1
|
Lantero-Rodriguez J, Montoliu-Gaya L, Ashton NJ, Pola I, Therriault J, Rahmouni N, Brum WS, Servaes S, Stevenson J, Di Molfetta G, Arslan B, Klostranec J, Vitali P, Montembeault M, Gauthier S, Tissot C, Macedo AC, Pascoal TA, Jeromin A, Gobom J, Blennow K, Zetterberg H, Rosa-Neto P, Benedet AL. Biofluid-based staging of Alzheimer's disease. Acta Neuropathol 2025; 149:27. [PMID: 40095069 PMCID: PMC11913990 DOI: 10.1007/s00401-025-02863-w] [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: 11/06/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Recently, conceptual systems for the in vivo staging of Alzheimer's disease (AD) using fluid biomarkers have been suggested. Thus, it is important to assess whether available fluid biomarkers can successfully stage AD into clinically and biologically relevant categories. In the TRIAD cohort, we explored whether p-tau217, p-tau205 and NTA-tau (biomarkers of early, intermediate and late AD pathology, respectively) have potential for biofluid-based staging in cerebrospinal fluid (CSF; n = 219) and plasma (n = 150), and compared them in a paired CSF and plasma subset (n = 76). Our findings suggest a good concordance between biofluid staging and underlying pathology when classifying amyloid-positivity into three categories based on neurofibrillary pathology: minimal/non-existent (p-tau217 positive), early-to-intermediate (p-tau217 and p-tau205 positivity), and advanced tau tangle deposition (p-tau217, p-tau205 and NTA-tau positive), as indexed by tau-PET. Discordant cases accounted for 4.6% and 13.3% of all CSF and plasma measurements respectively (9.2% and 11.8% in paired samples). Notably, CSF- and plasma-based staging matched one another in 61.7% of the cases, while approximately 32% of the remaining participants were one to three biofluid stages higher in CSF as compared to plasma. Overall, these exploratory results suggest that biofluid staging of AD holds potential for offering valuable insights into underlying AD hallmarks and disease severity. However, its applicability beyond molecular characterization at research settings has yet to be demonstrated.
Collapse
Affiliation(s)
- Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Ilaria Pola
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Joseph Therriault
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & 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
| | - Stijn Servaes
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Jenna Stevenson
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jesse Klostranec
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Paolo Vitali
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Maxime Montembeault
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Cecile Tissot
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Arthur C Macedo
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | | | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for 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
| | - Pedro Rosa-Neto
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
2
|
Antonioni A, Raho EM, Di Lorenzo F, Manzoli L, Flacco ME, Koch G. Blood phosphorylated Tau217 distinguishes amyloid-positive from amyloid-negative subjects in the Alzheimer's disease continuum. A systematic review and meta-analysis. J Neurol 2025; 272:252. [PMID: 40047958 PMCID: PMC11885345 DOI: 10.1007/s00415-025-12996-3] [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: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia worldwide, and cost-effective tools to detect amyloid pathology are urgently needed. Blood-based Tau phosphorylated at threonine 217 (pTau217) seems promising, but its reliability as a proxy for cerebrospinal fluid (CSF) status and ability to identify patients within the AD spectrum remain unclear. METHODS We performed a systematic review and meta-analysis on the potential of blood pTau217 to differentiate amyloid-positive (A+) and amyloid-negative (A-) subjects. We included original studies reporting quantitative data on pTau217 concentrations in both blood and CSF in the AD continuum. The single-group meta-analysis computed the pooled pTau217 levels in blood and in CSF, separately in the A+ and A- groups, while the head-to-head meta-analysis compared the mean pTau217 concentrations in the A+ versus A- subjects, both in blood and CSF, stratifying by assessment method in both cases. RESULTS Ten studies (819 A+; 1055 A-) were included. The mean pTau217 levels resulted higher in CSF than in blood and, crucially, in A+ individuals than in A- ones, regardless of the laboratory method employed. Most importantly, all laboratory techniques reliably distinguished A+ from A- subjects, whether applied to CSF or blood samples. CONCLUSIONS These results confirm that blood-based pTau217 is a reliable marker of amyloid pathology with significant implications for clinical practice in the AD continuum. Indeed, pTau217 might be a non-invasive, scalable biomarker for early AD detection, reducing the reliance on more invasive, expansive, and less accessible methods. CLINICAL TRIAL REGISTRATION Prospero CRD42024565187.
Collapse
Affiliation(s)
- Annibale Antonioni
- Doctoral Program in Translational Neurosciences and Neurotechnologies, Department of Neuroscience and Rehabilitation, University of Ferrara, Via Ludovico Ariosto, 35, 44121, Ferrara, Italy.
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy.
- Department of Neuroscience, Ferrara University Hospital, 44124, Ferrara, Italy.
| | - Emanuela Maria Raho
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
- Department of Neuroscience, Ferrara University Hospital, 44124, Ferrara, Italy
| | - Francesco Di Lorenzo
- Neuropsychophysiology Lab, Santa Lucia Foundation IRCCS, Via Ardeatina, 306, 00179, Rome, Italy.
| | - Lamberto Manzoli
- Department of Medical and Surgical Sciences, University of Bologna, 40126, Bologna, Italy
| | - Maria Elena Flacco
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Giacomo Koch
- Neuropsychophysiology Lab, Santa Lucia Foundation IRCCS, Via Ardeatina, 306, 00179, Rome, Italy.
- Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy.
- Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, 44121, Ferrara, Italy.
| |
Collapse
|
3
|
Yun J, Shin D, Lee EH, Kim JP, Ham H, Gu Y, Chun MY, Kang SH, Kim HJ, Na DL, Kim CH, Kim KW, Kim SE, Kim Y, Kim J, Jung NY, Kim YJ, Cho SH, Zetterberg H, Blennow K, Gonzalez-Ortiz F, Ashton NJ, Therriault J, Rahmouni N, Rosa-Neto P, Weiner MW, Seo SW, Jang H. Temporal Dynamics and Biological Variability of Alzheimer Biomarkers. JAMA Neurol 2025:2830247. [PMID: 39960728 PMCID: PMC11833660 DOI: 10.1001/jamaneurol.2024.5263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/10/2024] [Indexed: 02/20/2025]
Abstract
Importance Understanding the characteristics of discordance between plasma biomarkers and positron emission tomography (PET) results in Alzheimer disease (AD) is crucial for accurate interpretation of the findings. Objective To compare (1) medical comorbidities affecting plasma biomarker concentrations, (2) imaging and clinical features, and (3) cognitive changes between plasma biomarker and PET discordant and concordant cases. Design, Setting, and Participants This multicenter cohort study, conducted between 2016 and 2023, included individuals with unimpaired cognition, mild cognitive impairment, or Alzheimer-type dementia, who had both amyloid β (Aβ) PET imaging and plasma biomarkers. A subset of participants also underwent tau PET imaging. Exposures Participants were categorized into 4 groups based on their plasma and PET biomarker results: plasma-/PET-, plasma+/PET-, plasma-/PET+, and plasma+/PET+. Main Outcomes and Measures Clinical characteristics were compared between the 4 groups, focusing on the discordant groups. Results A total of 2611 participants (mean [SD] age was 71.2 [8.7] years; 1656 female [63.4%]), of whom 124 additionally underwent tau PET, were included. Among the plasma biomarkers, phosphorylated tau (p-tau) 217 exhibited the highest concordance rate with Aβ (2326 of 2571 [90.5%]) and tau (100 of 120 [83.3%]) PET. The p-tau217+/Aβ PET- group was older (mean [SD] age, 75.8 [7.2] years vs 70.0 [8.8] years; P < .001) with a higher prevalence of hypertension (56 of 152 [36.8%] vs 266 of 1073 [25.0%]), diabetes (40 of 152 [26.3%] vs 156 of 1059 [14.7%]), and chronic kidney disease (17 of 152 [11.2%] vs 21 of 1073 [2.0%]) compared with the p-tau217-/Aβ PET- group (P < .001 for all). Body mass index was higher in p-tau217-/Aβ PET+ than in p-tau217+/Aβ PET+ (mean [SD], 24.1 [2.8] vs 23.1 [3.1], respectively; P = .001; calculated as weight in kilograms divided by height in meters squared). The p-tau217+/Aβ PET- group had lower hippocampal volume (mean [SD], 2555.4 [576.9] vs 2979.1 [545.8]; P < .001) and worse clinical trajectory compared with p-tau217-/Aβ PET- (β = -0.53; P < .001). In contrast, tau PET discordant cases did not show significant differences in medical comorbidities or clinical outcomes compared with the p-tau217-/tau PET- group. Only the p-tau 217+/tau PET+ group demonstrated faster cognitive deterioration compared with the p-tau 217-/tau PET- group (β = -1.66; P < .001). Conclusions and Relevance Results of this cohort study suggest that the mechanisms underlying the discordance between plasma biomarkers and PET findings may be multifaceted, underscoring the need to consider the temporal dynamics and biological variability of plasma biomarkers.
Collapse
Affiliation(s)
- Jihwan Yun
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
| | - Daeun Shin
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Eun Hye Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hongki Ham
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University. Seoul, South Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, South Korea
| | - Yuna Gu
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Min Young Chun
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Sung Hoon Kang
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University. Seoul, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Happymid Clinic, Seoul, South Korea
| | - Chi-Hun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Anyang, South Korea
| | - Ko Woon Kim
- Department of Neurology, Jeonbuk National University Medical School and Hospital, Jeonju, Korea
| | - Si Eun Kim
- Department of Neurology, Inje University College of Medicine, Haeundae Paik Hospital, Busan, Korea
| | - Yeshin Kim
- Department of Neurology, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Jaeho Kim
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Yeo Jin Kim
- Department of Neurology, Kangwon National University College of Medicine, Chuncheon-si, Korea
| | - Soo Hyun Cho
- Department of Neurology, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju, Korea
| | - 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 Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792
| | - 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
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People’s Republic of China
| | - Fernando Gonzalez-Ortiz
- 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
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl 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
| | - Joseph Therriault
- McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Michael W. Weiner
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University. Seoul, South Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, South Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Hyemin Jang
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
4
|
Islam T, Hill E, Abrahamson EE, Servaes S, Smirnov DS, Zeng X, Sehrawat A, Chen Y, Kac PR, Kvartsberg H, Olsson M, Sjons E, Gonzalez-Ortiz F, Therriault J, Tissot C, Del Popolo I, Rahmouni N, Richardson A, Mitchell V, Zetterberg H, Pascoal TA, Lashley T, Wall MJ, Galasko D, Rosa-Neto P, Ikonomovic MD, Blennow K, Karikari TK. Phospho-tau serine-262 and serine-356 as biomarkers of pre-tangle soluble tau assemblies in Alzheimer's disease. Nat Med 2025; 31:574-588. [PMID: 39930142 PMCID: PMC11835754 DOI: 10.1038/s41591-024-03400-0] [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: 03/28/2024] [Accepted: 11/02/2024] [Indexed: 02/20/2025]
Abstract
Patients with Alzheimer's disease (AD) with little or no quantifiable insoluble brain tau neurofibrillary tangle (NFT) pathology demonstrate stronger clinical benefits of therapies than those with advanced NFTs. The formation of NFTs can be prevented by targeting the intermediate soluble tau assemblies (STAs). However, biochemical understanding and biomarkers of STAs are lacking. We show that Tris-buffered saline-soluble tau aggregates from autopsy-verified AD brain tissues include the core sequence ~tau258-368. In neuropathological assessments, antibodies against the phosphorylation sites serine-262 and serine-356 within the STA core almost exclusively stained granular (that is, prefibrillar) tau aggregates in pre-NFTs while antibodies against phosphorylation at serine-202 and threonine-205 and threonine-231, outside the STA core, stained the entire spectrum of tau aggregates in pre-NFTs and mature NFTs, dystrophic neurites and neuropil threads in the hippocampus. Functionally, a recombinantly produced STA core peptide robustly altered neuronal excitability and synaptic transmission in mouse hippocampal brain slices. Furthermore, we developed a cerebrospinal fluid assay that differentiated STAs in AD from non-AD tauopathies, correlated with the severity of NFT burden and cognitive decline independently of amyloid beta deposition, and with tau positron emission tomography uptake across Braak NFT stages. Together, our findings inform about the status of early-stage tau aggregation, reveal aggregation-relevant phosphorylation epitopes in tau and offer a diagnostic biomarker and targeted therapeutic opportunities for AD.
