5 minutes Recall had the best performance, with the highest mean AUC of up to 0.80 (SD 0.02) and an F-score of up to 258.70. The scalability of model 5 (Hopkins Verbal Learning Test-5 minutes Recall and Trail Making Test-B) was the lowest. Model 5 achieved a higher level of discrimination than the Hong Kong Brief Cognitive test score in distinguishing between the MCI and CN groups (P<.05). Model 5 also provided the highest sensitivity of up to 0.82 (range 0.72-0.92) and 0.83 (range 0.75-0.91) according to LR and SVC, respectively. This model yielded a similar robust discriminative performance in the ADNI-3 cohort regarding differentiation between the MCI and CN groups, with a mean AUC of up to 0.81 (SD 0) according to both LR and SVC algorithms. CONCLUSIONS We developed a stable and scalable composite neurocognitive test based on ML that could differentiate not only between patients with MCI and controls but also between patients with different stages of cognitive impairment. This composite neurocognitive test is a feasible and practical digital biomarker that can potentially be used in large-scale cognitive screening and intervention studies.
Collapse
Affiliation(s)
- Dongmei Gu
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Xiaozhen Lv
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Chuan Shi
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Tianhong Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sha Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zili Fan
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Lihui Tu
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Ming Zhang
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
- Department of Psychiatry, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Liming Chen
- China Telecom Digital Intelligence Technology Co.,Ltd, Beijing, China
| | - Zhijiang Wang
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Jing Wang
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Ying Zhang
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Huizi Li
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Luchun Wang
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Jiahui Zhu
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Yaonan Zheng
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Huali Wang
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| | - Xin Yu
- Clinical Research Division, Dementia Care and Research Center, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders (Peking University), National Health Committee Key Laboratory of Mental Health, Beijing, China
| |
Collapse
|
27
|
Welsh‐Bohmer KA, Kerchner GA, Dhadda S, Garcia M, Miller DS, Natanegara F, Raket LL, Robieson W, Siemers ER, Carrillo MC, Weber CJ. Decision making in clinical trials: Interim analyses, innovative design, and biomarkers. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12421. [PMID: 37867532 PMCID: PMC10585126 DOI: 10.1002/trc2.12421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/14/2023] [Indexed: 10/24/2023]
Abstract
The efficient and accurate execution of clinical trials testing novel treatments for Alzheimer's disease (AD) is a critical component of the field's collective efforts to develop effective disease-modifying treatments for AD. The lengthy and heterogeneous nature of clinical progression in AD contributes to the challenges inherent in demonstrating a clinically meaningful benefit of any potential new AD therapy. The failure of many large and expensive clinical trials to date has prompted a focus on optimizing all aspects of decision making, to not only expedite the development of new treatments, but also maximize the value of the information that each clinical trial yields, so that all future clinical trials (including those that are negative) will contribute toward advancing the field. To address this important topic the Alzheimer's Association Research Roundtable convened December 1-2, 2020. The goals focused around identifying new directions and actionable steps to enhance clinical trial decision making in planned future studies.
Collapse
Affiliation(s)
| | | | | | - Miguel Garcia
- Boehringer Ingelheim Pharmaceuticals Inc.RidgefieldConnecticutUSA
| | | | - Fanni Natanegara
- Eli Lilly and Company Lilly Corporate CenterIndianapolisIndianaUSA
| | | | | | | | | | | |
Collapse
|
28
|
Sims JR, Zimmer JA, Evans CD, Lu M, Ardayfio P, Sparks J, Wessels AM, Shcherbinin S, Wang H, Monkul Nery ES, Collins EC, Solomon P, Salloway S, Apostolova LG, Hansson O, Ritchie C, Brooks DA, Mintun M, Skovronsky DM. Donanemab in Early Symptomatic Alzheimer Disease: The TRAILBLAZER-ALZ 2 Randomized Clinical Trial. JAMA 2023; 330:512-527. [PMID: 37459141 PMCID: PMC10352931 DOI: 10.1001/jama.2023.13239] [Citation(s) in RCA: 983] [Impact Index Per Article: 491.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/28/2023] [Indexed: 07/20/2023]
Abstract
Importance There are limited efficacious treatments for Alzheimer disease. Objective To assess efficacy and adverse events of donanemab, an antibody designed to clear brain amyloid plaque. Design, Setting, and Participants Multicenter (277 medical research centers/hospitals in 8 countries), randomized, double-blind, placebo-controlled, 18-month phase 3 trial that enrolled 1736 participants with early symptomatic Alzheimer disease (mild cognitive impairment/mild dementia) with amyloid and low/medium or high tau pathology based on positron emission tomography imaging from June 2020 to November 2021 (last patient visit for primary outcome in April 2023). Interventions Participants were randomized in a 1:1 ratio to receive donanemab (n = 860) or placebo (n = 876) intravenously every 4 weeks for 72 weeks. Participants in the donanemab group were switched to receive placebo in a blinded manner if dose completion criteria were met. Main Outcomes and Measures The primary outcome was change in integrated Alzheimer Disease Rating Scale (iADRS) score from baseline to 76 weeks (range, 0-144; lower scores indicate greater impairment). There were 24 gated outcomes (primary, secondary, and exploratory), including the secondary outcome of change in the sum of boxes of the Clinical Dementia Rating Scale (CDR-SB) score (range, 0-18; higher scores indicate greater impairment). Statistical testing allocated α of .04 to testing low/medium tau population outcomes, with the remainder (.01) for combined population outcomes. Results Among 1736 randomized participants (mean age, 73.0 years; 996 [57.4%] women; 1182 [68.1%] with low/medium tau pathology and 552 [31.8%] with high tau pathology), 1320 (76%) completed the trial. Of the 24 gated outcomes, 23 were statistically significant. The least-squares mean (LSM) change in iADRS score at 76 weeks was -6.02 (95% CI, -7.01 to -5.03) in the donanemab group and -9.27 (95% CI, -10.23 to -8.31) in the placebo group (difference, 3.25 [95% CI, 1.88-4.62]; P < .001) in the low/medium tau population and -10.2 (95% CI, -11.22 to -9.16) with donanemab and -13.1 (95% CI, -14.10 to -12.13) with placebo (difference, 2.92 [95% CI, 1.51-4.33]; P < .001) in the combined population. LSM change in CDR-SB score at 76 weeks was 1.20 (95% CI, 1.00-1.41) with donanemab and 1.88 (95% CI, 1.68-2.08) with placebo (difference, -0.67 [95% CI, -0.95 to -0.40]; P < .001) in the low/medium tau population and 1.72 (95% CI, 1.53-1.91) with donanemab and 2.42 (95% CI, 2.24-2.60) with placebo (difference, -0.7 [95% CI, -0.95 to -0.45]; P < .001) in the combined population. Amyloid-related imaging abnormalities of edema or effusion occurred in 205 participants (24.0%; 52 symptomatic) in the donanemab group and 18 (2.1%; 0 symptomatic during study) in the placebo group and infusion-related reactions occurred in 74 participants (8.7%) with donanemab and 4 (0.5%) with placebo. Three deaths in the donanemab group and 1 in the placebo group were considered treatment related. Conclusions and Relevance Among participants with early symptomatic Alzheimer disease and amyloid and tau pathology, donanemab significantly slowed clinical progression at 76 weeks in those with low/medium tau and in the combined low/medium and high tau pathology population. Trial Registration ClinicalTrials.gov Identifier: NCT04437511.
Collapse
Affiliation(s)
| | | | | | - Ming Lu
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | - Hong Wang
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | - Paul Solomon
- Boston Center for Memory and Boston University Alzheimer’s Disease Center, Boston, Massachusetts
| | - Stephen Salloway
- Department of Neurology and Department of Psychiatry, Alpert Medical School of Brown University, Providence, Rhode Island
- Butler Hospital, Providence, Rhode Island
| | - Liana G. Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Lund, Sweden
| | | | | | - Mark Mintun
- Eli Lilly and Company, Indianapolis, Indiana
| | | |
Collapse
|
29
|
Quadalti C, Palmqvist S, Hall S, Rossi M, Mammana A, Janelidze S, Dellavalle S, Mattsson-Carlgren N, Baiardi S, Stomrud E, Hansson O, Parchi P. Clinical effects of Lewy body pathology in cognitively impaired individuals. Nat Med 2023; 29:1964-1970. [PMID: 37464058 PMCID: PMC10427416 DOI: 10.1038/s41591-023-02449-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
There is poor knowledge about the clinical effects of Lewy body (LB) pathology in patients with cognitive impairment, especially when coexisting with Alzheimer's disease (AD) pathology (amyloid-β and tau). Using a seed amplification assay, we analyzed cerebrospinal fluid for misfolded LB-associated α-synuclein in 883 memory clinic patients with mild cognitive impairment or dementia from the BioFINDER study. Twenty-three percent had LB pathology, of which only 21% fulfilled clinical criteria of Parkinson's disease or dementia with Lewy bodies at baseline. Among these LB-positive patients, 48% had AD pathology. Fifty-four percent had AD pathology in the whole sample (17% of mild cognitive impairment and 24% of patients with dementia were also LB-positive). When examining independent cross-sectional effects, LB pathology but not amyloid-β or tau, was associated with hallucinations and worse attention/executive, visuospatial and motor function. LB pathology was also associated with faster longitudinal decline in all examined cognitive functions, independent of amyloid-β, tau, cognitive stage and a baseline diagnosis of dementia with Lewy bodies/Parkinson's disease. LB status provides a better precision-medicine approach to predict clinical trajectories independent of AD biomarkers and a clinical diagnosis, which could have implications for the clinical management of cognitive impairment and the design of AD and LB drug trials.
Collapse
Affiliation(s)
- Corinne Quadalti
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sara Hall
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Marcello Rossi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Angela Mammana
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Sofia Dellavalle
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Neurology Clinic, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Simone Baiardi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Erik Stomrud
- 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.
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
30
|
Palmqvist S, Rossi M, Hall S, Quadalti C, Mattsson-Carlgren N, Dellavalle S, Tideman P, Pereira JB, Nilsson MH, Mammana A, Janelidze S, Baiardi S, Stomrud E, Parchi P, Hansson O. Cognitive effects of Lewy body pathology in clinically unimpaired individuals. Nat Med 2023; 29:1971-1978. [PMID: 37464059 PMCID: PMC10427420 DOI: 10.1038/s41591-023-02450-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
α-Synuclein aggregates constitute the pathology of Lewy body (LB) disease. Little is known about the effects of LB pathology in preclinical (presymptomatic) individuals, either as isolated pathology or coexisting with Alzheimer's disease (AD) pathology (β-amyloid (Aβ) and tau). We examined the effects of LB pathology using a cerebrospinal fluid α-synuclein-seed amplification assay in 1,182 cognitively and neurologically unimpaired participants from the BioFINDER study: 8% were LB positive, 26% Aβ positive (13% of those were LB positive) and 16% tau positive. LB positivity occurred more often in the presence of Aβ positivity but not tau positivity. LB pathology had independently negative effects on cross-sectional and longitudinal global cognition and memory and on longitudinal attention/executive function. Tau had cognitive effects of a similar magnitude, but these were less pronounced for Aβ. Participants with both LB and AD (Aβ and tau) pathology exhibited faster cognitive decline than those with only LB or AD pathology. LB, but not AD, pathology was associated with reduced sense of smell. Only LB-positive participants progressed to clinical LB disease over 10 years. These results are important for individualized prognosis, recruitment and choice of outcome measures in preclinical LB disease trials, but also for the design of early AD trials because >10% of individuals with preclinical AD have coexisting LB pathology.
Collapse
Affiliation(s)
- Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Marcello Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Sara Hall
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Corinne Quadalti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sofia Dellavalle
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Maria H Nilsson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Department of Health Sciences, Lund University, Lund, Sweden
| | - Angela Mammana
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Simone Baiardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
31
|
Insel PS, Kumar A, Hansson O, Mattsson-Carlgren N. Genetic Moderation of the Association of β-Amyloid With Cognition and MRI Brain Structure in Alzheimer Disease. Neurology 2023; 101:e20-e29. [PMID: 37085326 PMCID: PMC10351305 DOI: 10.1212/wnl.0000000000207305] [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: 07/27/2022] [Accepted: 03/03/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES There is considerable heterogeneity in the association between increasing β-amyloid (Aβ) pathology and early cognitive dysfunction in preclinical Alzheimer disease (AD). At this stage, some individuals show no signs of cognitive dysfunction, while others show clear signs of decline. The factors explaining this heterogeneity are particularly important for understanding progression in AD but remain largely unknown. In this study, we examined an array of genetic variants that may influence the relationships among Aβ, brain structure, and cognitive performance in 2 large cohorts. METHODS In 2,953 cognitively unimpaired participants from the Anti-Amyloid Treatment in Asymptomatic Alzheimer disease (A4) study, interactions between genetic variants and 18F-Florbetapir PET standardized uptake value ratio (SUVR) to predict the Preclinical Alzheimer Cognitive Composite (PACC) were assessed. Genetic variants identified in the A4 study were evaluated in the Alzheimer Disease Neuroimaging Initiative (ADNI, N = 527) for their association with longitudinal cognition and brain atrophy in both cognitively unimpaired participants and those with mild cognitive impairment. RESULTS In the A4 study, 4 genetic variants significantly moderated the association between Aβ load and cognition. Minor alleles of 3 variants were associated with additional decreases in PACC scores with increasing Aβ SUVR (rs78021285, β = -2.29, SE = 0.40, p FDR = 0.02, nearest gene ARPP21; rs71567499, β = -2.16, SE = 0.38, p FDR = 0.02, nearest gene PPARD; and rs10974405, β = -1.68, SE = 0.29, p FDR = 0.02, nearest gene GLIS3). The minor allele of rs7825645 was associated with less decrease in PACC scores with increasing Aβ SUVR (β = 0.71, SE = 0.13, p FDR = 0.04, nearest gene FGF20). The genetic variant rs76366637, in linkage disequilibrium with rs78021285, was available in both the A4 and ADNI. In the A4, rs76366637 was strongly associated with reduced PACC scores with increasing Aβ SUVR (β = -1.01, SE = 0.21, t = -4.90, p < 0.001). In the ADNI, rs76366637 was associated with accelerated cognitive decline (χ2 = 15.3, p = 0.004) and atrophy over time (χ2 = 26.8, p < 0.001), with increasing Aβ SUVR. DISCUSSION Patterns of increased cognitive dysfunction and accelerated atrophy due to specific genetic variation may explain some of the heterogeneity in cognition in preclinical and prodromal AD. The genetic variant near ARPP21 associated with lower cognitive scores in the A4 and accelerated cognitive decline and brain atrophy in the ADNI may help to identify those at the highest risk of accelerated progression of AD.
Collapse
Affiliation(s)
- Philip S Insel
- From the Clinical Memory Research Unit (P.S.I., A.K., O.H., N.M.-C.), Faculty of Medicine, Lund University, Sweden; Department of Psychiatry and Behavioral Sciences (P.S.I.), University of California, San Francisco; Memory Clinic (O.H.), Department of Neurology (N.M.-C.), Skåne University Hospital, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden.
| | - Atul Kumar
- From the Clinical Memory Research Unit (P.S.I., A.K., O.H., N.M.-C.), Faculty of Medicine, Lund University, Sweden; Department of Psychiatry and Behavioral Sciences (P.S.I.), University of California, San Francisco; Memory Clinic (O.H.), Department of Neurology (N.M.-C.), Skåne University Hospital, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit (P.S.I., A.K., O.H., N.M.-C.), Faculty of Medicine, Lund University, Sweden; Department of Psychiatry and Behavioral Sciences (P.S.I.), University of California, San Francisco; Memory Clinic (O.H.), Department of Neurology (N.M.-C.), Skåne University Hospital, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| | - Niklas Mattsson-Carlgren
- From the Clinical Memory Research Unit (P.S.I., A.K., O.H., N.M.-C.), Faculty of Medicine, Lund University, Sweden; Department of Psychiatry and Behavioral Sciences (P.S.I.), University of California, San Francisco; Memory Clinic (O.H.), Department of Neurology (N.M.-C.), Skåne University Hospital, and Wallenberg Center for Molecular Medicine (N.M.-C.), Lund University, Sweden
| |
Collapse
|
32
|
Giorgio J, Tanna A, Malpetti M, White SR, Wang J, Baker S, Landau S, Tanaka T, Chen C, Rowe JB, O'Brien J, Fripp J, Breakspear M, Jagust W, Kourtzi Z. A robust harmonization approach for cognitive data from multiple aging and dementia cohorts. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12453. [PMID: 37502020 PMCID: PMC10369372 DOI: 10.1002/dad2.12453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Although many cognitive measures have been developed to assess cognitive decline due to Alzheimer's disease (AD), there is little consensus on optimal measures, leading to varied assessments across research cohorts and clinical trials making it difficult to pool cognitive measures across studies. METHODS We used a two-stage approach to harmonize cognitive data across cohorts and derive a cross-cohort score of cognitive impairment due to AD. First, we pool and harmonize cognitive data from international cohorts of varying size and ethnic diversity. Next, we derived cognitive composites that leverage maximal data from the harmonized dataset. RESULTS We show that our cognitive composites are robust across cohorts and achieve greater or comparable sensitivity to AD-related cognitive decline compared to the Mini-Mental State Examination and Preclinical Alzheimer Cognitive Composite. Finally, we used an independent cohort validating both our harmonization approach and composite measures. DISCUSSION Our easy to implement and readily available pipeline offers an approach for researchers to harmonize their cognitive data with large publicly available cohorts, providing a simple way to pool data for the development or validation of findings related to cognitive decline due to AD.