Collapse
Grants
- R01 AG075336 NIA NIH HHS
- R01 AG083874 NIA NIH HHS
- R01 AG072641 NIA NIH HHS
- P30 AG062429 NIA NIH HHS
- AARF-21-850325 Alzheimer's Association
- P01AG14449 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- P01 AG014449 NIA NIH HHS
- P50 AG005133 NIA NIH HHS
- RF1 AG025516 NIA NIH HHS
- P30 AG066468 NIA NIH HHS
- R01 AG073267 NIA NIH HHS
- P01 AG025204 NIA NIH HHS
- R01AG083874 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R37 AG023651 NIA NIH HHS
- U24 AG082930 NIA NIH HHS
- 2021-03244 Vetenskapsrådet (Swedish Research Council)
- P01AG025204 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- P30AG066468 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- RF1 AG052525 NIA NIH HHS
- R01 AG053952 NIA NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- ELH is supported by a Race Against Dementia Fellowship (funded by the Barbara Naylor Foundation)
- HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2022-01018 and #2019-02397), the European Union’s Horizon Europe research and innovation programme under grant agreement No 101053962, Swedish State Support for Clinical Research (#ALFGBG-71320), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), the AD Strategic Fund and the Alzheimer's Association (#ADSF-21-831376-C, #ADSF-21-831381-C, and #ADSF-21-831377-C), the Bluefield Project, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2022-0270), the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 860197 (MIRIADE), the European Union Joint Programme – Neurodegenerative Disease Research (JPND2021-00694), the National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, and the UK Dementia Research Institute at UCL (UKDRI-1003)
Collapse
Affiliation(s)
- Tohidul Islam
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Emily Hill
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Eric E Abrahamson
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre (BIC), Montréal Neurological Institute, Montréal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Denis S Smirnov
- Shiley-Marcos Alzheimer's Disease Research Center, Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
- Pathology Residency Program, Mass General and Brigham and Women's Hospitals, Harvard Medical School, Boston, MA, USA
| | - Xuemei Zeng
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anuradha Sehrawat
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yijun Chen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Przemysław R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Hlin Kvartsberg
- 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
| | - Maria Olsson
- 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
| | - Emma Sjons
- 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
| | - Fernando Gonzalez-Ortiz
- 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
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre (BIC), Montréal Neurological Institute, Montréal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre (BIC), Montréal Neurological Institute, Montréal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | | | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre (BIC), Montréal Neurological Institute, Montréal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | | | | | - 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 Diseases, Queen Square Institute of Neurology UCL, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Tharick A Pascoal
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, UCL, London, UK
| | - Mark J Wall
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Douglas Galasko
- Shiley-Marcos Alzheimer's Disease Research Center, Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre (BIC), Montréal Neurological Institute, Montréal, Québec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Québec, Canada
| | - Milos D Ikonomovic
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - 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
| | - 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, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Quispialaya KM, Therriault J, Aliaga A, Benedet AL, Ashton NJ, Karikari T, Cassa Macedo A, Rahmouni N, Tissot C, Fernandez Arias J, Wang YTT, Trudel L, Hosseini SA, Matsudaira T, Chan T, Pascoal T, Gilfix B, Vitali P, Zimmer ER, Provost K, Soucy JP, Gauthier S, Zetterberg H, Jean-Claude BJ, Blennow K, Rosa-Neto P. Comparison of Plasma p-tau217 and [ 18F]FDG-PET for Identifying Alzheimer Disease in People With Early-Onset or Atypical Dementia. Neurology 2025; 104:e210211. [PMID: 39715476 PMCID: PMC11666273 DOI: 10.1212/wnl.0000000000210211] [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: 05/13/2024] [Accepted: 10/23/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES To compare the diagnostic performance of an immunoassay for plasma concentrations of phosphorylated tau (p-tau) 217 with visual assessments of fluorine-18 fluorodeoxyglucose [18F]FDG-PET in individuals who meet appropriate use criteria for Alzheimer dementia (AD) biomarker assessments. METHODS We performed a retrospective analysis of individuals with early-onset (age <65 years at onset) and/or atypical dementia (features other than memory at onset), who were evaluated at a tertiary care memory clinic. All participants underwent measurements of CSF biomarkers (Aβ42, p-tau181, and total tau levels), as well as [18F]FDG-PET scans, amyloid-PET scans, and plasma p-tau217 quantifications. To determine whether the [18F]FDG-PET images were compatible with AD, images were visually rated by 2 nuclear medicine experts. Using a contingency analysis, we evaluated the accuracy of [18F]FDG-PET scan interpretation and plasma p-tau217 for an AD biomarker profile in CSF and for amyloid-PET positivity. RESULTS A total of 81 individuals with early onset and/or atypical dementia were included in this study (mean age = 65 years; 48/81 female (59%). Both [18F]FDG-PET and plasma p-tau217 showed high levels of agreement with reference standard AD biomarkers ([18F]FDG-PET area under the curve [AUC]: 71%; plasma p-tau217 AUC: 81%). Although both biomarkers had similar specificity for AD [18F]FDG-PET: 70%, CI: 0.56-0.81; plasma p-tau217: 70%, CI: 0.56-0.81), plasma p-tau217 had higher sensitivity for AD (plasma p-tau217: 97%, CI: 0.85-0.99 vs [18F]FDG-PET: 73%, CI: 0.57-0.85) (p = 0.01). Overall accuracy was also higher for plasma p-tau217 (AUC = 84%, CI: 0.75-0.93 vs 72%, CI: 0.60-0.83 of [18F]FDG-PET) (p = 0.02). The same pattern of results was observed when using amyloid-PET as the reference standard. DISCUSSION Our study provides evidence that plasma p-tau217 has strong discriminative accuracy for AD among patients with early-onset and/or atypical dementia assessed in specialized settings. Future work should replicate these findings in secondary care settings.
Collapse
Affiliation(s)
- Kely Monica Quispialaya
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Experimental Medicine, McGill University
- Montreal Neurological Institute
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Antonio Aliaga
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Experimental Medicine, McGill University
- Graduate Program in Biological Sciences: Biochemistry (PPGBioq) and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Jaime Fernandez Arias
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Yi-Ting Tina Wang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Lydia Trudel
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Seyyed Ali Hosseini
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Takashi Matsudaira
- Department of Biofunctional Imaging, Hamamatsu University School of Medicine, Shizuoka, Japan
- Department of Neurology, NHO Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
| | - Tevy Chan
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Experimental Medicine, McGill University
- Montreal Neurological Institute
| | - Tharick Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Brian Gilfix
- Department of Specialized Medicine, McGill University, Montreal, Quebec, Canada
| | - Paolo Vitali
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Eduardo R Zimmer
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Graduate Program in Biological Sciences: Biochemistry (PPGBioq) and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Jean-Paul Soucy
- Montreal Neurological Institute
- Centre hospitalier de l'Université de Montreal
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, China; and
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Experimental Medicine, McGill University
- Montreal Neurological Institute
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Wang YT, Ashton NJ, Therriault J, Benedet AL, Macedo AC, Pola I, Aumont E, Di Molfetta G, Fernandez-Arias J, Tan K, Rahmouni N, Servaes SJG, Isaacson R, Chan T, Hosseini SA, Tissot C, Mathotaarachchi S, Stevenson J, Lussier FZ, Pascoal TA, Gauthier S, Blennow K, Zetterberg H, Rosa-Neto P. Identify biological Alzheimer's disease using a novel nucleic acid-linked protein immunoassay. Brain Commun 2025; 7:fcaf004. [PMID: 39845736 PMCID: PMC11753389 DOI: 10.1093/braincomms/fcaf004] [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: 06/13/2024] [Revised: 11/04/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Blood-based biomarkers have been revolutionizing the detection, diagnosis and screening of Alzheimer's disease. Specifically, phosphorylated-tau variants (p-tau181, p-tau217 and p-tau231) are promising biomarkers for identifying Alzheimer's disease pathology. Antibody-based assays such as single molecule arrays immunoassays are powerful tools to investigate pathological changes indicated by blood-based biomarkers and have been studied extensively in the Alzheimer's disease research field. A novel proteomic technology-NUcleic acid Linked Immuno-Sandwich Assay (NULISA)-was developed to improve the sensitivity of traditional proximity ligation assays and offer a comprehensive outlook for 120 protein biomarkers in neurodegenerative diseases. Due to the relative novelty of the NULISA technology in quantifying Alzheimer's disease biomarkers, validation through comparisons with more established methods is required. The main objective of the current study was to determine the capability of p-tau variants quantified using NULISA for identifying abnormal amyloid-β and tau pathology. We assessed 397 participants [mean (standard deviation) age, 64.8 (15.7) years; 244 females (61.5%) and 153 males (38.5%)] from the Translational Biomarkers in Aging and Dementia (TRIAD) cohort where participants had plasma measurements of p-tau181, p-tau217 and p-tau231 from NULISA and single molecule arrays immunoassays. Participants also underwent neuroimaging assessments, including structural MRI, amyloid-PET and tau-PET. Our findings suggest an excellent agreement between plasma p-tau variants quantified using NULISA and single molecule arrays immunoassays. Plasma p-tau217 measured with NULISA shows excellent discriminative accuracy for abnormal amyloid-PET (area under the receiver operating characteristic curve = 0.918, 95% confidence interval = 0.883 to 0.953, P < 0.0001) and tau-PET (area under the receiver operating characteristic curve = 0.939; 95% confidence interval = 0.909 to 0.969, P < 0.0001). It also presents the capability for differentiating tau-PET staging. Validation of the NULISA-measured plasma biomarkers adds to the current analytical methods for Alzheimer's disease diagnosis, screening and staging and could potentially expedite the development of a blood-based biomarker panel.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada H3A 0G4
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 39 Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, 4011 Stavanger, Norway
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London SE5 9RX, UK
- NIHR Maudsley Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London SE5 8AZ, UK
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 39 Mölndal, Sweden
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada H3A 0G4
| | - Ilaria Pola
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 39 Mölndal, Sweden
| | - Etienne Aumont
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada H3A 0G4
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 39 Mölndal, Sweden
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada H3A 0G4
| | - Kubra Tan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 39 Mölndal, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada H3A 0G4
| | - Stijn Johannes G Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
| | - Richard Isaacson
- Department of Neurology, Weill Cornell Medicine and New York-Presbyterian, New York, NY 10065, USA
- Department of Neurology, Florida Atlantic University, Charles E. Schmidt College of Medicine, Boca Raton, FL 33431, USA
| | - Tevy Chan
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
| | - Seyyed Ali Hosseini
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada H3A 0G4
| | - Cécile Tissot
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
| | - Firoza Z Lussier
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada H3A 0G4
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 39 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, 75013 Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei 101127, P. R.China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 39 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London W1CE 6BT, UK
- Hong Kong Center for 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 53792, USA
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Montreal, QC, Canada H4H 1R2
- Montreal Neurological Institute, Montreal, QC, Canada H3A 2B4
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada H3A 0G4
| |
Collapse
|
7
|
Quispialaya KM, Therriault J, Aliaga A, Tissot C, Servaes S, Rahmouni N, Karikari TK, Benedet AL, Ashton NJ, Macedo AC, Lussier FZ, Stevenson J, Wang Y, Arias JF, Hosseini A, Matsudaira T, Jean‐Claude B, Gilfix BM, Zimmer ER, Soucy J, Pascoal TA, Gauthier S, Zetterberg H, Blennow K, Rosa‐Neto P. Plasma phosphorylated tau181 outperforms [ 18F] fluorodeoxyglucose positron emission tomography in the identification of early Alzheimer disease. Eur J Neurol 2024; 31:e16255. [PMID: 39447157 PMCID: PMC11555153 DOI: 10.1111/ene.16255] [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: 10/20/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND AND PURPOSE This study was undertaken to compare the performance of plasma p-tau181 with that of [18F]fluorodeoxyglucose (FDG) positron emission tomography (PET) in the identification of early biological Alzheimer disease (AD). METHODS We included 533 cognitively impaired participants from the Alzheimer's Disease Neuroimaging Initiative. Participants underwent PET scans, biofluid collection, and cognitive tests. Receiver operating characteristic analyses were used to determine the diagnostic accuracy of plasma p-tau181 and [18F]FDG-PET using clinical diagnosis and core AD biomarkers ([18F]florbetapir-PET and cerebrospinal fluid [CSF] p-tau181) as reference standards. Differences in the diagnostic accuracy between plasma p-tau181 and [18F]FDG-PET were determined by bootstrap-based tests. Correlations of [18F]FDG-PET and plasma p-tau181 with CSF p-tau181, amyloid β (Aβ) PET, and cognitive performance were evaluated to compare associations between measurements. RESULTS We observed that both plasma p-tau181 and [18F]FDG-PET identified individuals with positive AD biomarkers in CSF or on Aβ-PET. In the MCI group, plasma p-tau181 outperformed [18F]FDG-PET in identifying AD measured by CSF (p = 0.0007) and by Aβ-PET (p = 0.001). We also observed that both plasma p-tau181 and [18F]FDG-PET metabolism were associated with core AD biomarkers. However, [18F]FDG-PET uptake was more closely associated with cognitive outcomes (Montreal Cognitive Assessment, Mini-Mental State Examination, Clinical Dementia Rating Sum of Boxes, and logical memory delayed recall, p < 0.001) than plasma p-tau181. CONCLUSIONS Overall, although both plasma p-tau181 and [18F]FDG-PET were associated with core AD biomarkers, plasma p-tau181 outperformed [18F]FDG-PET in identifying individuals with early AD pathophysiology. Taken together, our study suggests that plasma p-tau181 may aid in detecting individuals with underlying early AD.
Collapse
|
8
|
Therriault J, Janelidze S, Benedet AL, Ashton NJ, Arranz Martínez J, Gonzalez-Escalante A, Bellaver B, Alcolea D, Vrillon A, Karim H, Mielke MM, Hyung Hong C, Roh HW, Contador J, Puig Pijoan A, Algeciras-Schimnich A, Vemuri P, Graff-Radford J, Lowe VJ, Karikari TK, Jonaitis E, Brum W, Tissot C, Servaes S, Rahmouni N, Macedo AC, Stevenson J, Fernandez-Arias J, Wang YT, Woo MS, Friese MA, Jia WL, Dumurgier J, Hourregue C, Cognat E, Ferreira PL, Vitali P, Johnson S, Pascoal TA, Gauthier S, Lleó A, Paquet C, Petersen RC, Salmon D, Mattsson-Carlgren N, Palmqvist S, Stomrud E, Galasko D, Son SJ, Zetterberg H, Fortea J, Suárez-Calvet M, Jack CR, Blennow K, Hansson O, Rosa-Neto P. Diagnosis of Alzheimer's disease using plasma biomarkers adjusted to clinical probability. NATURE AGING 2024; 4:1529-1537. [PMID: 39533113 PMCID: PMC11564087 DOI: 10.1038/s43587-024-00731-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 09/25/2024] [Indexed: 11/16/2024]
Abstract
Recently approved anti-amyloid immunotherapies for Alzheimer's disease (AD) require evidence of amyloid-β pathology from positron emission tomography (PET) or cerebrospinal fluid (CSF) before initiating treatment. Blood-based biomarkers promise to reduce the need for PET or CSF testing; however, their interpretation at the individual level and the circumstances requiring confirmatory testing are poorly understood. Individual-level interpretation of diagnostic test results requires knowledge of disease prevalence in relation to clinical presentation (clinical pretest probability). Here, in a study of 6,896 individuals evaluated from 11 cohort studies from six countries, we determined the positive and negative predictive value of five plasma biomarkers for amyloid-β pathology in cognitively impaired individuals in relation to clinical pretest probability. We observed that p-tau217 could rule in amyloid-β pathology in individuals with probable AD dementia (positive predictive value above 95%). In mild cognitive impairment, p-tau217 interpretation depended on patient age. Negative p-tau217 results could rule out amyloid-β pathology in individuals with non-AD dementia syndromes (negative predictive value between 90% and 99%). Our findings provide a framework for the individual-level interpretation of plasma biomarkers, suggesting that p-tau217 combined with clinical phenotyping can identify patients where amyloid-β pathology can be ruled in or out without the need for PET or CSF confirmatory testing.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Javier Arranz Martínez
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Armand Gonzalez-Escalante
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Agathe Vrillon
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | - Helmet Karim
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Michelle M Mielke
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Chang Hyung Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyun Woong Roh
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - José Contador
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
| | - Albert Puig Pijoan
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
- Cognitive Decline and Movement Disorders Unit, Neurology Department, Hospital del Mar, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- ERA-Net on Cardiovascular Diseases Consortium, Barcelona, Spain
| | | | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Erin Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Wagner Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidad Federal do RioGrande do Sul, Porto Alegre, Brazil
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Marcel S Woo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wan Lu Jia
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Julien Dumurgier
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | - Claire Hourregue
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | - Emmanuel Cognat
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | | | - Paolo Vitali
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Sterling Johnson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Claire Paquet
- Institut National de la Santé et de la Recherche Médicale, Université de Paris Cité, Paris, France
- Centre de Neurologie Cognitive, Paris, France
| | - Ronald C Petersen
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - David Salmon
- San Diego and Shiley-Marcos Alzheimer's Disease Research Center, University of California, La Jolla, CA, USA
| | - 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
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmo, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmo, Sweden
| | - Douglas Galasko
- San Diego and Shiley-Marcos Alzheimer's Disease Research Center, University of California, La Jolla, CA, USA
| | - Sang Joon Son
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
- ERA-Net on Cardiovascular Diseases Consortium, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Madrid, Spain
| | - Marc Suárez-Calvet
- Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Medical Research Institute, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Madrid, Spain
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Centre intégré universitaire de santé et de services sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Frank G, Gualtieri P, Cianci R, Caldarelli M, Palma R, De Santis GL, Porfilio C, Nicoletti F, Bigioni G, Di Renzo L. Body Composition and Alzheimer's Disease: A Holistic Review. Int J Mol Sci 2024; 25:9573. [PMID: 39273520 PMCID: PMC11395597 DOI: 10.3390/ijms25179573] [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: 07/25/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Alzheimer's disease (AD) represents a significant global health challenge and affects approximately 50 million people worldwide. This overview of published reviews provides a comprehensive understanding of the intricate correlations between AD and body composition, focusing particularly on obesity. We used a systematic approach to collect and analyze relevant reviews on the topic of obesity and Alzheimer's disease. A comprehensive search of electronic databases, including PubMed, MEDLINE, and Google Scholar, was conducted. We searched keywords such as "Alzheimer's disease", "body composition", "lean mass", "bone mass", and "fat mass". We considered only reviews written within the past 5 years and in English. Fifty-six relevant reviews were identified that shed light on the multiple connections between AD and body composition. The review involves several aspects, including the impact of lean mass, bone mass, and endocrinological factors related to obesity, as well as inflammation, neuroinflammation, and molecular/genetic factors. The findings highlight the complex interplay of these elements in the development of AD, underscoring the need for holistic approaches to reduce the risk of AD and to explore innovative strategies for diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Giulia Frank
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Paola Gualtieri
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Roselisa Palma
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Gemma Lou De Santis
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Chiara Porfilio
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Francesco Nicoletti
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Giulia Bigioni
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
10
|
Fortea J, Pegueroles J, Alcolea D, Belbin O, Dols-Icardo O, Vaqué-Alcázar L, Videla L, Gispert JD, Suárez-Calvet M, Johnson SC, Sperling R, Bejanin A, Lleó A, Montal V. APOE4 homozygozity represents a distinct genetic form of Alzheimer's disease. Nat Med 2024; 30:1284-1291. [PMID: 38710950 DOI: 10.1038/s41591-024-02931-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/19/2024] [Indexed: 05/08/2024]
Abstract
This study aimed to evaluate the impact of APOE4 homozygosity on Alzheimer's disease (AD) by examining its clinical, pathological and biomarker changes to see whether APOE4 homozygotes constitute a distinct, genetically determined form of AD. Data from the National Alzheimer's Coordinating Center and five large cohorts with AD biomarkers were analyzed. The analysis included 3,297 individuals for the pathological study and 10,039 for the clinical study. Findings revealed that almost all APOE4 homozygotes exhibited AD pathology and had significantly higher levels of AD biomarkers from age 55 compared to APOE3 homozygotes. By age 65, nearly all had abnormal amyloid levels in cerebrospinal fluid, and 75% had positive amyloid scans, with the prevalence of these markers increasing with age, indicating near-full penetrance of AD biology in APOE4 homozygotes. The age of symptom onset was earlier in APOE4 homozygotes at 65.1, with a narrower 95% prediction interval than APOE3 homozygotes. The predictability of symptom onset and the sequence of biomarker changes in APOE4 homozygotes mirrored those in autosomal dominant AD and Down syndrome. However, in the dementia stage, there were no differences in amyloid or tau positron emission tomography across haplotypes, despite earlier clinical and biomarker changes. The study concludes that APOE4 homozygotes represent a genetic form of AD, suggesting the need for individualized prevention strategies, clinical trials and treatments.