Collapse
Affiliation(s)
- Joseph Giorgio
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
- School of Psychological SciencesCollege of Engineering, Science and the EnvironmentUniversity of NewcastleNewcastleNew South WalesAustralia
| | - Ankeet Tanna
- Department of PsychologyUniversity of CambridgeCambridgeUK
| | - Maura Malpetti
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Simon R. White
- Department of PsychiatryUniversity of CambridgeCambridgeUK
- MRC Biostatistics UnitUniversity of CambridgeshireCambridgeUK
| | - Jingshen Wang
- Division of BiostatisticsUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Suzanne Baker
- Molecular Biophysics & Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Susan Landau
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Tomotaka Tanaka
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
| | - Christopher Chen
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
| | - James B. Rowe
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - John O'Brien
- Department of PsychiatryUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Jurgen Fripp
- The Australian eHealth Research CentreCSIRO Health and BiosecurityBrisbaneQueenslandAustralia
| | - Michael Breakspear
- School of Psychological SciencesCollege of Engineering, Science and the EnvironmentUniversity of NewcastleNewcastleNew South WalesAustralia
| | - William Jagust
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
- Molecular Biophysics & Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Zoe Kourtzi
- Department of PsychologyUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
33
|
Wijaya A, Setiawan NA, Ahmad AH, Zakaria R, Othman Z. Electroencephalography and mild cognitive impairment research: A scoping review and bibliometric analysis (ScoRBA). AIMS Neurosci 2023; 10:154-171. [PMID: 37426780 PMCID: PMC10323261 DOI: 10.3934/neuroscience.2023012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/27/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Mild cognitive impairment (MCI) is often considered a precursor to Alzheimer's disease (AD) and early diagnosis may help improve treatment effectiveness. To identify accurate MCI biomarkers, researchers have utilized various neuroscience techniques, with electroencephalography (EEG) being a popular choice due to its low cost and better temporal resolution. In this scoping review, we analyzed 2310 peer-reviewed articles on EEG and MCI between 2012 and 2022 to track the research progress in this field. Our data analysis involved co-occurrence analysis using VOSviewer and a Patterns, Advances, Gaps, Evidence of Practice, and Research Recommendations (PAGER) framework. We found that event-related potentials (ERP), EEG, epilepsy, quantitative EEG (QEEG), and EEG-based machine learning were the primary research themes. The study showed that ERP/EEG, QEEG, and EEG-based machine learning frameworks provide high-accuracy detection of seizure and MCI. These findings identify the main research themes in EEG and MCI and suggest promising avenues for future research in this field.
Collapse
Affiliation(s)
- Adi Wijaya
- Department of Health Information Management, Universitas Indonesia Maju, Jakarta, Indonesia
| | - Noor Akhmad Setiawan
- Department of Electrical and Information Engineering, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Asma Hayati Ahmad
- School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kota Bharu, Malaysia
| | - Rahimah Zakaria
- School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kota Bharu, Malaysia
| | - Zahiruddin Othman
- School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kota Bharu, Malaysia
| |
Collapse
|
34
|
Petersen RC, Aisen PS, Andrews JS, Atri A, Matthews BR, Rentz DM, Siemers ER, Weber CJ, Carrillo MC. Expectations and clinical meaningfulness of randomized controlled trials. Alzheimers Dement 2023; 19:2730-2736. [PMID: 36748826 DOI: 10.1002/alz.12959] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) clinical trials are designed and powered to detect the impact of a therapeutic intervention, and there has been considerable discussion on what constitutes a clinically meaningful change in those receiving treatment versus placebo. The pathology of AD is complex, beginning many years before clinical symptoms are detectable, with multiple potential opportunities for therapeutic engagement. Introducing treatment strategies early in the disease and assessing meaningful change over the course of an 18-month clinical trial are critical to understanding the value to an effective intervention. With new clinical trial data expected soon on emerging therapeutics from several AD studies, the Alzheimer's Association convened a work group of experts to discuss key considerations for interpreting data from cognitive and functional measures and what is considered a clinically meaningful benefit or meaningful slowing of this fatal disease. Our expectations of outcomes from therapeutic interventions in AD may need to be modified.
Collapse
Affiliation(s)
| | - Paul S Aisen
- USC Alzheimer's Therapeutic Research Institute, San Diego, California, USA
| | | | - Alireza Atri
- Banner Sun Health Research Institute, Banner Health, Sun City, Arizona, USA
| | | | - Dorene M Rentz
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital and Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
35
|
Hampton OL, Mukherjee S, Properzi MJ, Schultz AP, Crane PK, Gibbons LE, Hohman TJ, Maruff P, Lim YY, Amariglio RE, Papp KV, Johnson KA, Rentz DM, Sperling RA, Buckley RF. Harmonizing the preclinical Alzheimer cognitive composite for multicohort studies. Neuropsychology 2023; 37:436-449. [PMID: 35862098 PMCID: PMC9859944 DOI: 10.1037/neu0000833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES Studies are increasingly examining research questions across multiple cohorts using data from the preclinical Alzheimer cognitive composite (PACC). Our objective was to use modern psychometric approaches to develop a harmonized PACC. METHOD We used longitudinal data from the Alzheimer's Disease Neuroimaging Initiative (ADNI), Harvard Aging Brain Study (HABS), and Australian Imaging, Biomarker and Lifestyle Study of Ageing (AIBL) cohorts (n = 2,712). We further demonstrated our method with the Anti-Amyloid Treatment of Asymptomatic Alzheimer's Disease (A4) Study prerandomized data (n = 4,492). For the harmonization method, we used confirmatory factor analysis (CFA) on the final visit of the longitudinal cohorts to determine parameters to generate latent PACC (lPACC) scores. Overlapping tests across studies were set as "anchors" that tied cohorts together, while parameters from unique tests were freely estimated. We performed validation analyses to assess the performance of lPACC versus the common standardized PACC (zPACC). RESULTS Baseline (BL) scores for the zPACC were centered on zero, by definition. The harmonized lPACC did not define a common mean of zero and demonstrated differences in baseline ability levels across the cohorts. Baseline lPACC slightly outperformed zPACC in the prediction of progression to dementia. Longitudinal change in the lPACC was more constrained and less variable relative to the zPACC. In combined-cohort analyses, longitudinal lPACC slightly outperformed longitudinal zPACC in its association with baseline β-amyloid status. CONCLUSIONS This study proposes procedures for harmonizing the PACC that make fewer strong assumptions than the zPACC, facilitating robust multicohort analyses. This implementation of item response theory lends itself to adapting across future cohorts with similar composites. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
- Olivia L. Hampton
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Shubhabrata Mukherjee
- Department of Medicine, Division of General Internal Medicine, University of Washington
| | - Michael J. Properzi
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Aaron P. Schultz
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Paul K. Crane
- Department of Medicine, Division of General Internal Medicine, University of Washington
| | - Laura E. Gibbons
- Department of Medicine, Division of General Internal Medicine, University of Washington
| | - Timothy J. Hohman
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Paul Maruff
- Cogstate Ltd., Melbourne, Victoria, Australia
| | - Yen Ying Lim
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria, Australia
| | - Rebecca E. Amariglio
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Neurology, Brigham and Women’s Hospital, Center for Alzheimer Research and Treatment, Boston, Massachusetts, United States
| | - Kathryn V. Papp
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Neurology, Brigham and Women’s Hospital, Center for Alzheimer Research and Treatment, Boston, Massachusetts, United States
| | - Keith A. Johnson
- Department of Neurology, Brigham and Women’s Hospital, Center for Alzheimer Research and Treatment, Boston, Massachusetts, United States
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Dorene M. Rentz
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Neurology, Brigham and Women’s Hospital, Center for Alzheimer Research and Treatment, Boston, Massachusetts, United States
| | - Reisa A. Sperling
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Neurology, Brigham and Women’s Hospital, Center for Alzheimer Research and Treatment, Boston, Massachusetts, United States
| | - Rachel F. Buckley
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Neurology, Brigham and Women’s Hospital, Center for Alzheimer Research and Treatment, Boston, Massachusetts, United States
- Melbourne School of Psychological Science, University of Melbourne
| |
Collapse
|
36
|
Hansson O, Blennow K, Zetterberg H, Dage J. Blood biomarkers for Alzheimer's disease in clinical practice and trials. NATURE AGING 2023; 3:506-519. [PMID: 37202517 PMCID: PMC10979350 DOI: 10.1038/s43587-023-00403-3] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Blood-based biomarkers hold great promise to revolutionize the diagnostic and prognostic work-up of Alzheimer's disease (AD) in clinical practice. This is very timely, considering the recent development of anti-amyloid-β (Aβ) immunotherapies. Several assays for measuring phosphorylated tau (p-tau) in plasma exhibit high diagnostic accuracy in distinguishing AD from all other neurodegenerative diseases in patients with cognitive impairment. Prognostic models based on plasma p-tau levels can also predict future development of AD dementia in patients with mild cognitive complaints. The use of such high-performing plasma p-tau assays in the clinical practice of specialist memory clinics would reduce the need for more costly investigations involving cerebrospinal fluid samples or positron emission tomography. Indeed, blood-based biomarkers already facilitate identification of individuals with pre-symptomatic AD in the context of clinical trials. Longitudinal measurements of such biomarkers will also improve the detection of relevant disease-modifying effects of new drugs or lifestyle interventions.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Lund, Sweden.