Collapse
Affiliation(s)
- Juan Fortea
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain.
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain.
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain
| | - Olivia Belbin
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain
| | - Lídia Vaqué-Alcázar
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Neurosciences Programme, IMIM - Hospital del Mar Medical Research Institute, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina. Instituto de Salud carlos III, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Neurosciences Programme, IMIM - Hospital del Mar Medical Research Institute, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina. Instituto de Salud carlos III, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Reisa Sperling
- Brigham and Women's Hospital Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain
| | - Víctor Montal
- Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau - Biomedical Research Institute Sant Pau, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Barcelona, Spain.
- Barcelona Supercomputing Center, Barcelona, Spain.
| |
Collapse
|
11
|
Wang YT, Therriault J, Servaes S, Tissot C, Rahmouni N, Macedo AC, Fernandez-Arias J, Mathotaarachchi SS, Benedet AL, Stevenson J, Ashton NJ, Lussier FZ, Pascoal TA, Zetterberg H, Rajah MN, Blennow K, Gauthier S, Rosa-Neto P. Sex-specific modulation of amyloid-β on tau phosphorylation underlies faster tangle accumulation in females. Brain 2024; 147:1497-1510. [PMID: 37988283 PMCID: PMC10994548 DOI: 10.1093/brain/awad397] [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: 08/10/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/23/2023] Open
Abstract
Females are disproportionately affected by dementia due to Alzheimer's disease. Despite a similar amyloid-β (Aβ) load, a higher load of neurofibrillary tangles (NFTs) is seen in females than males. Previous literature has proposed that Aβ and phosphorylated-tau (p-tau) synergism accelerates tau tangle formation, yet the effect of biological sex in this process has been overlooked. In this observational study, we examined longitudinal neuroimaging data from the TRIAD and ADNI cohorts from Canada and USA, respectively. We assessed 457 participants across the clinical spectrum of Alzheimer's disease. All participants underwent baseline multimodal imaging assessment, including MRI and PET, with radioligands targeting Aβ plaques and tau tangles, respectively. CSF data were also collected. Follow-up imaging assessments were conducted at 1- and 2-year intervals for the TRIAD cohort and 1-, 2- and 4-year intervals for the ADNI cohort. The upstream pathological events contributing to faster tau progression in females were investigated-specifically, whether the contribution of Aβ and p-tau synergism to accelerated tau tangle formation is modulated by biological sex. We hypothesized that cortical Aβ predisposes tau phosphorylation and tangle accumulation in a sex-specific manner. Findings revealed that Aβ-positive females presented higher CSF p-tau181 concentrations compared with Aβ-positive males in both the TRIAD (P = 0.04, Cohen's d = 0.51) and ADNI (P = 0.027, Cohen's d = 0.41) cohorts. In addition, Aβ-positive females presented faster NFT accumulation compared with their male counterparts (TRIAD: P = 0.026, Cohen's d = 0.52; ADNI: P = 0.049, Cohen's d = 1.14). Finally, the triple interaction between female sex, Aβ and CSF p-tau181 was revealed as a significant predictor of accelerated tau accumulation at the 2-year follow-up visit (Braak I: P = 0.0067, t = 2.81; Braak III: P = 0.017, t = 2.45; Braak IV: P = 0.002, t = 3.17; Braak V: P = 0.006, t = 2.88; Braak VI: P = 0.0049, t = 2.93). Overall, we report sex-specific modulation of cortical Aβ in tau phosphorylation, consequently facilitating faster NFT progression in female individuals over time. This presents important clinical implications and suggests that early intervention that targets Aβ plaques and tau phosphorylation may be a promising therapeutic strategy in females to prevent the further accumulation and spread of tau aggregates.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Sulantha S Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, 4011 Stavanger, Norway
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London SE5 9RX, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London SE5 8AF, UK
| | - Firoza Z Lussier
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tharick A Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
- Hong Kong Center for 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 53792, USA
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 431 41 Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC H3A 0G4, Canada
- Montreal Neurological Institute, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
12
|
Wei B, Xu Y, Du Y, Zhou J, Zhong F, Wu C, Lou Y. Feasibility of Using Magnetic Resonance Spectroscopy Test Biomarkers to Diagnose Alzheimer's Disease: Systematic Evaluation and Meta-Analysis. ACTAS ESPANOLAS DE PSIQUIATRIA 2024; 52:161-171. [PMID: 38622011 PMCID: PMC11016455 DOI: 10.62641/aep.v52i2.1552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of dementia, resulting in impairments in memory, cognition, decision-making, and social skills. Thus, accurate preclinical diagnosis of Alzheimer's disease is paramount. The identification of biomarkers for Alzheimer's disease through magnetic resonance spectroscopy (MRS) represents a novel adjunctive diagnostic approach. OBJECTIVE This study conducted a meta-analysis of the diagnostic results of this technology to explore its feasibility and accuracy. METHODS PubMed, Cochrane Library, EMBASE, and Web of Science databases were searched without restrictions, with the search period extending up to July 31, 2022. The search strategy employed a combination of subject headings and keywords. All retrieved documents underwent screening by two researchers, who selected them for meta-analysis. The included literature was analyzed using Review Manager 5.4 software, with corresponding bias maps, forest plots, and summary receiver operating characteristic (SROC) curves generated and analyzed. RESULTS A total of 344 articles were retrieved initially, with 11 articles meeting the criteria for inclusion in the analysis. The analysis encompassed data from approximately 1766 patients. In the forest plot, both sensitivity (95% CI) and specificity (95% CI) approached 1. Examining the true positive rate, false positive rate, true negative rate, and false negative rate, all studies on the summary receiver operating characteristic (SROC) curve clustered in the upper left quadrant, suggesting a very high accuracy of biomarkers detected by MRS for diagnosing Alzheimer's disease. CONCLUSION The detection of biomarkers by MRS demonstrates feasibility and high accuracy in diagnosing AD. This technology holds promise for widespread adoption in the clinical diagnosis of AD in the future.
Collapse
Affiliation(s)
- Bo Wei
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Yiqin Xu
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Ye Du
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Jie Zhou
- Department of Radiology, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), 312000 Shaoxing, Zhejiang, China
| | - Fangfang Zhong
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Chenglong Wu
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| | - Yiping Lou
- Department of Neurology, Shaoxing People's Hospital, 312000 Shaoxing, Zhejiang, China
| |
Collapse
|
13
|
Therriault J, Ashton NJ, Pola I, Triana-Baltzer G, Brum WS, Di Molfetta G, Arslan B, Rahmouni N, Tissot C, Servaes S, Stevenson J, Macedo AC, Pascoal TA, Kolb HC, Jeromin A, Blennow K, Zetterberg H, Rosa-Neto P, Benedet AL. Comparison of two plasma p-tau217 assays to detect and monitor Alzheimer's pathology. EBioMedicine 2024; 102:105046. [PMID: 38471397 PMCID: PMC10943661 DOI: 10.1016/j.ebiom.2024.105046] [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: 09/14/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Blood-based biomarkers of Alzheimer's disease (AD) have become increasingly important as scalable tools for diagnosis and determining clinical trial eligibility. P-tau217 is the most promising due to its excellent sensitivity and specificity for AD-related pathological changes. METHODS We compared the performance of two commercially available plasma p-tau217 assays (ALZpath p-tau217 and Janssen p-tau217+) in 294 individuals cross-sectionally. Correlations with amyloid PET and tau PET were assessed, and Receiver Operating Characteristic (ROC) analyses evaluated both p-tau217 assays for identifying AD pathology. FINDINGS Both plasma p-tau217 assays were strongly associated with amyloid and tau PET. Furthermore, both plasma p-tau217 assays identified individuals with AD vs other neurodegenerative diseases (ALZpath AUC = 0.95; Janssen AUC = 0.96). Additionally, plasma p-tau217 concentrations rose with AD severity and their annual changes correlated with tau PET annual change. INTERPRETATION Both p-tau217 assays had excellent diagnostic performance for AD. Our study supports the future clinical use of commercially-available assays for p-tau217. FUNDING This research is supported by the Weston Brain Institute, Canadian Institutes of Health Research (CIHR), Canadian Consortium on Neurodegeneration in Aging, the Alzheimer's Association, Brain Canada Foundation, the Fonds de Recherche du Québec - Santé and the Colin J. Adair Charitable Foundation.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montréal, Québec H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Nicholas James Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal 6 431 41, Sweden; Wallenberg Centre for Molecular Medicine, University of Gothenburg, Gothenburg 6 431 41, Sweden; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London SE5 9RT, UK; NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London SE5 8AF, UK
| | - Ilaria Pola
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal 6 431 41, Sweden
| | | | - Wagner Scheeren Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal 6 431 41, Sweden
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal 6 431 41, Sweden
| | - Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal 6 431 41, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montréal, Québec H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Cecile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montréal, Québec H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montréal, Québec H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montréal, Québec H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montréal, Québec H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Tharick Ali Pascoal
- Department of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh 15213, USA
| | | | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal 6 431 41, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 6 431 41, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal 6 431 41, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 6 431 41, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London SE5 9RT, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong 1512, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montréal, Québec H4H 1R3, Canada; Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Andrea Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal 6 431 41, Sweden.
| |
Collapse
|
14
|
Therriault J, Schindler SE, Salvadó G, Pascoal TA, Benedet AL, Ashton NJ, Karikari TK, Apostolova L, Murray ME, Verberk I, Vogel JW, La Joie R, Gauthier S, Teunissen C, Rabinovici GD, Zetterberg H, Bateman RJ, Scheltens P, Blennow K, Sperling R, Hansson O, Jack CR, Rosa-Neto P. Biomarker-based staging of Alzheimer disease: rationale and clinical applications. Nat Rev Neurol 2024; 20:232-244. [PMID: 38429551 DOI: 10.1038/s41582-024-00942-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Disease staging, whereby the spatial extent and load of brain pathology are used to estimate the severity of Alzheimer disease (AD), is pivotal to the gold-standard neuropathological diagnosis of AD. Current in vivo diagnostic frameworks for AD are based on abnormal concentrations of amyloid-β and tau in the cerebrospinal fluid or on PET scans, and breakthroughs in molecular imaging have opened up the possibility of in vivo staging of AD. Focusing on the key principles of disease staging shared across several areas of medicine, this Review highlights the potential for in vivo staging of AD to transform our understanding of preclinical AD, refine enrolment criteria for trials of disease-modifying therapies and aid clinical decision-making in the era of anti-amyloid therapeutics. We provide a state-of-the-art review of recent biomarker-based AD staging systems and highlight their contributions to the understanding of the natural history of AD. Furthermore, we outline hypothetical frameworks to stage AD severity using more accessible fluid biomarkers. In addition, by applying amyloid PET-based staging to recently published anti-amyloid therapeutic trials, we highlight how biomarker-based disease staging frameworks could illustrate the numerous pathological changes that have already taken place in individuals with mildly symptomatic AD. Finally, we discuss challenges related to the validation and standardization of disease staging and provide a forward-looking perspective on potential clinical applications.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- NIHR Biomedical Research Centre, South London and Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Liana Apostolova
- Department of Neurology, University of Indiana School of Medicine, Indianapolis, IN, USA
| | | | - Inge Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Charlotte Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, 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
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Reisa Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Cataldo A, Criscuolo S, De Benedetto E, Masciullo A, Pesola M, Schiavoni R. A Novel Metric for Alzheimer's Disease Detection Based on Brain Complexity Analysis via Multiscale Fuzzy Entropy. Bioengineering (Basel) 2024; 11:324. [PMID: 38671746 PMCID: PMC11048692 DOI: 10.3390/bioengineering11040324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative brain disorder that affects cognitive functioning and memory. Current diagnostic tools, including neuroimaging techniques and cognitive questionnaires, present limitations such as invasiveness, high costs, and subjectivity. In recent years, interest has grown in using electroencephalography (EEG) for AD detection due to its non-invasiveness, low cost, and high temporal resolution. In this regard, this work introduces a novel metric for AD detection by using multiscale fuzzy entropy (MFE) to assess brain complexity, offering clinicians an objective, cost-effective diagnostic tool to aid early intervention and patient care. To this purpose, brain entropy patterns in different frequency bands for 35 healthy subjects (HS) and 35 AD patients were investigated. Then, based on the resulting MFE values, a specific detection algorithm, able to assess brain complexity abnormalities that are typical of AD, was developed and further validated on 24 EEG test recordings. This MFE-based method achieved an accuracy of 83% in differentiating between HS and AD, with a diagnostic odds ratio of 25, and a Matthews correlation coefficient of 0.67, indicating its viability for AD diagnosis. Furthermore, the algorithm showed potential for identifying anomalies in brain complexity when tested on a subject with mild cognitive impairment (MCI), warranting further investigation in future research.
Collapse
Affiliation(s)
- Andrea Cataldo
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (A.M.); (R.S.)
| | - Sabatina Criscuolo
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples Federico II, 80125 Naples, Italy; (S.C.); (E.D.B.); (M.P.)
| | - Egidio De Benedetto
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples Federico II, 80125 Naples, Italy; (S.C.); (E.D.B.); (M.P.)
| | - Antonio Masciullo
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (A.M.); (R.S.)
| | - Marisa Pesola
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples Federico II, 80125 Naples, Italy; (S.C.); (E.D.B.); (M.P.)
| | - Raissa Schiavoni
- Department of Engineering for Innovation, University of Salento, 73100 Lecce, Italy; (A.M.); (R.S.)
| |
Collapse
|
16
|
Ashton NJ, Brum WS, Di Molfetta G, Benedet AL, Arslan B, Jonaitis E, Langhough RE, Cody K, Wilson R, Carlsson CM, Vanmechelen E, Montoliu-Gaya L, Lantero-Rodriguez J, Rahmouni N, Tissot C, Stevenson J, Servaes S, Therriault J, Pascoal T, Lleó A, Alcolea D, Fortea J, Rosa-Neto P, Johnson S, Jeromin A, Blennow K, Zetterberg H. Diagnostic Accuracy of a Plasma Phosphorylated Tau 217 Immunoassay for Alzheimer Disease Pathology. JAMA Neurol 2024; 81:255-263. [PMID: 38252443 PMCID: PMC10804282 DOI: 10.1001/jamaneurol.2023.5319] [Citation(s) in RCA: 190] [Impact Index Per Article: 190.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/10/2023] [Indexed: 01/23/2024]
Abstract
Importance Phosphorylated tau (p-tau) is a specific blood biomarker for Alzheimer disease (AD) pathology, with p-tau217 considered to have the most utility. However, availability of p-tau217 tests for research and clinical use has been limited. Expanding access to this highly accurate AD biomarker is crucial for wider evaluation and implementation of AD blood tests. Objective To determine the utility of a novel and commercially available immunoassay for plasma p-tau217 to detect AD pathology and evaluate reference ranges for abnormal amyloid β (Aβ) and longitudinal change across 3 selected cohorts. Design, Setting, and Participants This cohort study examined data from 3 single-center observational cohorts: cross-sectional and longitudinal data from the Translational Biomarkers in Aging and Dementia (TRIAD) cohort (visits October 2017-August 2021) and Wisconsin Registry for Alzheimer's Prevention (WRAP) cohort (visits February 2007-November 2020) and cross-sectional data from the Sant Pau Initiative on Neurodegeneration (SPIN) cohort (baseline visits March 2009-November 2021). Participants included individuals with and without cognitive impairment grouped by amyloid and tau (AT) status using PET or CSF biomarkers. Data were analyzed from February to June 2023. Exposures Magnetic resonance imaging, Aβ positron emission tomography (PET), tau PET, cerebrospinal fluid (CSF) biomarkers (Aβ42/40 and p-tau immunoassays), and plasma p-tau217 (ALZpath pTau217 assay). Main Outcomes and Measures Accuracy of plasma p-tau217 in detecting abnormal amyloid and tau pathology, longitudinal p-tau217 change according to baseline pathology status. Results The study included 786 participants (mean [SD] age, 66.3 [9.7] years; 504 females [64.1%] and 282 males [35.9%]). High accuracy was observed in identifying elevated Aβ (area under the curve [AUC], 0.92-0.96; 95% CI, 0.89-0.99) and tau pathology (AUC, 0.93-0.97; 95% CI, 0.84-0.99) across all cohorts. These accuracies were comparable with CSF biomarkers in determining abnormal PET signal. The detection of abnormal Aβ pathology using a 3-range reference yielded reproducible results and reduced confirmatory testing by approximately 80%. Longitudinally, plasma p-tau217 values showed an annual increase only in Aβ-positive individuals, with the highest increase observed in those with tau positivity. Conclusions and Relevance This study found that a commercially available plasma p-tau217 immunoassay accurately identified biological AD, comparable with results using CSF biomarkers, with reproducible cut-offs across cohorts. It detected longitudinal changes, including at the preclinical stage.
Collapse
Affiliation(s)
- Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, 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
| | - Wagner S. Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, 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
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Andrea L. Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Erin 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
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Rebecca E. 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
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Karly Cody
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Rachael Wilson
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Cynthia M. Carlsson
- 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
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | | | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Cecile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Tharick Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau, Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau, Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau, Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Sterling 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
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin–Madison, Madison
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| |
Collapse
|
17
|
Gunter NB, Gebre RK, Graff-Radford J, Heckman MG, Jack CR, Lowe VJ, Knopman DS, Petersen RC, Ross OA, Vemuri P, Ramanan VK. Machine Learning Models of Polygenic Risk for Enhanced Prediction of Alzheimer Disease Endophenotypes. Neurol Genet 2024; 10:e200120. [PMID: 38250184 PMCID: PMC10798228 DOI: 10.1212/nxg.0000000000200120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/01/2023] [Indexed: 01/23/2024]
Abstract
Background and Objectives Alzheimer disease (AD) has a polygenic architecture, for which genome-wide association studies (GWAS) have helped elucidate sequence variants (SVs) influencing susceptibility. Polygenic risk score (PRS) approaches show promise for generating summary measures of inherited risk for clinical AD based on the effects of APOE and other GWAS hits. However, existing PRS approaches, based on traditional regression models, explain only modest variation in AD dementia risk and AD-related endophenotypes. We hypothesized that machine learning (ML) models of polygenic risk (ML-PRS) could outperform standard regression-based PRS methods and therefore have the potential for greater clinical utility. Methods We analyzed combined data from the Mayo Clinic Study of Aging (n = 1,791) and the Alzheimer's Disease Neuroimaging Initiative (n = 864). An AD PRS was computed for each participant using the top common SVs obtained from a large AD dementia GWAS. In parallel, ML models were trained using those SV genotypes, with amyloid PET burden as the primary outcome. Secondary outcomes included amyloid PET positivity and clinical diagnosis (cognitively unimpaired vs impaired). We compared performance between ML-PRS and standard PRS across 100 training sessions with different data splits. In each session, data were split into 80% training and 20% testing, and then five-fold cross-validation was used within the training set to ensure the best model was produced for testing. We also applied permutation importance techniques to assess which genetic factors contributed most to outcome prediction. Results ML-PRS models outperformed the AD PRS (r2 = 0.28 vs r2 = 0.24 in test set) in explaining variation in amyloid PET burden. Among ML approaches, methods accounting for nonlinear genetic influences were superior to linear methods. ML-PRS models were also more accurate when predicting amyloid PET positivity (area under the curve [AUC] = 0.80 vs AUC = 0.63) and the presence of cognitive impairment (AUC = 0.75 vs AUC = 0.54) compared with the standard PRS. Discussion We found that ML-PRS approaches improved upon standard PRS for prediction of AD endophenotypes, partly related to improved accounting for nonlinear effects of genetic susceptibility alleles. Further adaptations of the ML-PRS framework could help to close the gap of remaining unexplained heritability for AD and therefore facilitate more accurate presymptomatic and early-stage risk stratification for clinical decision-making.
Collapse
Affiliation(s)
- Nathaniel B Gunter
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Robel K Gebre
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Jonathan Graff-Radford
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Michael G Heckman
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Clifford R Jack
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Val J Lowe
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - David S Knopman
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Ronald C Petersen
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Owen A Ross
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Prashanthi Vemuri
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Vijay K Ramanan
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| |
Collapse
|
18
|
Woo MS, Tissot C, Lantero‐Rodriguez J, Snellman A, Therriault J, Rahmouni N, Macedo AC, Servaes S, Wang Y, Arias JF, Hosseini SA, Chamoun M, Lussier FZ, Benedet AL, Ashton NJ, Karikari TK, Triana‐Baltzer G, Kolb HC, Stevenson J, Mayer C, Kobayashi E, Massarweh G, Friese MA, Pascoal TA, Gauthier S, Zetterberg H, Blennow K, Rosa‐Neto P. Plasma pTau-217 and N-terminal tau (NTA) enhance sensitivity to identify tau PET positivity in amyloid-β positive individuals. Alzheimers Dement 2024; 20:1166-1174. [PMID: 37920945 PMCID: PMC10916953 DOI: 10.1002/alz.13528] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION We set out to identify tau PET-positive (A+T+) individuals among amyloid-beta (Aβ) positive participants using plasma biomarkers. METHODS In this cross-sectional study we assessed 234 participants across the AD continuum who were evaluated by amyloid PET with [18 F]AZD4694 and tau-PET with [18 F]MK6240 and measured plasma levels of total tau, pTau-181, pTau-217, pTau-231, and N-terminal tau (NTA-tau). We evaluated the performances of plasma biomarkers to predict tau positivity in Aβ+ individuals. RESULTS Highest associations with tau positivity in Aβ+ individuals were found for plasma pTau-217 (AUC [CI95% ] = 0.89 [0.82, 0.96]) and NTA-tau (AUC [CI95% ] = 0.88 [0.91, 0.95]). Combining pTau-217 and NTA-tau resulted in the strongest agreement (Cohen's Kappa = 0.74, CI95% = 0.57/0.90, sensitivity = 92%, specificity = 81%) with PET for classifying tau positivity. DISCUSSION The potential for identifying tau accumulation in later Braak stages will be useful for patient stratification and prognostication in treatment trials and in clinical practice. HIGHLIGHTS We found that in a cohort without pre-selection pTau-181, pTau-217, and NTA-tau showed the highest association with tau PET positivity. We found that in Aβ+ individuals pTau-217 and NTA-tau showed the highest association with tau PET positivity. Combining pTau-217 and NTA-tau resulted in the strongest agreement with the tau PET-based classification.
Collapse
|
19
|
Therriault J, Woo MS, Salvadó G, Gobom J, Karikari TK, Janelidze S, Servaes S, Rahmouni N, Tissot C, Ashton NJ, Benedet AL, Montoliu-Gaya L, Macedo AC, Lussier FZ, Stevenson J, Vitali P, Friese MA, Massarweh G, Soucy JP, Pascoal TA, Stomrud E, Palmqvist S, Mattsson-Carlgren N, Gauthier S, Zetterberg H, Hansson O, Blennow K, Rosa-Neto P. Comparison of immunoassay- with mass spectrometry-derived p-tau quantification for the detection of Alzheimer's disease pathology. Mol Neurodegener 2024; 19:2. [PMID: 38185677 PMCID: PMC10773025 DOI: 10.1186/s13024-023-00689-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Antibody-based immunoassays have enabled quantification of very low concentrations of phosphorylated tau (p-tau) protein forms in cerebrospinal fluid (CSF), aiding in the diagnosis of AD. Mass spectrometry enables absolute quantification of multiple p-tau variants within a single run. The goal of this study was to compare the performance of mass spectrometry assessments of p-tau181, p-tau217 and p-tau231 with established immunoassay techniques. METHODS We measured p-tau181, p-tau217 and p-tau231 concentrations in CSF from 173 participants from the TRIAD cohort and 394 participants from the BioFINDER-2 cohort using both mass spectrometry and immunoassay methods. All subjects were clinically evaluated by dementia specialists and had amyloid-PET and tau-PET assessments. Bland-Altman analyses evaluated the agreement between immunoassay and mass spectrometry p-tau181, p-tau217 and p-tau231. P-tau associations with amyloid-PET and tau-PET uptake were also compared. Receiver Operating Characteristic (ROC) analyses compared the performance of mass spectrometry and immunoassays p-tau concentrations to identify amyloid-PET positivity. RESULTS Mass spectrometry and immunoassays of p-tau217 were highly comparable in terms of diagnostic performance, between-group effect sizes and associations with PET biomarkers. In contrast, p-tau181 and p-tau231 concentrations measured using antibody-free mass spectrometry had lower performance compared with immunoassays. CONCLUSIONS Our results suggest that while similar overall, immunoassay-based p-tau biomarkers are slightly superior to antibody-free mass spectrometry-based p-tau biomarkers. Future work is needed to determine whether the potential to evaluate multiple biomarkers within a single run offsets the slightly lower performance of antibody-free mass spectrometry-based p-tau quantification.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Marcel S Woo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology, Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Gemma Salvadó
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, 15213, USA
| | - Shorena Janelidze
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Wallenberg Centre for Molecular Medicine, University of Gothenburg, Gothenburg, S-413 45, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, SE5 9RT, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, SE5 8AF, UK
| | - Andréa Lessa Benedet
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, 15213, USA
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
| | - Paolo Vitali
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Manuel A Friese
- Department of Neurology, Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Gassan Massarweh
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, 15213, USA
| | - Erik Stomrud
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 6BG, UK
- UK Dementia Research Institute at UCL, London, WC1N 6BG, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Oskar Hansson
- Department of Clinical Sciences Malmö, Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, S-431 80, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, S-431 80, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre Intégré Universitaire de Santé Et de Services Sociaux (CIUSSS) de l'Ouest-de-L'Île-de-Montréal, 6875 La Salle Blvd - FBC Room 3149, Montréal, Québec, H4H 1R3, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
20
|
Lantero-Rodriguez J, Montoliu-Gaya L, Benedet AL, Vrillon A, Dumurgier J, Cognat E, Brum WS, Rahmouni N, Stevenson J, Servaes S, Therriault J, Becker B, Brinkmalm G, Snellman A, Huber H, Kvartsberg H, Ashton NJ, Zetterberg H, Paquet C, Rosa-Neto P, Blennow K. CSF p-tau205: a biomarker of tau pathology in Alzheimer's disease. Acta Neuropathol 2024; 147:12. [PMID: 38184490 PMCID: PMC10771353 DOI: 10.1007/s00401-023-02659-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 01/08/2024]
Abstract
Post-mortem staging of Alzheimer's disease (AD) neurofibrillary pathology is commonly performed by immunohistochemistry using AT8 antibody for phosphorylated tau (p-tau) at positions 202/205. Thus, quantification of p-tau205 and p-tau202 in cerebrospinal fluid (CSF) should be more reflective of neurofibrillary tangles in AD than other p-tau epitopes. We developed two novel Simoa immunoassays for CSF p-tau205 and p-tau202 and measured these phosphorylations in three independent cohorts encompassing the AD continuum, non-AD cases and cognitively unimpaired participants: a discovery cohort (n = 47), an unselected clinical cohort (n = 212) and a research cohort well-characterized by fluid and imaging biomarkers (n = 262). CSF p-tau205 increased progressively across the AD continuum, while CSF p-tau202 was increased only in AD and amyloid (Aβ) and tau pathology positive (A+T+) cases (P < 0.01). In A+ cases, CSF p-tau205 and p-tau202 showed stronger associations with tau-PET (rSp205 = 0.67, rSp202 = 0.45) than Aβ-PET (rSp205 = 0.40, rSp202 = 0.09). CSF p-tau205 increased gradually across tau-PET Braak stages (P < 0.01), whereas p-tau202 only increased in Braak V-VI (P < 0.0001). Both showed stronger regional associations with tau-PET than with Aβ-PET, and CSF p-tau205 was significantly associated with Braak V-VI tau-PET regions. When assessing the contribution of Aβ and tau pathologies (indexed by PET) to CSF p-tau205 and p-tau202 variance, tau pathology was found to be the most prominent contributor in both cases (CSF p-tau205: R2 = 69.7%; CSF p-tau202: R2 = 85.6%) Both biomarkers associated with brain atrophy measurements globally (rSp205 = - 0.36, rSp202 = - 0.33) and regionally, and correlated with cognition (rSp205 = - 0.38/- 0.40, rSp202 = - 0.20/- 0.29). In conclusion, we report the first high-throughput CSF p-tau205 immunoassay for the in vivo quantification of tau pathology in AD, and a potentially cost-effective alternative to tau-PET in clinical settings and clinical trials.