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for 27 Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeffrey Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
37
|
Mattsson-Carlgren N, Salvadó G, Ashton NJ, Tideman P, Stomrud E, Zetterberg H, Ossenkoppele R, Betthauser TJ, Cody KA, Jonaitis EM, Langhough R, Palmqvist S, Blennow K, Janelidze S, Johnson SC, Hansson O. Prediction of Longitudinal Cognitive Decline in Preclinical Alzheimer Disease Using Plasma Biomarkers. JAMA Neurol 2023; 80:360-369. [PMID: 36745413 PMCID: PMC10087054 DOI: 10.1001/jamaneurol.2022.5272] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/12/2022] [Indexed: 02/07/2023]
Abstract
Importance Alzheimer disease (AD) pathology starts with a prolonged phase of β-amyloid (Aβ) accumulation without symptoms. The duration of this phase differs greatly among individuals. While this disease phase has high relevance for clinical trial designs, it is currently unclear how to best predict the onset of clinical progression. Objective To evaluate combinations of different plasma biomarkers for predicting cognitive decline in Aβ-positive cognitively unimpaired (CU) individuals. Design, Setting, and Participants This prospective population-based prognostic study evaluated data from 2 prospective longitudinal cohort studies (the Swedish BioFINDER-1 and the Wisconsin Registry for Alzheimer Prevention [WRAP]), with data collected from February 8, 2010, to October 21, 2020, for the BioFINDER-1 cohort and from August 11, 2011, to June 27, 2021, for the WRAP cohort. Participants were CU individuals recruited from memory clinics who had brain Aβ pathology defined by cerebrospinal fluid (CSF) Aβ42/40 in the BioFINDER-1 study and by Pittsburgh Compound B (PiB) positron emission tomography (PET) in the WRAP study. A total of 564 eligible Aβ-positive and Aβ-negative CU participants with available relevant data from the BioFINDER-1 and WRAP cohorts were included in the study; of those, 171 Aβ-positive participants were included in the main analyses. Exposures Baseline P-tau181, P-tau217, P-tau231, glial fibrillary filament protein, and neurofilament light measured in plasma; CSF biomarkers in the BioFINDER-1 cohort, and PiB PET uptake in the WRAP cohort. Main Outcomes and Measures The primary outcome was longitudinal measures of cognition (using the Mini-Mental State Examination [MMSE] and the modified Preclinical Alzheimer Cognitive Composite [mPACC]) over a median of 6 years (range, 2-10 years). The secondary outcome was conversion to AD dementia. Baseline biomarkers were used in linear regression models to predict rates of longitudinal cognitive change (calculated separately). Models were adjusted for age, sex, years of education, apolipoprotein E ε4 allele status, and baseline cognition. Multivariable models were compared based on model R2 coefficients and corrected Akaike information criterion. Results Among 171 Aβ-positive CU participants included in the main analyses, 119 (mean [SD] age, 73.0 [5.4] years; 60.5% female) were from the BioFINDER-1 study, and 52 (mean [SD] age, 64.4 [4.6] years; 65.4% female) were from the WRAP study. In the BioFINDER-1 cohort, plasma P-tau217 was the best marker to predict cognitive decline in the mPACC (model R2 = 0.41) and the MMSE (model R2 = 0.34) and was superior to the covariates-only models (mPACC: R2 = 0.23; MMSE: R2 = 0.04; P < .001 for both comparisons). Results were validated in the WRAP cohort; for example, plasma P-tau217 was associated with mPACC slopes (R2 = 0.13 vs 0.01 in the covariates-only model; P = .01) and MMSE slopes (R2 = 0.29 vs 0.24 in the covariates-only model; P = .046). Sparse models were identified with plasma P-tau217 as a predictor of cognitive decline. Power calculations for enrichment in hypothetical clinical trials revealed large relative reductions in sample sizes when using plasma P-tau217 to enrich for CU individuals likely to experience cognitive decline over time. Conclusions and Relevance In this study, plasma P-tau217 predicted cognitive decline in patients with preclinical AD. These findings suggest that plasma P-tau217 may be used as a complement to CSF or PET for participant selection in clinical trials of novel disease-modifying treatments.
Collapse
Affiliation(s)
- Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, United Kingdom
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, United Kingdom
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Pontus Tideman
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at University College London, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, Hong Kong SAR, China
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration Program, Amsterdam University Medical Centers, the Netherlands
| | - Tobey J. Betthauser
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Karly Alex Cody
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Erin M. Jonaitis
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Rebecca Langhough
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
38
|
Wessels AM, Dennehy EB, Dowsett SA, Dickson SP, Hendrix SB. Meaningful Clinical Changes in Alzheimer Disease Measured With the iADRS and Illustrated Using the Donanemab TRAILBLAZER-ALZ Study Findings. Neurol Clin Pract 2023; 13:e200127. [PMID: 36891463 PMCID: PMC9987204 DOI: 10.1212/cpj.0000000000200127] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/09/2022] [Indexed: 02/18/2023]
Abstract
Purpose of Review To provide relevant background of the Integrated Alzheimer's Disease Rating Scale (iADRS), with examples, to assist the reader with the interpretation of iADRS findings from the TRAILBLAZER-ALZ study. Recent Findings The iADRS is an integrated measure of global Alzheimer disease (AD) severity for use in the clinical trial environment. It provides a single score that captures commonalities across cognitive and functional ability domains, reflecting disease-related impairment, while minimizing noise not related to disease progression that may exist within each domain. In AD, disease-modifying therapies (DMTs) are expected to slow the rate of clinical decline, changing the trajectory of disease progression. The overall percent slowing of disease progression with treatment is a more informative outcome of effect than absolute point differences between treatment and placebo groups at any given time point because the latter is influenced by treatment period and disease severity. The TRAILBLAZER-ALZ trial was a phase 2 study designed to evaluate the safety and efficacy of donanemab in participants with early symptomatic AD; the primary outcome measure was the change from baseline to 76 weeks on the iADRS. In the TRAILBLAZER-ALZ study, donanemab slowed disease progression by 32% at 18 months (p = 0.04 vs placebo), demonstrating clinical efficacy. At the patient level, one can assess whether the DMT effect is clinically meaningful by estimating the threshold of change consistent with clinically meaningful worsening; based on the TRAILBLAZER-ALZ findings, treatment with donanemab would delay reaching this threshold by approximately 6 months. Summary The iADRS is capable of accurately describing clinical changes associated with disease progression and detecting treatment effects and is an effective assessment tool for use in clinical trials of individuals with early symptomatic AD.