Collapse
Affiliation(s)
- Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Agathe Vrillon
- Cognitive Neurology Center, Université de Paris Cité, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Julien Dumurgier
- Cognitive Neurology Center, Université de Paris Cité, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Emmanuel Cognat
- Cognitive Neurology Center, Université de Paris Cité, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - 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
| | - Nesrine Rahmouni
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Jenna Stevenson
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Stijn Servaes
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Bruno Becker
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Hanna Huber
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Hlin Kvartsberg
- 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
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - 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, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Claire Paquet
- Cognitive Neurology Center, Université de Paris Cité, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Pedro Rosa-Neto
- Montreal Neurological Institute, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - 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
| |
Collapse
|
21
|
LoBue C, Stopschinski BE, Calveras NS, Douglas PM, Huebinger R, Cullum CM, Hart J, Gonzales MM. Blood Markers in Relation to a History of Traumatic Brain Injury Across Stages of Cognitive Impairment in a Diverse Cohort. J Alzheimers Dis 2024; 97:345-358. [PMID: 38143366 PMCID: PMC10947497 DOI: 10.3233/jad-231027] [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: 12/26/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) has been linked to multiple pathophysiological processes that could increase risk for Alzheimer's disease and related dementias (ADRD). However, the impact of prior TBI on blood biomarkers for ADRD remains unknown. OBJECTIVE Using cross-sectional data, we assessed whether a history of TBI influences serum biomarkers in a diverse cohort (approximately 50% Hispanic) with normal cognition, mild cognitive impairment, or dementia. METHODS Levels of glial fibrillary acidic protein (GFAP), neurofilament light (NFL), total tau (T-tau), and ubiquitin carboxy-terminal hydrolase-L1 (UCHL1) were measured for participants across the cognitive spectrum. Participants were categorized based on presence and absence of a history of TBI with loss of consciousness, and study samples were derived through case-control matching. Multivariable general linear models compared concentrations of biomarkers in relation to a history of TBI and smoothing splines modelled biomarkers non-linearly in the cognitively impaired groups as a function of time since symptom onset. RESULTS Each biomarker was higher across stages of cognitive impairment, characterized by clinical diagnosis and Mini-Mental State Examination performance, but these associations were not influenced by a history of TBI. However, modelling biomarkers in relation to duration of cognitive symptoms for ADRD showed differences by history of TBI, with only GFAP and UCHL1 being elevated. CONCLUSIONS Serum GFAP, NFL, T-tau, and UCHL1 were higher across stages of cognitive impairment in this diverse clinical cohort, regardless of TBI history, though longitudinal investigation of the timing, order, and trajectory of the biomarkers in relation to prior TBI is warranted.
Collapse
Affiliation(s)
- Christian LoBue
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas,TX
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Barbara E. Stopschinski
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX
| | - Nil Saez Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX
| | - Peter M. Douglas
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Ryan Huebinger
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - C. Munro Cullum
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas,TX
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
| | - John Hart
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas,TX
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Mitzi M. Gonzales
- Department of Neurology, Cedars Sinai Medical Center, Los Angeles, CA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX
| |
Collapse
|
22
|
Zhuang X, Cordes D, Bender AR, Nandy R, Oh EC, Kinney J, Caldwell JZ, Cummings J, Miller J. Classifying Alzheimer's Disease Neuropathology Using Clinical and MRI Measurements. J Alzheimers Dis 2024; 100:843-862. [PMID: 38943387 PMCID: PMC11307063 DOI: 10.3233/jad-231321] [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] [Accepted: 05/21/2024] [Indexed: 07/01/2024]
Abstract
Background Computer-aided machine learning models are being actively developed with clinically available biomarkers to diagnose Alzheimer's disease (AD) in living persons. Despite considerable work with cross-sectional in vivo data, many models lack validation against postmortem AD neuropathological data. Objective Train machine learning models to classify the presence or absence of autopsy-confirmed severe AD neuropathology using clinically available features. Methods AD neuropathological status are assessed at postmortem for participants from the National Alzheimer's Coordinating Center (NACC). Clinically available features are utilized, including demographics, Apolipoprotein E(APOE) genotype, and cortical thicknesses derived from ante-mortem MRI scans encompassing AD meta regions of interest (meta-ROI). Both logistic regression and random forest models are trained to identify linearly and nonlinearly separable features between participants with the presence (N = 91, age-at-MRI = 73.6±9.24, 38 women) or absence (N = 53, age-at-MRI = 68.93±19.69, 24 women) of severe AD neuropathology. The trained models are further validated in an external data set against in vivo amyloid biomarkers derived from PET imaging (amyloid-positive: N = 71, age-at-MRI = 74.17±6.37, 26 women; amyloid-negative: N = 73, age-at-MRI = 71.59±6.80, 41 women). Results Our models achieve a cross-validation accuracy of 84.03% in classifying the presence or absence of severe AD neuropathology, and an external-validation accuracy of 70.14% in classifying in vivo amyloid positivity status. Conclusions Our models show that clinically accessible features, including APOE genotype and cortical thinning encompassing AD meta-ROIs, are able to classify both postmortem confirmed AD neuropathological status and in vivo amyloid status with reasonable accuracies. These results suggest the potential utility of AD meta-ROIs in determining AD neuropathological status in living persons.
Collapse
Affiliation(s)
- Xiaowei Zhuang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- Interdisciplinary Neuroscience PhD Program, University of Nevada, Las Vegas, NV, USA
- Laboratory of Neurogenetics and Precision Medicine, University of Nevada, Las Vegas, NV, USA
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- University of Colorado Boulder, Boulder, CO, USA
| | - Andrew R. Bender
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Rajesh Nandy
- Department of Biostatistics and Epidemiology, School of Public Health, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Edwin C. Oh
- Interdisciplinary Neuroscience PhD Program, University of Nevada, Las Vegas, NV, USA
- Laboratory of Neurogenetics and Precision Medicine, University of Nevada, Las Vegas, NV, USA
- Department of Internal Medicine, School of Medicine, University of Nevada, Las Vegas, NV, USA
| | - Jefferson Kinney
- Interdisciplinary Neuroscience PhD Program, University of Nevada, Las Vegas, NV, USA
- Department of Brain Health, Chambers-Grundy Center for Transformative Neuroscience, School of Integrated Health Sciences, University of Nevada, Las Vegas, NV, USA
| | | | - Jeffrey Cummings
- Department of Brain Health, Chambers-Grundy Center for Transformative Neuroscience, School of Integrated Health Sciences, University of Nevada, Las Vegas, NV, USA
| | - Justin Miller
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| |
Collapse
|
23
|
Ramanan VK, Armstrong MJ, Choudhury P, Coerver KA, Hamilton RH, Klein BC, Wolk DA, Wessels SR, Jones LK. Antiamyloid Monoclonal Antibody Therapy for Alzheimer Disease: Emerging Issues in Neurology. Neurology 2023; 101:842-852. [PMID: 37495380 PMCID: PMC10663011 DOI: 10.1212/wnl.0000000000207757] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/30/2023] [Indexed: 07/28/2023] Open
Abstract
With recent data demonstrating that lecanemab treatment can slow cognitive and functional decline in early symptomatic Alzheimer disease (AD), it is widely anticipated that this drug and potentially other monoclonal antibody infusions targeting β-amyloid protein will imminently be realistic options for some patients with AD. Given that these new antiamyloid monoclonal antibodies (mAbs) are associated with nontrivial risks and burdens of treatment that are radically different from current mainstays of AD management, effectively and equitably translating their use to real-world clinical care will require systematic and practice-specific modifications to existing workflows and infrastructure. In this Emerging Issues in Neurology article, we provide practical guidance for a wide audience of neurology clinicians on logistic adaptations and decision making around emerging antiamyloid mAbs. Specifically, we briefly summarize the rationale and available evidence supporting antiamyloid mAb use in AD to facilitate appropriate communication with patients and care partners on potential benefits. We also discuss pragmatic approaches to optimizing patient selection and treatment monitoring, with a particular focus on the value of incorporating shared decision making and multidisciplinary collaboration. In addition, we review some of the recognized limitations of current knowledge and highlight areas of future evolution to guide the development of sustainable and flexible models for treatment and follow-up. As the field enters a new era with disease-modifying treatment options for AD, it will be critical for neurology practices to prepare and continually innovate to ensure optimal outcomes for patients.
Collapse
Affiliation(s)
- Vijay K Ramanan
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| | - Melissa J Armstrong
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| | - Parichita Choudhury
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| | - Katherine A Coerver
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| | - Roy H Hamilton
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| | - Brad C Klein
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| | - David A Wolk
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| | - Scott R Wessels
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| | - Lyell K Jones
- From the Department of Neurology (V.K.R., L.K.J.), Mayo Clinic, Rochester, MN; Department of Neurology (M.J.A.), University of Florida College of Medicine; Norman Fixel Institute for Neurologic Diseases (M.J.A.), University of Florida, Gainesville; Cleo Roberts Center (P.C.), Banner Sun Health Research Institute, Sun City, AZ; Rocky Mountain Neurology (K.C.), Lone Tree, CO; Department of Neurology (R.H.H., D.A.W.), Department of Physical Medicine and Rehabilitation (R.H.H.), and Department of Psychiatry (R.H.H.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Abington Neurological Associates (B.C.K.), Ltd., Abington, PA; and American Academy of Neurology (S.R.W.), Minneapolis, MN
| |
Collapse
|
24
|
Schaffer Aguzzoli C, Ferreira PCL, Povala G, Ferrari-Souza JP, Bellaver B, Soares Katz C, Zalzale H, Lussier FZ, Rohden F, Abbas S, Leffa DT, Scop Medeiros M, Therriault J, Benedet AL, Tissot C, Servaes S, Rahmouni N, Cassa Macedo A, Bezgin G, Kang MS, Stevenson J, Pallen V, Cohen A, Lopez OL, Tudorascu DL, Klunk WE, Villemagne VL, Soucy JP, Zimmer ER, Schilling LP, Karikari TK, Ashton NJ, Zetterberg H, Blennow K, Gauthier S, Valcour V, Miller BL, Rosa-Neto P, Pascoal TA. Neuropsychiatric Symptoms and Microglial Activation in Patients with Alzheimer Disease. JAMA Netw Open 2023; 6:e2345175. [PMID: 38010651 PMCID: PMC10682836 DOI: 10.1001/jamanetworkopen.2023.45175] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/15/2023] [Indexed: 11/29/2023] Open
Abstract
Importance Neuropsychiatric symptoms are commonly encountered and are highly debilitating in patients with Alzheimer disease. Understanding their underpinnings has implications for identifying biomarkers and treatment for these symptoms. Objective To evaluate whether glial markers are associated with neuropsychiatric symptoms in individuals across the Alzheimer disease continuum. Design, Setting, and Participants This cross-sectional study was conducted from January to June 2023, leveraging data from the Translational Biomarkers in Aging and Dementia cohort at McGill University, Canada. Recruitment was based on referrals of individuals from the community or from outpatient clinics. Exclusion criteria included active substance abuse, major surgery, recent head trauma, safety contraindications for positron emission tomography (PET) or magnetic resonance imaging, being currently enrolled in other studies, and having inadequately treated systemic conditions. Main Outcomes and Measures All individuals underwent assessment for neuropsychiatric symptoms (Neuropsychiatry Inventory Questionnaire [NPI-Q]), and imaging for microglial activation ([11C]PBR28 PET), amyloid-β ([18F]AZD4694 PET), and tau tangles ([18F]MK6240 PET). Results Of the 109 participants, 72 (66%) were women and 37 (34%) were men; the median age was 71.8 years (range, 38.0-86.5 years). Overall, 70 had no cognitive impairment and 39 had cognitive impairment (25 mild; 14 Alzheimer disease dementia). Amyloid-β PET positivity was present in 21 cognitively unimpaired individuals (30%) and in 31 cognitively impaired individuals (79%). The NPI-Q severity score was associated with microglial activation in the frontal, temporal, and parietal cortices (β = 7.37; 95% CI, 1.34-13.41; P = .01). A leave-one-out approach revealed that irritability was the NPI-Q domain most closely associated with the presence of brain microglial activation (β = 6.86; 95% CI, 1.77-11.95; P = .008). Furthermore, we found that microglia-associated irritability was associated with study partner burden measured by NPI-Q distress score (β = 5.72; 95% CI, 0.33-11.10; P = .03). Conclusions and Relevance In this cross-sectional study of 109 individuals across the AD continuum, microglial activation was associated with and a potential biomarker of neuropsychiatric symptoms in Alzheimer disease. Moreover, our findings suggest that the combination of amyloid-β- and microglia-targeted therapies could have an impact on relieving these symptoms.