Collapse
Affiliation(s)
- Alette M Wessels
- Eli Lilly and Company (AMW, EBD, SAD), Indianapolis, IN; Department of Psychological Sciences (EBD), Purdue University, West Lafayette, IN; and Pentara Corporation (SPD, SBH), Millcreek, UT
| | - Ellen B Dennehy
- Eli Lilly and Company (AMW, EBD, SAD), Indianapolis, IN; Department of Psychological Sciences (EBD), Purdue University, West Lafayette, IN; and Pentara Corporation (SPD, SBH), Millcreek, UT
| | - Sherie A Dowsett
- Eli Lilly and Company (AMW, EBD, SAD), Indianapolis, IN; Department of Psychological Sciences (EBD), Purdue University, West Lafayette, IN; and Pentara Corporation (SPD, SBH), Millcreek, UT
| | - Samuel P Dickson
- Eli Lilly and Company (AMW, EBD, SAD), Indianapolis, IN; Department of Psychological Sciences (EBD), Purdue University, West Lafayette, IN; and Pentara Corporation (SPD, SBH), Millcreek, UT
| | - Suzanne B Hendrix
- Eli Lilly and Company (AMW, EBD, SAD), Indianapolis, IN; Department of Psychological Sciences (EBD), Purdue University, West Lafayette, IN; and Pentara Corporation (SPD, SBH), Millcreek, UT
| |
Collapse
|
39
|
Caputo A, Racine A, Paule I, Tariot PN, Langbaum JB, Coello N, Riviere ME, Ryan JM, Lopez CL, Graf A. Rationale for the selection of dual primary endpoints in prevention studies of cognitively unimpaired individuals at genetic risk for developing symptoms of Alzheimer's disease. Alzheimers Res Ther 2023; 15:45. [PMID: 36879340 PMCID: PMC9987044 DOI: 10.1186/s13195-023-01183-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND There is a critical need for novel primary endpoints designed to detect early and subtle changes in cognition in clinical trials targeting the asymptomatic (preclinical) phase of Alzheimer's disease (AD). The Alzheimer's Prevention Initiative (API) Generation Program, conducted in cognitively unimpaired individuals at risk of developing AD (e.g., enriched by the apolipoprotein E (APOE) genotype), used a novel dual primary endpoints approach, whereby demonstration of treatment effect in one of the two endpoints is sufficient for trial success. The two primary endpoints were (1) time to event (TTE)-with an event defined as a diagnosis of mild cognitive impairment (MCI) due to AD and/or dementia due to AD-and (2) change from baseline to month 60 in the API Preclinical Composite Cognitive (APCC) test score. METHODS Historical observational data from three sources were used to fit models to describe the TTE and the longitudinal APCC decline, both in people who do and do not progress to MCI or dementia due to AD. Clinical endpoints were simulated based on the TTE and APCC models to assess the performance of the dual endpoints versus each of the two single endpoints, with the selected treatment effect ranging from a hazard ratio (HR) of 0.60 (40% risk reduction) to 1 (no effect). RESULTS A Weibull model was selected for TTE, and power and linear models were selected to describe the APCC score for progressors and non-progressors, respectively. Derived effect sizes in terms of reduction of the APCC change from baseline to year 5 were low (0.186 for HR = 0.67). The power for the APCC alone was consistently lower compared to the power of TTE alone (58% [APCC] vs 84% [TTE] for HR = 0.67). Also, the overall power was higher for the 80%/20% distribution (82%) of the family-wise type 1 error rate (alpha) between TTE and APCC compared to 20%/80% (74%). CONCLUSIONS Dual endpoints including TTE and a measure of cognitive decline perform better than the cognitive decline measure as a single primary endpoint in a cognitively unimpaired population at risk of AD (based on the APOE genotype). Clinical trials in this population, however, need to be large, include older age, and have a long follow-up period of at least 5 years to be able to detect treatment effects.
Collapse
Affiliation(s)
| | - Amy Racine
- Novartis Pharma AG, PostfachCH-4002, Basel, Switzerland
| | - Ines Paule
- Novartis Pharma AG, PostfachCH-4002, Basel, Switzerland
| | | | | | - Neva Coello
- Novartis Pharma AG, PostfachCH-4002, Basel, Switzerland
| | | | | | | | - Ana Graf
- Novartis Pharma AG, PostfachCH-4002, Basel, Switzerland
| |
Collapse
|
40
|
Liu KY, Thambisetty M, Howard R. How can secondary dementia prevention trials of Alzheimer's disease be clinically meaningful? Alzheimers Dement 2023; 19:1073-1085. [PMID: 36161763 PMCID: PMC10039957 DOI: 10.1002/alz.12788] [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: 04/21/2022] [Revised: 07/06/2022] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
After clinical trial failures in symptomatic Alzheimer's disease (AD), our field has moved to earlier intervention in cognitively normal individuals with biomarker evidence of AD. This offers potential for dementia prevention, but mainly low and variable rates of progression to AD dementia reduce the usefulness of trials' data in decision making by potential prescribers. With results from several Phase 3 secondary prevention studies anticipated within the next few years and the Food and Drug Administration's recent endorsement of amyloid beta as a surrogate outcome biomarker for AD clinical trials, it is time to question the clinical significance of changes in biomarkers, adequacy of current trial durations, and criteria for treatment success if cognitively unimpaired patients and their doctors are to meaningfully evaluate the potential value of new agents. We argue for a change of direction toward trial designs that can unambiguously inform clinical decision making about dementia risk and progression.
Collapse
Affiliation(s)
- Kathy Y. Liu
- Division of Psychiatry, University College London, London, UK
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, USA
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
41
|
Insel PS, Young CB, Aisen PS, Johnson KA, Sperling RA, Mormino EC, Donohue MC. Tau positron emission tomography in preclinical Alzheimer's disease. Brain 2023; 146:700-711. [PMID: 35962782 PMCID: PMC10169284 DOI: 10.1093/brain/awac299] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/01/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Rates of tau accumulation in cognitively unimpaired older adults are subtle, with magnitude and spatial patterns varying in recent reports. Regional accumulation also likely varies in the degree to which accumulation is amyloid-β-dependent. Thus, there is a need to evaluate the pattern and consistency of tau accumulation across multiple cognitively unimpaired cohorts and how these patterns relate to amyloid burden, in order to design optimal tau end points for clinical trials. Using three large cohorts of cognitively unimpaired older adults, the Anti-Amyloid Treatment in Asymptomatic Alzheimer's and companion study, Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (n = 447), the Alzheimer's Disease Neuroimaging Initiative (n = 420) and the Harvard Aging Brain Study (n = 190), we attempted to identify regions with high rates of tau accumulation and estimate how these rates evolve over a continuous spectrum of baseline amyloid deposition. Optimal combinations of regions, tailored to multiple ranges of baseline amyloid burden as hypothetical clinical trial inclusion criteria, were tested and validated. The inferior temporal cortex, fusiform gyrus and middle temporal cortex had the largest effect sizes of accumulation in both longitudinal cohorts when considered individually. When tau regions of interest were combined to find composite weights to maximize the effect size of tau change over time, both longitudinal studies exhibited a similar pattern-inferior temporal cortex, almost exclusively, was optimal for participants with mildly elevated amyloid β levels. For participants with highly elevated baseline amyloid β levels, combined optimal composite weights were 53% inferior temporal cortex, 31% amygdala and 16% fusiform. At mildly elevated levels of baseline amyloid β, a sample size of 200/group required a treatment effect of 0.40-0.45 (40-45% slowing of tau accumulation) to power an 18-month trial using the optimized composite. Neither a temporal lobe composite nor a global composite reached 80% power with 200/group with an effect size under 0.5. The focus of early tau accumulation on the medial temporal lobe has resulted from the observation that the entorhinal cortex is the initial site to show abnormal levels of tau with age. However, these abnormal levels do not appear to be the result of a high rate of accumulation in the short term, but possibly a more moderate rate occurring early with respect to age. While the entorhinal cortex plays a central role in the early appearance of tau, it may be the inferior temporal cortex that is the critical region for rapid tau accumulation in preclinical Alzheimer's disease.