Collapse
Affiliation(s)
- Cristiano Schaffer Aguzzoli
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Global Brain Health Institute, University of California, San Francisco
| | - Pâmela C. L. Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Carolina Soares Katz
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Hussein Zalzale
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Firoza Z. Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Francieli Rohden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sarah Abbas
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Douglas T. Leffa
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina Scop Medeiros
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Andréa L. Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Arthur Cassa Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Ann Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dana L. Tudorascu
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William E. Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jean Paul Soucy
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmacology, Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lucas P. Schilling
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Neurology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Thomas K. Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, 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, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Victor Valcour
- Global Brain Health Institute, University of California, San Francisco
- Department of Neurology, University of California, San Francisco
| | - Bruce L. Miller
- Global Brain Health Institute, University of California, San Francisco
- Department of Neurology, University of California, San Francisco
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux de l’Ouest-de-l’Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
25
|
Balu D, Valencia-Olvera AC, Islam Z, Mielczarek C, Hansen A, Perez Ramos TM, York J, LaDu MJ, Tai LM. APOE genotype and sex modulate Alzheimer's disease pathology in aged EFAD transgenic mice. Front Aging Neurosci 2023; 15:1279343. [PMID: 38020764 PMCID: PMC10644540 DOI: 10.3389/fnagi.2023.1279343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Increasing evidence supports that age, APOE and sex interact to modulate Alzheimer's disease (AD) risk, however the underlying pathways are unclear. One way that AD risk factors may modulate cognition is by impacting amyloid beta (Aβ) accumulation as plaques, and/or neuroinflammation Therefore, the goal of the present study was to evaluate the extent to which age, APOE and sex modulate Aβ pathology, neuroinflammation and behavior in vivo. To achieve this goal, we utilized the EFAD mice, which express human APOE3 or APOE4 and have five familial AD mutations (FAD) that result in Aβ42 overproduction. We assessed Aβ levels, reactive glia and Morris water maze performance in 6-, 10-, 14-, and 18-month-old EFAD mice. Female APOE4 mice had the highest Aβ deposition, fibrillar amyloid deposits and neuroinflammation as well as earlier behavior deficits. Interestingly, we found that female APOE3 mice and male APOE4 mice had similar levels of pathology. Collectively our data support that the combination of APOE4 and female sex is the most detrimental combination for AD, and that at older ages, female sex may be equivalent to APOE4 genotype.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zarak Islam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Clare Mielczarek
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Allison Hansen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Peoria, IL, United States
| | - Tamara M. Perez Ramos
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- School of Medicine, St. George’s University, St. George’s, Grenada
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
26
|
Nabizadeh F. sTREM2 is associated with attenuated tau aggregate accumulation in the presence of amyloid-β pathology. Brain Commun 2023; 5:fcad286. [PMID: 37942087 PMCID: PMC10629471 DOI: 10.1093/braincomms/fcad286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/16/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023] Open
Abstract
Triggering Receptor Expressed on Myeloid Cell 2 (TREM2) plays a crucial role in the transition of microglia from a state of homeostasis to a state associated with the disease. Mutations in TREM2 are strongly linked with a higher risk of developing neurodegenerative diseases, including Alzheimer's disease. There have been contradictory findings regarding the potential detrimental or protective effects of microglial activation and TREM2-related microglial responses in Alzheimer's disease. Although previous studies reported increased CSF soluble TREM2 (sTREM2) in different clinical stages of Alzheimer's disease, the exact association between Alzheimer's disease hallmarks such as amyloid-beta and tau pathology remains unclear. In the present study, I aimed to investigate the association between TREM2-related microglial responses and tau accumulation in the presence and absence of amyloid-beta pathology in order to give a better view of the role of microglial activation in Alzheimer's disease development. Imaging data of 178 non-demented participants including 107 amyloid-beta-negative participants, 71 amyloid-beta-positive were recruited from Alzheimer's disease Neuroimaging Initiative. The CSF sTREM2 was used as an in vivo indicator of microglial responses associated with TREM2. Furthermore, I used longitudinal tau-PET and resting-state functional MRI connectomes in order to investigate the association of TREM2-related microglial activation and tau spreading through functional connections. A higher level of sTREM2 was associated with slower tau aggregate accumulation in non-demented amyloid-beta-positive. Furthermore, measuring the tau spreading through inter-connected regions using functional MRI connectomes confirms that the TREM2-related microglial activity might be a protective factor against tau pathology in brain tissue. These findings demonstrate that in individuals with initial amyloid-beta abnormalities, TREM2-related microglial activation is linked to reduced regional accumulation of tau aggregates and also, spreading across inter-connected brain regions, as evaluated through functional MRI connectomes during the early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| |
Collapse
|
27
|
Belasso CJ, Cai Z, Bezgin G, Pascoal T, Stevenson J, Rahmouni N, Tissot C, Lussier F, Rosa-Neto P, Soucy JP, Rivaz H, Benali H. Bayesian workflow for the investigation of hierarchical classification models from tau-PET and structural MRI data across the Alzheimer's disease spectrum. Front Aging Neurosci 2023; 15:1225816. [PMID: 37920382 PMCID: PMC10619155 DOI: 10.3389/fnagi.2023.1225816] [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: 05/19/2023] [Accepted: 09/26/2023] [Indexed: 11/04/2023] Open
Abstract
Background Alzheimer's disease (AD) diagnosis in its early stages remains difficult with current diagnostic approaches. Though tau neurofibrillary tangles (NFTs) generally follow the stereotypical pattern described by the Braak staging scheme, the network degeneration hypothesis (NDH) has suggested that NFTs spread selectively along functional networks of the brain. To evaluate this, we implemented a Bayesian workflow to develop hierarchical multinomial logistic regression models with increasing levels of complexity of the brain from tau-PET and structural MRI data to investigate whether it is beneficial to incorporate network-level information into an ROI-based predictive model for the presence/absence of AD. Methods This study included data from the Translational Biomarkers in Aging and Dementia (TRIAD) longitudinal cohort from McGill University's Research Centre for Studies in Aging (MCSA). Baseline and 1 year follow-up structural MRI and [18F]MK-6240 tau-PET scans were acquired for 72 cognitive normal (CN), 23 mild cognitive impairment (MCI), and 18 Alzheimer's disease dementia subjects. We constructed the four following hierarchical Bayesian models in order of increasing complexity: (Model 1) a complete-pooling model with observations, (Model 2) a partial-pooling model with observations clustered within ROIs, (Model 3) a partial-pooling model with observations clustered within functional networks, and (Model 4) a partial-pooling model with observations clustered within ROIs that are also clustered within functional brain networks. We then investigated which of the models had better predictive performance given tau-PET or structural MRI data as an input, in the form of a relative annualized rate of change. Results The Bayesian leave-one-out cross-validation (LOO-CV) estimate of the expected log pointwise predictive density (ELPD) results indicated that models 3 and 4 were substantially better than other models for both tau-PET and structural MRI inputs. For tau-PET data, model 3 was slightly better than 4 with an absolute difference in ELPD of 3.10 ± 1.30. For structural MRI data, model 4 was considerably better than other models with an absolute difference in ELPD of 29.83 ± 7.55 relative to model 3, the second-best model. Conclusion Our results suggest that representing the data generating process in terms of a hierarchical model that encompasses both ROI-level and network-level heterogeneity leads to better predictive ability for both tau-PET and structural MRI inputs over all other model iterations.
Collapse
Affiliation(s)
- Clyde J. Belasso
- Department of Electrical and Computer Engineering, Concordia University, Montréal, QC, Canada
- PERFORM Centre, Concordia University, Montréal, QC, Canada
| | - Zhengchen Cai
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Gleb Bezgin
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Tharick Pascoal
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Firoza Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
- McConnell Brain Imaging Centre (BIC), Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Jean-Paul Soucy
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Hassan Rivaz
- Department of Electrical and Computer Engineering, Concordia University, Montréal, QC, Canada
- PERFORM Centre, Concordia University, Montréal, QC, Canada
| | - Habib Benali
- Department of Electrical and Computer Engineering, Concordia University, Montréal, QC, Canada
- PERFORM Centre, Concordia University, Montréal, QC, Canada
| |
Collapse
|
28
|
Ferrari-Souza JP, Bellaver B, Ferreira PCL, Benedet AL, Povala G, Lussier FZ, Leffa DT, Therriault J, Tissot C, Soares C, Wang YT, Chamoun M, Servaes S, Macedo AC, Vermeiren M, Bezgin G, Kang MS, Stevenson J, Rahmouni N, Pallen V, Poltronetti NM, Cohen A, Lopez OL, Klunk WE, Soucy JP, Gauthier S, Souza DO, Triana-Baltzer G, Saad ZS, Kolb HC, Karikari TK, Villemagne VL, Tudorascu DL, Ashton NJ, Zetterberg H, Blennow K, Zimmer ER, Rosa-Neto P, Pascoal TA. APOEε4 potentiates amyloid β effects on longitudinal tau pathology. NATURE AGING 2023; 3:1210-1218. [PMID: 37749258 PMCID: PMC10592050 DOI: 10.1038/s43587-023-00490-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 08/16/2023] [Indexed: 09/27/2023]
Abstract
The mechanisms by which the apolipoprotein E ε4 (APOEε4) allele influences the pathophysiological progression of Alzheimer's disease (AD) are poorly understood. Here we tested the association of APOEε4 carriership and amyloid-β (Aβ) burden with longitudinal tau pathology. We longitudinally assessed 94 individuals across the aging and AD spectrum who underwent clinical assessments, APOE genotyping, magnetic resonance imaging, positron emission tomography (PET) for Aβ ([18F]AZD4694) and tau ([18F]MK-6240) at baseline, as well as a 2-year follow-up tau-PET scan. We found that APOEε4 carriership potentiates Aβ effects on longitudinal tau accumulation over 2 years. The APOEε4-potentiated Aβ effects on tau-PET burden were mediated by longitudinal plasma phosphorylated tau at threonine 217 (p-tau217+) increase. This longitudinal tau accumulation as measured by PET was accompanied by brain atrophy and clinical decline. Our results suggest that the APOEε4 allele plays a key role in Aβ downstream effects on the aggregation of phosphorylated tau in the living human brain.
Collapse
Affiliation(s)
- João Pedro Ferrari-Souza
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pâmela C L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andréa L Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Firoza Z Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Douglas T Leffa
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Cécile Tissot
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Carolina Soares
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Gleb Bezgin
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Artificial Intelligence and Computational Neurosciences Laboratory, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nina Margherita Poltronetti
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Ann Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - William E Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Ziad S Saad
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, USA
| | - Hartmuth C Kolb
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, USA
| | - Thomas K Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Victor L Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L Tudorascu
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 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
- UW Department of Medicine, School of Medicine and Public Health, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Brum WS, Cullen NC, Janelidze S, Ashton NJ, Zimmer ER, Therriault J, Benedet AL, Rahmouni N, Tissot C, Stevenson J, Servaes S, Triana-Baltzer G, Kolb HC, Palmqvist S, Stomrud E, Rosa-Neto P, Blennow K, Hansson O. A two-step workflow based on plasma p-tau217 to screen for amyloid β positivity with further confirmatory testing only in uncertain cases. NATURE AGING 2023; 3:1079-1090. [PMID: 37653254 PMCID: PMC10501903 DOI: 10.1038/s43587-023-00471-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/18/2023] [Indexed: 09/02/2023]
Abstract
Cost-effective strategies for identifying amyloid-β (Aβ) positivity in patients with cognitive impairment are urgently needed with recent approvals of anti-Aβ immunotherapies for Alzheimer's disease (AD). Blood biomarkers can accurately detect AD pathology, but it is unclear whether their incorporation into a full diagnostic workflow can reduce the number of confirmatory cerebrospinal fluid (CSF) or positron emission tomography (PET) tests needed while accurately classifying patients. We evaluated a two-step workflow for determining Aβ-PET status in patients with mild cognitive impairment (MCI) from two independent memory clinic-based cohorts (n = 348). A blood-based model including plasma tau protein 217 (p-tau217), age and APOE ε4 status was developed in BioFINDER-1 (area under the curve (AUC) = 89.3%) and validated in BioFINDER-2 (AUC = 94.3%). In step 1, the blood-based model was used to stratify the patients into low, intermediate or high risk of Aβ-PET positivity. In step 2, we assumed referral only of intermediate-risk patients to CSF Aβ42/Aβ40 testing, whereas step 1 alone determined Aβ-status for low- and high-risk groups. Depending on whether lenient, moderate or stringent thresholds were used in step 1, the two-step workflow overall accuracy for detecting Aβ-PET status was 88.2%, 90.5% and 92.0%, respectively, while reducing the number of necessary CSF tests by 85.9%, 72.7% and 61.2%, respectively. In secondary analyses, an adapted version of the BioFINDER-1 model led to successful validation of the two-step workflow with a different plasma p-tau217 immunoassay in patients with cognitive impairment from the TRIAD cohort (n = 84). In conclusion, using a plasma p-tau217-based model for risk stratification of patients with MCI can substantially reduce the need for confirmatory testing while accurately classifying patients, offering a cost-effective strategy to detect AD in memory clinic settings.
Collapse
Affiliation(s)
- Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia, South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biological Sciences: Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- McGill Centre for Studies in Aging, McGill University, Montreal, Québec, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | | | - Hartmuth C Kolb
- Neuroscience Biomarkers, Janssen Research & Development, La Jolla, CA, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Québec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
30
|
Ashton NJ, Brum WS, Di Molfetta G, Benedet AL, Arslan B, Jonatis E, Langhough RE, Cody K, Wilson R, Carlsson CM, Vanmechelen E, Montoliu-Gaya L, Lantero-Rodriguez J, Rahmouni N, Tissot C, Stevenson J, Servaes S, Therriault J, Pascoal T, Lleó A, Alcolea D, Fortea J, Rosa-Neto P, Johnson S, Jeromin A, Blennow K, Zetterberg H. Diagnostic accuracy of the plasma ALZpath pTau217 immunoassay to identify Alzheimer's disease pathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.11.23292493. [PMID: 37502842 PMCID: PMC10370224 DOI: 10.1101/2023.07.11.23292493] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Importance Phosphorylated tau (pTau) is a specific blood biomarker for Alzheimer's disease (AD) pathology, with pTau217 considered to have the most utility. However, availability of pTau217 tests for research and clinical use has been limited. Expanding access to this highly accurate AD biomarker is crucial for wider evaluation and implementation of AD blood tests. Objective To determine the utility of a novel and commercially available Single molecule array (Simoa) for plasma pTau217 (ALZpath) to detect AD pathology. To evaluate references ranges for abnormal Aβ across three selected cohorts. Design Setting Participants Three single-centre observational cohorts were involved in the study: Translational Biomarkers in Aging and Dementia (TRIAD), Wisconsin Registry for Alzheimer's Prevention (WRAP), and Sant Pau Initiative on Neurodegeneration (SPIN). MRI, Aβ-PET, and tau-PET data were available for TRIAD and WRAP, while CSF biomarkers were additionally measured in a subset of TRIAD and SPIN. Plasma measurements of pTau181, pTau217 (ALZpath), pTau231, Aβ42/40, GFAP, and NfL, were available for all cohorts. Longitudinal blood biomarker data spanning 3 years for TRIAD and 8 years for WRAP were included. Exposures MRI, Aβ-PET, tau-PET, CSF biomarkers (Aβ42/40 and pTau immunoassays) and plasma pTau217 (ALZpath Simoa). Main Outcomes and Measures The accuracy of plasma pTau217 for detecting abnormal amyloid and tau pathology. Longitudinal pTau217 change according to baseline pathology status. Results The study included 786 participants (mean [SD] age, 66.3 [9.7] years; 504 females [64.1%]) were included in the study. High accuracy was observed in identifying elevated Aβ (AUC, 0.92-0.96; 95%CI 0.89-0.99) and tau pathology (AUC, 0.93-0.97; 95%CI 0.84-0.99) across all cohorts. These accuracies were significantly higher than other plasma biomarker combinations and comparable to CSF biomarkers. The detection of abnormal Aβ pathology using binary or three-range references yielded reproducible results. Longitudinally, plasma pTau217 showed an annual increase only in Aβ-positive individuals, with the highest increase observed in those with tau-positivity. Conclusions and Relevance The ALZpath plasma pTau217 Simoa assay accurately identifies biological AD, comparable to CSF biomarkers, with reproducible cut-offs across cohorts. It detects longitudinal changes, including at the preclinical stage, and is the first widely available, accessible, and scalable blood test for pTau217 detection.