Collapse
Affiliation(s)
- Philip S Insel
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Paul S Aisen
- Alzheimer’s Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA, USA
| | - Keith A Johnson
- Department of Neurology, Harvard Aging Brain Study, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Harvard Aging Brain Study, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael C Donohue
- Alzheimer’s Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA, USA
| |
Collapse
|
42
|
Hauber B, Paulsen R, Krasa HB, Vradenburg G, Comer M, Callahan LF, Winfield J, Potashman M, Hartry A, Lee D, Wilson H, Hoffman DL, Wieberg D, Kremer IN, Taylor GA, Taylor JM, Lappin D, Martin AD, Frangiosa T, Biggar V, Slota C, Romano C, DiBenedetti DB. Assessing What Matters to People Affected by Alzheimer's Disease: A Quantitative Analysis. Neurol Ther 2023; 12:505-527. [PMID: 36763306 PMCID: PMC10043143 DOI: 10.1007/s40120-023-00445-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
INTRODUCTION In this phase of the ongoing What Matters Most study series, designed to evaluate concepts that are meaningful to people affected by Alzheimer's disease (AD), we quantified the importance of symptoms, impacts, and outcomes of AD to people at risk for or with AD and care partners of people with AD. METHODS We administered a web-based survey to individuals at risk for or with AD (Group 1: unimpaired cognition with evidence of AD pathology; Group 2: AD risk factors and subjective cognitive complaints/mild cognitive impairment; Group 3: mild AD) and to care partners of individuals with moderate AD (Group 4) or severe AD (Group 5). Respondents rated the importance of 42 symptoms, impacts, and outcomes on a scale ranging from 1 ("not at all important") to 5 ("extremely important"). RESULTS Among the 274 respondents (70.4% female; 63.1% white), over half of patient respondents rated all 42 items as "very important" or "extremely important," while care partners rated fewer items as "very important" or "extremely important." Among the three patient groups, the minimum (maximum) mean importance rating for any item was 3.4 (4.6), indicating that all items were at least moderately to very important. Among care partners of people with moderate or severe AD, the minimum (maximum) mean importance rating was 2.1 (4.4), indicating that most items were rated as at least moderately important. Overall, taking medications correctly, not feeling down or depressed, and staying safe had the highest importance ratings among both patients and care partners, regardless of AD phase. CONCLUSION Concepts of importance to individuals affected by AD go beyond the common understanding of "cognition" or "function" alone, reflecting a desire to maintain independence, overall physical and mental health, emotional well-being, and safety. Preservation of these attributes may be key to understanding whether interventions deliver clinically meaningful outcomes.
Collapse
Affiliation(s)
- Brett Hauber
- RTI Health Solutions, 3040 Cornwallis Road, PO Box 12194, Research Triangle Park, NC, 27709-2194, USA
| | - Russ Paulsen
- UsAgainstAlzheimer's, Washington, DC, 20043, USA
| | - Holly B Krasa
- Blue Persimmon Group LLC, Washington, DC, 20016, USA
| | | | - Meryl Comer
- UsAgainstAlzheimer's, Washington, DC, 20043, USA
| | | | - John Winfield
- University of North Carolina, Chapel Hill, NC, 27599, USA
| | | | | | - Daniel Lee
- Otsuka Pharmaceutical Development and Commercialization Inc., Princeton, NJ, 08540, USA
| | - Hilary Wilson
- Boehringer Ingelheim, Burlington, ON, L7L 5H4, Canada
| | | | | | - Ian N Kremer
- LEAD Coalition (Leaders Engaged on Alzheimer's Disease), Washington, DC, 20043, USA
| | | | | | - Debra Lappin
- Faegre Drinker Consulting, Washington, DC, 20001, USA
| | | | | | | | - Christina Slota
- RTI Health Solutions, 3040 Cornwallis Road, PO Box 12194, Research Triangle Park, NC, 27709-2194, USA
| | - Carla Romano
- RTI Health Solutions, 3040 Cornwallis Road, PO Box 12194, Research Triangle Park, NC, 27709-2194, USA
| | - Dana B DiBenedetti
- RTI Health Solutions, 3040 Cornwallis Road, PO Box 12194, Research Triangle Park, NC, 27709-2194, USA.
| |
Collapse
|
43
|
van Dyck CH, Swanson CJ, Aisen P, Bateman RJ, Chen C, Gee M, Kanekiyo M, Li D, Reyderman L, Cohen S, Froelich L, Katayama S, Sabbagh M, Vellas B, Watson D, Dhadda S, Irizarry M, Kramer LD, Iwatsubo T. Lecanemab in Early Alzheimer's Disease. N Engl J Med 2023; 388:9-21. [PMID: 36449413 DOI: 10.1056/nejmoa2212948] [Citation(s) in RCA: 2233] [Impact Index Per Article: 1116.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND The accumulation of soluble and insoluble aggregated amyloid-beta (Aβ) may initiate or potentiate pathologic processes in Alzheimer's disease. Lecanemab, a humanized IgG1 monoclonal antibody that binds with high affinity to Aβ soluble protofibrils, is being tested in persons with early Alzheimer's disease. METHODS We conducted an 18-month, multicenter, double-blind, phase 3 trial involving persons 50 to 90 years of age with early Alzheimer's disease (mild cognitive impairment or mild dementia due to Alzheimer's disease) with evidence of amyloid on positron-emission tomography (PET) or by cerebrospinal fluid testing. Participants were randomly assigned in a 1:1 ratio to receive intravenous lecanemab (10 mg per kilogram of body weight every 2 weeks) or placebo. The primary end point was the change from baseline at 18 months in the score on the Clinical Dementia Rating-Sum of Boxes (CDR-SB; range, 0 to 18, with higher scores indicating greater impairment). Key secondary end points were the change in amyloid burden on PET, the score on the 14-item cognitive subscale of the Alzheimer's Disease Assessment Scale (ADAS-cog14; range, 0 to 90; higher scores indicate greater impairment), the Alzheimer's Disease Composite Score (ADCOMS; range, 0 to 1.97; higher scores indicate greater impairment), and the score on the Alzheimer's Disease Cooperative Study-Activities of Daily Living Scale for Mild Cognitive Impairment (ADCS-MCI-ADL; range, 0 to 53; lower scores indicate greater impairment). RESULTS A total of 1795 participants were enrolled, with 898 assigned to receive lecanemab and 897 to receive placebo. The mean CDR-SB score at baseline was approximately 3.2 in both groups. The adjusted least-squares mean change from baseline at 18 months was 1.21 with lecanemab and 1.66 with placebo (difference, -0.45; 95% confidence interval [CI], -0.67 to -0.23; P<0.001). In a substudy involving 698 participants, there were greater reductions in brain amyloid burden with lecanemab than with placebo (difference, -59.1 centiloids; 95% CI, -62.6 to -55.6). Other mean differences between the two groups in the change from baseline favoring lecanemab were as follows: for the ADAS-cog14 score, -1.44 (95% CI, -2.27 to -0.61; P<0.001); for the ADCOMS, -0.050 (95% CI, -0.074 to -0.027; P<0.001); and for the ADCS-MCI-ADL score, 2.0 (95% CI, 1.2 to 2.8; P<0.001). Lecanemab resulted in infusion-related reactions in 26.4% of the participants and amyloid-related imaging abnormalities with edema or effusions in 12.6%. CONCLUSIONS Lecanemab reduced markers of amyloid in early Alzheimer's disease and resulted in moderately less decline on measures of cognition and function than placebo at 18 months but was associated with adverse events. Longer trials are warranted to determine the efficacy and safety of lecanemab in early Alzheimer's disease. (Funded by Eisai and Biogen; Clarity AD ClinicalTrials.gov number, NCT03887455.).
Collapse
Affiliation(s)
- Christopher H van Dyck
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Chad J Swanson
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Paul Aisen
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Randall J Bateman
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Christopher Chen
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Michelle Gee
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Michio Kanekiyo
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - David Li
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Larisa Reyderman
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Sharon Cohen
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Lutz Froelich
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Sadao Katayama
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Marwan Sabbagh
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Bruno Vellas
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - David Watson
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Shobha Dhadda
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Michael Irizarry
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Lynn D Kramer
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| | - Takeshi Iwatsubo
- From the Alzheimer's Disease Research Unit, Yale School of Medicine, New Haven, CT (C.H.D.); Eisai, Nutley, NJ (C.J.S., M.K., D.L., L.R., S.D., M.I., L.D.K.); the Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego (P.A.); Washington University School of Medicine in St. Louis, St. Louis (R.B.); the Memory, Aging, and Cognition Center, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.C.); Eisai, Hatfield, United Kingdom (M.G.); Toronto Memory Program, Toronto (S.C.); Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Germany (L.F.); Katayama Medical Clinic, Okayama (S.K.), and the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, and the National Center of Neurology and Psychiatry, Tokyo (T.I.) - all in Japan; Barrow Neurological Institute, Phoenix, AZ (M.S.); Toulouse Gerontopole University Hospital, Université Paul Sabatier, INSERM Unité 1295, Toulouse, France (B.V.); and Alzheimer's Research and Treatment Center, Wellington, FL (D.W.)