Collapse
Affiliation(s)
- Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & 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
| | - Wagner S. Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & 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
| | - Guglielmo Di Molfetta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Andrea L. Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Burak Arslan
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Erin Jonatis
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Rebecca E. Langhough
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Karly Cody
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Rachael Wilson
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medicine, Division of Geriatrics and Gerontology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | | | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Cecile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Tharick Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal
- Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Sterling Johnson
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
- 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 Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| |
Collapse
|
31
|
Therriault J, Lussier FZ, Tissot C, Chamoun M, Stevenson J, Rahmouni N, Pallen V, Bezgin G, Servaes S, Kunach P, Wang Y, Fernandez‐Arias J, Vermeiren M, Pascoal TA, Massarweh G, Vitali P, Soucy J, Saha‐Chaudhuri P, Gauthie S, Rosa‐Neto P. Amyloid beta plaque accumulation with longitudinal [18F]AZD4694 PET. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12391. [PMID: 37644990 PMCID: PMC10461075 DOI: 10.1002/dad2.12391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 11/01/2022] [Accepted: 11/29/2022] [Indexed: 08/31/2023]
Abstract
Introduction [18F]AZD4694 is an amyloid beta (Aβ) imaging agent used in several observational studies and clinical trials. However, no studies have yet published data on longitudinal Aβ accumulation measured with [18F]AZD4694. Methods We assessed 146 individuals who were evaluated with [18F]AZD4694 at baseline and 2-year follow-up. We calculated annual rates of [18F]AZD4694 change for clinically defined and biomarker-defined groups. Results Cognitively unimpaired (CU) older adults displayed subtle [18F]AZD4694 standardized uptake value ratio (SUVR) accumulation over the follow-up period. In contrast, Aβ positive CU older adults displayed higher annual [18F]AZD4694 SUVR increases. [18F]AZD4694 SUVR accumulation in Aβ positive mild cognitive impairment (MCI) and dementia was modest across the neocortex. Discussion Larger increases in [18F]AZD4694 SUVR were observed in CU individuals who had abnormal amyloid positron emission tomography levels at baseline. [18F]AZD4694 can be used to monitor Aβ levels in therapeutic trials as well as clinical settings, particularly prior to initiating anti-amyloid therapies.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Firoza Z. Lussier
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Cécile Tissot
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Mira Chamoun
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Jenna Stevenson
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Nesrine Rahmouni
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Vanessa Pallen
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Gleb Bezgin
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Stijn Servaes
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Peter Kunach
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Yi‐Ting Wang
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Jaime Fernandez‐Arias
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Marie Vermeiren
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
| | - Tharick A. Pascoal
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Gassan Massarweh
- Department of RadiochemistryMcGill UniversityMontrealQuebecCanada
| | - Paolo Vitali
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Jean‐Paul Soucy
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Paramita Saha‐Chaudhuri
- Department of EpidemiologyBiostatistics and Occupational HealthMcGill UniversityMontrealQuebecCanada
- Department of Mathematics & StatisticsUniversity of VermontBurlingtonVermontUSA
| | - Serge Gauthie
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| | - Pedro Rosa‐Neto
- Translational Neuroimaging LaboratoryDouglas Mental Health InstituteMcGill University Research Centre for Studies in AgingMontrealQuebecCanada
- Department of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
32
|
Montoliu-Gaya L, Benedet AL, Tissot C, Vrillon A, Ashton NJ, Brum WS, Lantero-Rodriguez J, Stevenson J, Nilsson J, Sauer M, Rahmouni N, Brinkmalm G, Lussier FZ, Pascoal TA, Skoog I, Kern S, Zetterberg H, Paquet C, Gobom J, Rosa-Neto P, Blennow K. Mass spectrometric simultaneous quantification of tau species in plasma shows differential associations with amyloid and tau pathologies. NATURE AGING 2023; 3:661-669. [PMID: 37198279 PMCID: PMC10275761 DOI: 10.1038/s43587-023-00405-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 03/22/2023] [Indexed: 05/19/2023]
Abstract
Blood phosphorylated tau (p-tau) biomarkers, at differing sites, demonstrate high accuracy to detect Alzheimer's disease (AD). However, knowledge on the optimal marker for disease identification across the AD continuum and the link to pathology is limited. This is partly due to heterogeneity in analytical methods. In this study, we employed an immunoprecipitation mass spectrometry method to simultaneously quantify six phosphorylated (p-tau181, p-tau199, p-tau202, p-tau205, p-tau217 and p-tau231) and two non-phosphorylated plasma tau peptides in a total of 214 participants from the Paris Lariboisière and Translational Biomarkers of Aging and Dementia cohorts. Our results indicate that p-tau217, p-tau231 and p-tau205 are the plasma tau forms that best reflect AD-related brain changes, although with distinct emergences along the disease course and correlations with AD features-amyloid and tau. These findings support the differential association of blood p-tau variants with AD pathology, and our method offers a potential tool for disease staging in clinical trials.
Collapse
Grants
- R01 AG068398 NIA NIH HHS
- BrightFocus Foundation (BrightFocus)
- Alzheimerfonden
- Stiftelsen för Gamla Tjänarinnor (Foundation for Old Servants)
- AV is funded by Fondation Ophtalmologique Adolphe de Rothschild, Fondation Philipe Chatrier, Amicale des Anciens Internes des Hôpitaux de Paris, Fondation Vaincre Alzheimer, the Swedish Dementia Foundation (Demensfonden), Gun and Bertil Stohnes Foundation and Gamla Tjänarinnor Foundation.
- JN is supported by Demensfonden and the Foundation for Gamla Tjänarinnor (#2020-00959 and #2021-01153).
- HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Union’s Horizon Europe research and innovation programme under grant agreement No 101053962, Swedish State Support for Clinical Research (#ALFGBG-71320), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), the AD Strategic Fund and the Alzheimer’s Association (#ADSF-21-831376-C, #ADSF-21-831381-C, and #ADSF-21-831377-C), the Bluefield Project, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2022-0270), the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 860197 (MIRIADE), the European Union Joint Programme – Neurodegenerative Disease Research (JPND2021-00694), and the UK Dementia Research Institute at UCL (UKDRI-1003).
- KB is supported by the Swedish Research Council (#2017-00915 and #2022-00732), the Alzheimer Drug Discovery Foundation (ADDF), USA (#RDAPB-201809-2016615), the Swedish Alzheimer Foundation (#AF-930351, #AF-939721 and #AF-968270), Hjärnfonden, Sweden (#FO2017-0243 and #ALZ2022-0006), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986 and #ALFGBG-965240), the European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236), the National Institute of Health (NIH), USA, (grant #1R01AG068398-01), the Alzheimer’s Association 2021 Zenith Award (ZEN-21-848495), and the Alzheimer’s Association 2022-2025 Grant (SG-23-1038904 QC)
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Agathe Vrillon
- Université de Paris, Cognitive Neurology Center, GHUNord APHP Hospital Lariboisière Fernand Widal, Paris, France
- Université de Paris, Inserm UMRS11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, 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
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Johanna Nilsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Mathias Sauer
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ingmar Skoog
- Department of Neuropsychiatric Epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AgeCap) at the University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Silke Kern
- Department of Neuropsychiatric Epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AgeCap) at the University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- UW Department of Medicine, School of Medicine and Public Health, Madison, WI, USA
| | - Claire Paquet
- Université de Paris, Cognitive Neurology Center, GHUNord APHP Hospital Lariboisière Fernand Widal, Paris, France
- Université de Paris, Inserm UMRS11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| |
Collapse
|
33
|
Tissot C, Servaes S, Lussier FZ, Ferrari-Souza JP, Therriault J, Ferreira PCL, Bezgin G, Bellaver B, Leffa DT, Mathotaarachchi SS, Chamoun M, Stevenson J, Rahmouni N, Kang MS, Pallen V, Margherita-Poltronetti N, Wang YT, Fernandez-Arias J, Benedet AL, Zimmer ER, Soucy JP, Tudorascu DL, Cohen AD, Sharp M, Gauthier S, Massarweh G, Lopresti B, Klunk WE, Baker SL, Villemagne VL, Rosa-Neto P, Pascoal TA. The Association of Age-Related and Off-Target Retention with Longitudinal Quantification of [ 18F]MK6240 Tau PET in Target Regions. J Nucl Med 2023; 64:452-459. [PMID: 36396455 PMCID: PMC10071794 DOI: 10.2967/jnumed.122.264434] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022] Open
Abstract
6-(fluoro-18F)-3-(1H-pyrrolo[2,3-c]pyridin-1-yl)isoquinolin-5-amine ([18F]MK6240) tau PET tracer quantifies the brain tau neurofibrillary tangle load in Alzheimer disease. The aims of our study were to test the stability of common reference region estimates in the cerebellum over time and across diagnoses and evaluate the effects of age-related and off-target retention on the longitudinal quantification of [18F]MK6240 in target regions. Methods: We assessed reference, target, age-related, and off-target regions in 125 individuals across the aging and Alzheimer disease spectrum with longitudinal [18F]MK6240 SUVs and SUV ratios (SUVRs) (mean ± SD, 2.25 ± 0.40 y of follow-up). We obtained SUVR from meninges, exhibiting frequent off-target retention with [18F]MK6240. Additionally, we compared tracer uptake between 37 cognitively unimpaired young (CUY) (mean age, 23.41 ± 3.33 y) and 27 cognitively unimpaired older (CU) adults (amyloid-β-negative and tau-negative, 58.50 ± 9.01 y) to identify possible nonvisually apparent, age-related signal. Two-tailed t testing and Pearson correlation testing were used to determine the difference between groups and associations between changes in region uptake, respectively. Results: Inferior cerebellar gray matter SUV did not differ on the basis of diagnosis and amyloid-β status, cross-sectionally and over time. [18F]MK6240 uptake significantly differed between CUY and CU adults in the putamen or pallidum (affecting ∼75% of the region) and in the Braak II region (affecting ∼35%). Changes in meningeal and putamen or pallidum SUVRs did not significantly differ from zero, nor did they vary across diagnostic groups. We did not observe significant correlations between longitudinal changes in age-related or meningeal off-target retention and changes in target regions, whereas changes in all target regions were strongly correlated. Conclusion: Inferior cerebellar gray matter was similar across diagnostic groups cross-sectionally and stable over time and thus was deemed a suitable reference region for quantification. Despite not being visually perceptible, [18F]MK6240 has age-related retention in subcortical regions, at a much lower magnitude but topographically colocalized with significant off-target signal of the first-generation tau tracers. The lack of correlation between changes in age-related or meningeal and target retention suggests little influence of possible off-target signals on longitudinal tracer quantification. Nevertheless, the age-related retention in the Braak II region needs to be further investigated. Future postmortem studies should elucidate the source of the newly reported age-related [18F]MK6240 signal, and in vivo studies should further explore its impact on tracer quantification.
Collapse
Affiliation(s)
- Cécile Tissot
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Stijn Servaes
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Firoza Z Lussier
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - João Pedro Ferrari-Souza
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Porto Alegre, Brazil
| | - Joseph Therriault
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Pâmela C L Ferreira
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gleb Bezgin
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Bruna Bellaver
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Porto Alegre, Brazil
| | - Douglas Teixeira Leffa
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sulantha S Mathotaarachchi
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Mira Chamoun
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Jenna Stevenson
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Min Su Kang
- Artificial Intelligence and Computational Neurosciences Lab, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- L.C. Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Vanessa Pallen
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Nina Margherita-Poltronetti
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Yi-Ting Wang
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Jaime Fernandez-Arias
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | | | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Porto Alegre, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Dana L Tudorascu
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Annie D Cohen
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Serge Gauthier
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
- Douglas Mental Health Institute, Montreal, Quebec, Canada
| | - Gassan Massarweh
- Department of Radiochemistry, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Brian Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - William E Klunk
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Victor L Villemagne
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Pedro Rosa-Neto
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
- Montreal Neurological Institute, Montreal, Quebec, Canada
- Douglas Mental Health Institute, Montreal, Quebec, Canada
| | - Tharick A Pascoal
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
| |
Collapse
|
34
|
Therriault J, Gauthier S, Rosa-Neto P. Applications of Alzheimer's disease staging to clinical trials. Aging (Albany NY) 2023; 15:4-5. [PMID: 36622284 PMCID: PMC9876645 DOI: 10.18632/aging.204482] [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: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 01/07/2023]
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Canada
- Department of Neurology and Neurosurgery, McGill University, Canada
- Montreal Neurological Institute, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Canada
- Department of Neurology and Neurosurgery, McGill University, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, Canada
- Department of Neurology and Neurosurgery, McGill University, Canada
- Montreal Neurological Institute, Canada
| |
Collapse
|
35
|
Shuping JL, Matthews DC, Adamczuk K, Scott D, Rowe CC, Kreisl WC, Johnson SC, Lukic AS, Johnson KA, Rosa‐Neto P, Andrews RD, Van Laere K, Cordes L, Ward L, Wilde CL, Barakos J, Purcell DD, Devanand DP, Stern Y, Luchsinger JA, Sur C, Price JC, Brickman AM, Klunk WE, Boxer AL, Mathotaarachchi SS, Lao PJ, Evelhoch JL. Development, initial validation, and application of a visual read method for [ 18F]MK-6240 tau PET. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12372. [PMID: 36873926 PMCID: PMC9983143 DOI: 10.1002/trc2.12372] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 02/15/2023]
Abstract
Background The positron emission tomography (PET) radiotracer [18F]MK-6240 exhibits high specificity for neurofibrillary tangles (NFTs) of tau protein in Alzheimer's disease (AD), high sensitivity to medial temporal and neocortical NFTs, and low within-brain background. Objectives were to develop and validate a reproducible, clinically relevant visual read method supporting [18F]MK-6240 use to identify and stage AD subjects versus non-AD and controls. Methods Five expert readers used their own methods to assess 30 scans of mixed diagnosis (47% cognitively normal, 23% mild cognitive impairment, 20% AD, 10% traumatic brain injury) and provided input regarding regional and global positivity, features influencing assessment, confidence, practicality, and clinical relevance. Inter-reader agreement and concordance with quantitative values were evaluated to confirm that regions could be read reliably. Guided by input regarding clinical applicability and practicality, read classifications were defined. The readers read the scans using the new classifications, establishing by majority agreement a gold standard read for those scans. Two naïve readers were trained and read the 30-scan set, providing initial validation. Inter-rater agreement was further tested by two trained independent readers in 131 scans. One of these readers used the same method to read a full, diverse database of 1842 scans; relationships between read classification, clinical diagnosis, and amyloid status as available were assessed. Results Four visual read classifications were determined: no uptake, medial temporal lobe (MTL) only, MTL and neocortical uptake, and uptake outside MTL. Inter-rater kappas were 1.0 for the naïve readers gold standard scans read and 0.98 for the independent readers 131-scan read. All scans in the full database could be classified; classification frequencies were concordant with NFT histopathology literature. Discussion This four-class [18F]MK-6240 visual read method captures the presence of medial temporal signal, neocortical expansion associated with disease progression, and atypical distributions that may reflect different phenotypes. The method demonstrates excellent trainability, reproducibility, and clinical relevance supporting clinical use. Highlights A visual read method has been developed for [18F]MK-6240 tau positron emission tomography.The method is readily trainable and reproducible, with inter-rater kappas of 0.98.The read method has been applied to a diverse set of 1842 [18F]MK-6240 scans.All scans from a spectrum of disease states and acquisitions could be classified.Read classifications are consistent with histopathological neurofibrillary tangle staging literature.