| |
Collapse
|
44
|
The treatment of sleep dysfunction to improve cognitive function: A meta-analysis of randomized controlled trials. Sleep Med 2023; 101:118-126. [PMID: 36370516 DOI: 10.1016/j.sleep.2022.10.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/12/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE This meta-analysis of randomized controlled trials (RCTs) evaluates if treating sleep disturbances improves cognitive function over at least 12 weeks. METHODS Multiple data sources were searched until November 1, 2021. RCTs were included if they examined the effect of an intervention (behavioral or medical) on sleep and cognition in an adult sample with sleep disturbances and had an intervention duration and follow-up of at least 12 weeks. Two independent reviewers located 3784 studies; 16 satisfied the inclusion criteria. Primary outcomes included the broad cognitive domains of visual processing, short-term memory, long-term storage and retrieval, processing speed, and reaction time. RESULTS Most trials were conducted in participants with obstructive sleep apnea (OSA; N = 13); the most studied intervention was continuous positive airway pressure (CPAP; N = 10). All RCTs were 12 months in duration or less. The estimates of mean pooled effects were not indicative of significant treatment effect for any primary outcome. Although the interventions reduced daytime sleepiness (Hedge's g, 0.51; 95% confidence interval, 0.29-0.74; p < 0.01), this did not lead to cognitive enhancement. CONCLUSIONS Overall, there was insufficient evidence to suggest that treating sleep dysfunction can improve cognition. Further studies with longer follow-up duration and supporting biomarkers are needed.
Collapse
|
45
|
Dhiman K, Villemagne VL, Fowler C, Bourgeat P, Li QX, Collins S, Rowe CC, Masters CL, Ames D, Blennow K, Zetterberg H, Martins RN, Gupta V. Cerebrospinal fluid levels of fatty acid-binding protein 3 are associated with likelihood of amyloidopathy in cognitively healthy individuals. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12377. [PMID: 36479019 PMCID: PMC9719998 DOI: 10.1002/dad2.12377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 12/12/2022]
Abstract
Introduction Fatty acid-binding protein 3 (FABP3) is a biomarker of neuronal membrane disruption, associated with lipid dyshomeostasis-a notable Alzheimer's disease (AD) pathophysiological change. We assessed the association of cerebrospinal fluid (CSF) FABP3 levels with brain amyloidosis and the likelihood/risk of developing amyloidopathy in cognitively healthy individuals. Methods FABP3 levels were measured in CSF samples of cognitively healthy participants, > 60 years of age (n = 142), from the Australian Imaging, Biomarkers & Lifestyle Flagship Study of Ageing (AIBL). Results FABP3 levels were positively associated with baseline brain amyloid beta (Aβ) load as measured by standardized uptake value ratio (SUVR, standardized β = 0.22, P = .009) and predicted the change in brain Aβ load (standardized β = 0.32, P = .004). Higher levels of CSF FABP3 (above median) were associated with a likelihood of amyloidopathy (odds ratio [OR] 2.28, 95% confidence interval [CI] 1.12 to 4.65, P = .023). Discussion These results support inclusion of CSF FABP3 as a biomarker in risk-prediction models of AD.
Collapse
Affiliation(s)
- Kunal Dhiman
- IMPACT - The Institute for Mental and Physical Health and Clinical Translation School of Medicine Deakin University Geelong Victoria Australia
- Western Health Partnership School of Nursing and Midwifery (Centre for Quality and Patient Safety Research in the Institute of Health Transformation) Faculty of Health Deakin University Melbourne Victoria Australia
- School of Medical and Health Sciences Edith Cowan University Joondalup Western Australia Australia
| | - Victor L Villemagne
- Department of Psychiatry University of Pittsburgh Pittsburgh Pennsylvania USA
- Department of Molecular Imaging & Therapy and Centre for PET Austin Health Heidelberg Victoria Australia
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria Australia
| | - Pierrick Bourgeat
- Australian e-Health Research Centre CSIRO Health and Biosecurity Brisbane Queensland Australia
| | - Qiao-Xin Li
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria Australia
| | - Steven Collins
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy and Centre for PET Austin Health Heidelberg Victoria Australia
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria Australia
| | - David Ames
- National Ageing Research Institute Parkville Victoria Australia
- Academic Unit for Psychiatry of Old age St. George's Hospital The University of Melbourne Melbourne Victoria Australia
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology the Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Gothenburg Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology the Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Gothenburg Sweden
- Department of Neurodegenerative Disease UCL Queen Square Institute of Neurology London UK
- UK Dementia Research Institute at UCL London UK
- Hong Kong Center for Neurodegenerative Diseases Hong Kong China
| | - Ralph N Martins
- School of Medical and Health Sciences Edith Cowan University Joondalup Western Australia Australia
- Australian Alzheimer's Research Foundation Ralph and Patricia Sarich Neuroscience Research Institute Nedlands Western Australia Australia
- Department of Biomedical Sciences Macquarie University Sydney New South Wales Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Perth Western Australia Australia
- KaRa Institute of Neurological Diseases Sydney New South Wales Australia
- Co-operative Research Centre for Mental Health Carlton Victoria Australia
| | - Veer Gupta
- IMPACT - The Institute for Mental and Physical Health and Clinical Translation School of Medicine Deakin University Geelong Victoria Australia
- School of Medical and Health Sciences Edith Cowan University Joondalup Western Australia Australia
| |
Collapse
|
46
|
Hansson O, Edelmayer RM, Boxer AL, Carrillo MC, Mielke MM, Rabinovici GD, Salloway S, Sperling R, Zetterberg H, Teunissen CE. The Alzheimer's Association appropriate use recommendations for blood biomarkers in Alzheimer's disease. Alzheimers Dement 2022; 18:2669-2686. [PMID: 35908251 PMCID: PMC10087669 DOI: 10.1002/alz.12756] [Citation(s) in RCA: 308] [Impact Index Per Article: 102.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/28/2022] [Accepted: 07/08/2022] [Indexed: 01/31/2023]
Abstract
Blood-based markers (BBMs) have recently shown promise to revolutionize the diagnostic and prognostic work-up of Alzheimer's disease (AD), as well as to improve the design of interventional trials. Here we discuss in detail further research needed to be performed before widespread use of BBMs. We already now recommend use of BBMs as (pre-)screeners to identify individuals likely to have AD pathological changes for inclusion in trials evaluating disease-modifying therapies, provided the AD status is confirmed with positron emission tomography (PET) or cerebrospinal fluid (CSF) testing. We also encourage studying longitudinal BBM changes in ongoing as well as future interventional trials. However, BBMs should not yet be used as primary endpoints in pivotal trials. Further, we recommend to cautiously start using BBMs in specialized memory clinics as part of the diagnostic work-up of patients with cognitive symptoms and the results should be confirmed whenever possible with CSF or PET. Additional data are needed before use of BBMs as stand-alone diagnostic AD markers, or before considering use in primary care.
Collapse
Affiliation(s)
- Oskar Hansson
- ClinicalMemory Research UnitDepartment of Clinical Sciences MalmöLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | | | - Adam L. Boxer
- Department of NeurologyUniversity of California San FranciscoMemory and Aging CenterSan FranciscoCaliforniaUSA
| | | | - Michelle M. Mielke
- Department of Epidemiology and PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Gil D. Rabinovici
- Department of NeurologyUniversity of California San FranciscoMemory and Aging CenterSan FranciscoCaliforniaUSA
| | - Stephen Salloway
- Departments of Neurology and PsychiatryAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Reisa Sperling
- Centerfor Alzheimer Research and TreatmentBrigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongPeople's Republic of China
| | - Charlotte E. Teunissen
- NeurochemistryLaboratoryDepartment of Clinical ChemistryAmsterdam University Medical CentersVrije UniversiteitAmsterdam NeuroscienceAmsterdamthe Netherlands
| |
Collapse
|
47
|
Ashton NJ, Janelidze S, Mattsson-Carlgren N, Binette AP, Strandberg O, Brum WS, Karikari TK, González-Ortiz F, Di Molfetta G, Meda FJ, Jonaitis EM, Koscik RL, Cody K, Betthauser TJ, Li Y, Vanmechelen E, Palmqvist S, Stomrud E, Bateman RJ, Zetterberg H, Johnson SC, Blennow K, Hansson O. Differential roles of Aβ42/40, p-tau231 and p-tau217 for Alzheimer's trial selection and disease monitoring. Nat Med 2022; 28:2555-2562. [PMID: 36456833 PMCID: PMC9800279 DOI: 10.1038/s41591-022-02074-w] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 10/03/2022] [Indexed: 12/03/2022]
Abstract
Blood biomarkers indicative of Alzheimer's disease (AD) pathology are altered in both preclinical and symptomatic stages of the disease. Distinctive biomarkers may be optimal for the identification of AD pathology or monitoring of disease progression. Blood biomarkers that correlate with changes in cognition and atrophy during the course of the disease could be used in clinical trials to identify successful interventions and thereby accelerate the development of efficient therapies. When disease-modifying treatments become approved for use, efficient blood-based biomarkers might also inform on treatment implementation and management in clinical practice. In the BioFINDER-1 cohort, plasma phosphorylated (p)-tau231 and amyloid-β42/40 ratio were more changed at lower thresholds of amyloid pathology. Longitudinally, however, only p-tau217 demonstrated marked amyloid-dependent changes over 4-6 years in both preclinical and symptomatic stages of the disease, with no such changes observed in p-tau231, p-tau181, amyloid-β42/40, glial acidic fibrillary protein or neurofilament light. Only longitudinal increases of p-tau217 were also associated with clinical deterioration and brain atrophy in preclinical AD. The selective longitudinal increase of p-tau217 and its associations with cognitive decline and atrophy was confirmed in an independent cohort (Wisconsin Registry for Alzheimer's Prevention). These findings support the differential association of plasma biomarkers with disease development and strongly highlight p-tau217 as a surrogate marker of disease progression in preclinical and prodromal AD, with impact for the development of new disease-modifying treatments.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Shorena Janelidze
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fernándo González-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Francisco J Meda
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Rebecca Langhough Koscik
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Karly Cody
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Sebastian Palmqvist
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- ADx NeuroSciences, Technologiepark 94, Ghent, Belgium.
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
48
|
Reed RG, Carroll JE, Marsland AL, Manuck SB. DNA methylation-based measures of biological aging and cognitive decline over 16-years: preliminary longitudinal findings in midlife. Aging (Albany NY) 2022; 14:9423-9444. [PMID: 36374219 PMCID: PMC9792211 DOI: 10.18632/aging.204376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/29/2022] [Indexed: 11/13/2022]
Abstract
DNA methylation-based (DNAm) measures of biological aging associate with increased risk of morbidity and mortality, but their links with cognitive decline are less established. This study examined changes over a 16-year interval in epigenetic clocks (the traditional and principal components [PC]-based Horvath, Hannum, PhenoAge, GrimAge) and pace of aging measures (Dunedin PoAm, Dunedin PACE) in 48 midlife adults enrolled in the longitudinal arm of the Adult Health and Behavior project (56% Female, baseline AgeM = 44.7 years), selected for discrepant cognitive trajectories. Cognitive Decliners (N = 24) were selected based on declines in a composite score derived from neuropsychological tests and matched with participants who did not show any decline, Maintainers (N = 24). Multilevel models with repeated DNAm measures within person tested the main effects of time, group, and group by time interactions. DNAm measures significantly increased over time generally consistent with elapsed time between study visits. There were also group differences: overall, Cognitive Decliners had an older PC-GrimAge and faster pace of aging (Dunedin PoAm, Dunedin PACE) than Cognitive Maintainers. There were no significant group by time interactions, suggesting accelerated epigenetic aging in Decliners remained constant over time. Older PC-GrimAge and faster pace of aging may be particularly sensitive to cognitive decline in midlife.
Collapse
Affiliation(s)
- Rebecca G. Reed
- Department of Psychology, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Judith E. Carroll
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry and Biobehavioral Science, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Anna L. Marsland
- Department of Psychology, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Stephen B. Manuck
- Department of Psychology, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
49
|
Pichet Binette A, Franzmeier N, Spotorno N, Ewers M, Brendel M, Biel D, Strandberg O, Janelidze S, Palmqvist S, Mattsson-Carlgren N, Smith R, Stomrud E, Ossenkoppele R, Hansson O. Amyloid-associated increases in soluble tau relate to tau aggregation rates and cognitive decline in early Alzheimer's disease. Nat Commun 2022; 13:6635. [PMID: 36333294 PMCID: PMC9636262 DOI: 10.1038/s41467-022-34129-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
For optimal design of anti-amyloid-β (Aβ) and anti-tau clinical trials, we need to better understand the pathophysiological cascade of Aβ- and tau-related processes. Therefore, we set out to investigate how Aβ and soluble phosphorylated tau (p-tau) relate to the accumulation of tau aggregates assessed with PET and subsequent cognitive decline across the Alzheimer's disease (AD) continuum. Using human cross-sectional and longitudinal neuroimaging and cognitive assessment data, we show that in early stages of AD, increased concentration of soluble CSF p-tau is strongly associated with accumulation of insoluble tau aggregates across the brain, and CSF p-tau levels mediate the effect of Aβ on tau aggregation. Further, higher soluble p-tau concentrations are mainly related to faster accumulation of tau aggregates in the regions with strong functional connectivity to individual tau epicenters. In this early stage, higher soluble p-tau concentrations is associated with cognitive decline, which is mediated by faster increase of tau aggregates. In contrast, in AD dementia, when Aβ fibrils and soluble p-tau levels have plateaued, cognitive decline is related to the accumulation rate of insoluble tau aggregates. Our data suggest that therapeutic approaches reducing soluble p-tau levels might be most favorable in early AD, before widespread insoluble tau aggregates.
Collapse
Affiliation(s)
- Alexa Pichet Binette
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden.
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Nicola Spotorno
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Olof Strandberg
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Department of Neurology, Skåne University Hospital, Lund, 205 02, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Department of Neurology, Skåne University Hospital, Lund, 205 02, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund, 205 02, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
50
|
Svenningsson AL, Stomrud E, Palmqvist S, Hansson O, Ossenkoppele R. Axonal degeneration and amyloid pathology predict cognitive decline beyond cortical atrophy. Alzheimers Res Ther 2022; 14:144. [PMID: 36192766 PMCID: PMC9531524 DOI: 10.1186/s13195-022-01081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 09/11/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Cortical atrophy is associated with cognitive decline, but the association is not perfect. We aimed to identify factors explaining the discrepancy between the degree of cortical atrophy and cognitive decline in cognitively unimpaired elderly. METHODS The discrepancy between atrophy and cognitive decline was measured using the residuals from a linear regression analysis between change in whole brain cortical thickness over time and change in a cognitive composite measure over time in 395 cognitively unimpaired participants from the Swedish BioFINDER study. We tested for bivariate associations of this residual measure with demographic, imaging, and fluid biomarker variables using Pearson correlations and independent-samples t-tests, and for multivariate associations using linear regression models. Mediation analyses were performed to explore possible paths between the included variables. RESULTS In bivariate analyses, older age (r = -0.11, p = 0.029), male sex (t = -3.00, p = 0.003), larger intracranial volume (r = -0.17, p < 0.001), carrying an APOEe4 allele (t = -2.71, p = 0.007), larger white matter lesion volume (r = -0.16, p = 0.002), lower cerebrospinal fluid (CSF) β-amyloid (Aβ) 42/40 ratio (t = -4.05, p < 0.001), and higher CSF levels of phosphorylated tau (p-tau) 181 (r = -0.22, p < 0.001), glial fibrillary acidic protein (GFAP; r = -0.15, p = 0.003), and neurofilament light (NfL; r = -0.34, p < 0.001) were negatively associated with the residual measure, i.e., associated with worse than expected cognitive trajectory given the level of atrophy. In a multivariate analysis, only lower CSF Aβ42/40 ratio and higher CSF NfL levels explained cognition beyond brain atrophy. Mediation analyses showed that associations between the residual measure and APOEe4 allele, CSF Aβ42/40 ratio, and CSF GFAP and p-tau181 levels were mediated by levels of CSF NfL, as were the associations with the residual measure for age, sex, and WML volume. CONCLUSIONS Our results suggest that axonal degeneration and amyloid pathology independently affect the rate of cognitive decline beyond the degree of cortical atrophy. Furthermore, axonal degeneration mediated the negative effects of old age, male sex, and white matter lesions, and in part also amyloid and tau pathology, on cognition over time when accounting for cortical atrophy.
Collapse
Affiliation(s)
- Anna Linnéa Svenningsson
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.484519.5Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
| |
Collapse
|