Collapse
Affiliation(s)
| | | | | | | | - Christopher C. Rowe
- Department of Molecular Imaging and TherapyAustin HealthMelbourneVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | - William C. Kreisl
- Department of NeurologyThe Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Columbia University Irving Medical CenterVagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
| | - Sterling C. Johnson
- Department of MedicineDivision of GeriatricsAlzheimer's Disease Research Center, University of WisconsinMadisonWisconsinUSA
| | | | - Keith A. Johnson
- The Gordon Center for Medical ImagingDepartment of NeurologyCenter for Alzheimer Research and TreatmentBrigham and Women's HospitalBostonMassachusettsUSA
- Department of RadiologyAthinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalHarvard Medical SchoolCharlestownMassachusettsUSA
| | - Pedro Rosa‐Neto
- Montreal Neurological InstituteMcGill UniversityMontréalQuebecCanada
| | | | - Koen Van Laere
- Nuclear Medicine and Molecular ImagingDepartment of Imaging and Pathology KU LeuvenLeuvenBelgium
| | | | - Larry Ward
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| | | | | | | | - Davangere P. Devanand
- Department of NeurologyThe Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Columbia University Irving Medical CenterVagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
- Department of PsychiatryColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Yaakov Stern
- Department of NeurologyThe Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Columbia University Irving Medical CenterVagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
- Department of PsychiatryColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of NeurologyGertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Jose A. Luchsinger
- Columbia University Irving Medical CenterVagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
- Department of Medicine and EpidemiologyColumbia University Irving Medical CenterNew York, NY, 10032 USA For Dr. LuchsingerUSA
| | | | - Julie C. Price
- Department of RadiologyAthinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalHarvard Medical SchoolCharlestownMassachusettsUSA
| | - Adam M. Brickman
- Department of NeurologyThe Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Columbia University Irving Medical CenterVagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
- Department of NeurologyGertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - William E. Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Adam L. Boxer
- Department of NeurologyMemory and Aging CenterUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | | - Patrick J. Lao
- Department of NeurologyThe Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Columbia University Irving Medical CenterVagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
- Department of NeurologyGertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | | |
Collapse
|
36
|
Hou J, Chen Y, Grajales-Reyes G, Colonna M. TREM2 dependent and independent functions of microglia in Alzheimer's disease. Mol Neurodegener 2022; 17:84. [PMID: 36564824 PMCID: PMC9783481 DOI: 10.1186/s13024-022-00588-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/02/2022] [Indexed: 12/25/2022] Open
Abstract
Microglia are central players in brain innate immunity and have been the subject of extensive research in Alzheimer's disease (AD). In this review, we aim to summarize the genetic and functional discoveries that have advanced our understanding of microglia reactivity to AD pathology. Given the heightened AD risk posed by rare variants of the microglial triggering receptor expressed on myeloid cells 2 (TREM2), we will focus on the studies addressing the impact of this receptor on microglia responses to amyloid plaques, tauopathy and demyelination pathologies in mouse and human. Finally, we will discuss the implications of recent discoveries on microglia and TREM2 biology on potential therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jinchao Hou
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Yun Chen
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA ,grid.4367.60000 0001 2355 7002Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Gary Grajales-Reyes
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Marco Colonna
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
37
|
Ferrari-Souza JP, Ferreira PCL, Bellaver B, Tissot C, Wang YT, Leffa DT, Brum WS, Benedet AL, Ashton NJ, De Bastiani MA, Rocha A, Therriault J, Lussier FZ, Chamoun M, Servaes S, Bezgin G, Kang MS, Stevenson J, Rahmouni N, Pallen V, Poltronetti NM, Klunk WE, Tudorascu DL, Cohen AD, Villemagne VL, Gauthier S, Blennow K, Zetterberg H, Souza DO, Karikari TK, Zimmer ER, Rosa-Neto P, Pascoal TA. Astrocyte biomarker signatures of amyloid-β and tau pathologies in Alzheimer's disease. Mol Psychiatry 2022; 27:4781-4789. [PMID: 35948658 PMCID: PMC9734046 DOI: 10.1038/s41380-022-01716-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 01/07/2023]
Abstract
Astrocytes can adopt multiple molecular phenotypes in the brain of Alzheimer's disease (AD) patients. Here, we studied the associations of cerebrospinal fluid (CSF) glial fibrillary acidic protein (GFAP) and chitinase-3-like protein 1 (YKL-40) levels with brain amyloid-β (Aβ) and tau pathologies. We assessed 121 individuals across the aging and AD clinical spectrum with positron emission tomography (PET) brain imaging for Aβ ([18F]AZD4694) and tau ([18F]MK-6240), as well as CSF GFAP and YKL-40 measures. We observed that higher CSF GFAP levels were associated with elevated Aβ-PET but not tau-PET load. By contrast, higher CSF YKL-40 levels were associated with elevated tau-PET but not Aβ-PET burden. Structural equation modeling revealed that CSF GFAP and YKL-40 mediate the effects of Aβ and tau, respectively, on hippocampal atrophy, which was further associated with cognitive impairment. Our results suggest the existence of distinct astrocyte biomarker signatures in response to brain Aβ and tau accumulation, which may contribute to our understanding of the complex link between reactive astrogliosis heterogeneity and AD progression.
Collapse
Affiliation(s)
- João Pedro Ferrari-Souza
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cécile Tissot
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Douglas T Leffa
- ADHD Outpatient Program & Development Psychiatry Program, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Wagner S Brum
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Andréa L Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Marco Antônio De Bastiani
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andréia Rocha
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nina Margherita Poltronetti
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 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
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
38
|
Therriault J, Pascoal TA, Lussier FZ, Tissot C, Chamoun M, Bezgin G, Servaes S, Benedet AL, Ashton NJ, Karikari TK, Lantero-Rodriguez J, Kunach P, Wang YT, Fernandez-Arias J, Massarweh G, Vitali P, Soucy JP, Saha-Chaudhuri P, Blennow K, Zetterberg H, Gauthier S, Rosa-Neto P. Biomarker modeling of Alzheimer's disease using PET-based Braak staging. NATURE AGING 2022; 2:526-535. [PMID: 37118445 PMCID: PMC10154209 DOI: 10.1038/s43587-022-00204-0] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/08/2022] [Indexed: 04/30/2023]
Abstract
Gold-standard diagnosis of Alzheimer's disease (AD) relies on histopathological staging systems. Using the topographical information from [18F]MK6240 tau positron-emission tomography (PET), we applied the Braak tau staging system to 324 living individuals. We used PET-based Braak stage to model the trajectories of amyloid-β, phosphorylated tau (pTau) in cerebrospinal fluid (pTau181, pTau217, pTau231 and pTau235) and plasma (pTau181 and pTau231), neurodegeneration and cognitive symptoms. We identified nonlinear AD biomarker trajectories corresponding to the spatial extent of tau-PET, with modest biomarker changes detectable by Braak stage II and significant changes occurring at stages III-IV, followed by plateaus. Early Braak stages were associated with isolated memory impairment, whereas Braak stages V-VI were incompatible with normal cognition. In 159 individuals with follow-up tau-PET, progression beyond stage III took place uniquely in the presence of amyloid-β positivity. Our findings support PET-based Braak staging as a framework to model the natural history of AD and monitor AD severity in living humans.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Andrea L Benedet
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nicholas J Ashton
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- Biomedical Research Unit for Dementia at South London, NIHR Biomedical Research Centre for Mental Health and Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Juan Lantero-Rodriguez
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Kunach
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Gassan Massarweh
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Paolo Vitali
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | | | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
39
|
Ramanan VK, Heckman MG, Lesnick TG, Przybelski SA, Cahn EJ, Kosel ML, Murray ME, Mielke MM, Botha H, Graff-Radford J, Jones DT, Lowe VJ, Machulda MM, Jack CR, Knopman DS, Petersen RC, Ross OA, Vemuri P. Tau polygenic risk scoring: a cost-effective aid for prognostic counseling in Alzheimer's disease. Acta Neuropathol 2022; 143:571-583. [PMID: 35412102 PMCID: PMC9109940 DOI: 10.1007/s00401-022-02419-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 11/28/2022]
Abstract
Tau deposition is one of two hallmark features of biologically defined Alzheimer's disease (AD) and is more closely related to cognitive decline than amyloidosis. Further, not all amyloid-positive individuals develop tauopathy, resulting in wide heterogeneity in clinical outcomes across the population with AD. We hypothesized that a polygenic risk score (PRS) based on tau PET (tau PRS) would capture the aggregate inherited susceptibility/resistance architecture influencing tau accumulation, beyond solely the measurement of amyloid-β burden. Leveraging rich multimodal data from a population-based sample of older adults, we found that this novel tau PRS was a strong surrogate of tau PET deposition and captured a significant proportion of the variance in tau PET levels as compared with amyloid PET burden, APOE (apolipoprotein E) ε4 (the most common risk allele for AD), and a non-APOE PRS of clinical case-control AD risk variants. In independent validation samples, the tau PRS was associated with cerebrospinal fluid phosphorylated tau levels in one cohort and with postmortem Braak neurofibrillary tangle stage in another. We also observed an association of the tau PRS with longitudinal cognitive trajectories, including a statistical interaction of the tau PRS with amyloid burden on cognitive decline. Although additional study is warranted, these findings demonstrate the potential utility of a tau PRS for capturing the collective genetic background influencing tau deposition in the general population. In the future, a tau PRS could be leveraged for cost-effective screening and risk stratification to guide trial enrollment and clinical interventions in AD.
Collapse
Affiliation(s)
- Vijay K Ramanan
- Department of Neurology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Michael G Heckman
- Department of Quantitative Health Sciences, Mayo Clinic-Florida, Jacksonville, FL, 32224, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Elliot J Cahn
- Department of Quantitative Health Sciences, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Matthew L Kosel
- Department of Quantitative Health Sciences, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic-Florida, Jacksonville, FL, 32224, USA
| | - Michelle M Mielke
- Department of Neurology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Quantitative Health Sciences, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jonathan Graff-Radford
- Department of Neurology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Radiology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ronald C Petersen
- Department of Neurology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Quantitative Health Sciences, Mayo Clinic-Minnesota, Rochester, MN, 55905, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic-Florida, Jacksonville, FL, 32224, USA
- Department of Clinical Genomics, Mayo Clinic-Florida, Jacksonville, FL, 32224, USA
| | - Prashanthi Vemuri
- Department of Neuroscience, Mayo Clinic-Florida, Jacksonville, FL, 32224, USA.
- Department of Radiology, Mayo Clinic-Minnesota, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
40
|
Cassidy CM, Therriault J, Pascoal TA, Cheung V, Savard M, Tuominen L, Chamoun M, McCall A, Celebi S, Lussier F, Massarweh G, Soucy JP, Weinshenker D, Tardif C, Ismail Z, Gauthier S, Rosa-Neto P. Association of locus coeruleus integrity with Braak stage and neuropsychiatric symptom severity in Alzheimer's disease. Neuropsychopharmacology 2022; 47:1128-1136. [PMID: 35177805 PMCID: PMC8938499 DOI: 10.1038/s41386-022-01293-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/07/2022] [Accepted: 02/02/2022] [Indexed: 12/16/2022]
Abstract
The clinical and pathophysiological correlates of locus coeruleus (LC) degeneration in Alzheimer's disease (AD) could be clarified using a method to index LC integrity in vivo, neuromelanin-sensitive MRI (NM-MRI). We examined whether integrity of the LC-norepinephrine system, assessed with NM-MRI, is associated with stage of AD and with neuropsychiatric symptoms (NPS), independent of cortical pathophysiology (amyloid-β and tau burden). Cognitively normal older adults (n = 118), and individuals with mild cognitive impairment (MCI, n = 44), and AD (n = 28) underwent MR imaging and tau and amyloid-β positron emission tomography (with [18F]MK6240 and [18F]AZD4694, respectively). Integrity of the LC-norepinephrine system was assessed based on contrast-to-noise ratio of the LC on NM-MRI images. Braak stage of AD was derived from regional binding of [18F]MK6240. NPS were assessed with the Mild Behavioral Impairment Checklist (MBI-C). LC signal contrast was decreased in tau-positive participants (t186 = -4.00, p = 0.0001) and negatively correlated to Braak stage (Spearman ρ = -0.31, p = 0.00006). In tau-positive participants (n = 51), higher LC signal predicted NPS severity (ρ = 0.35, p = 0.019) independently of tau burden, amyloid-β burden, and cortical gray matter volume. This relationship appeared to be driven by the impulse dyscontrol domain of NPS, which was highly correlated to LC signal (ρ = 0.44, p = 0.0027). NM-MRI reveals loss of LC integrity that correlates to severity of AD. However, LC preservation in AD may also have negative consequences by conferring risk for impulse control symptoms. NM-MRI shows promise as a practical biomarker that could have utility in predicting the risk of NPS or guiding their treatment in AD.
Collapse
Affiliation(s)
- Clifford M. Cassidy
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada ,grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada
| | - Joseph Therriault
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Tharick A. Pascoal
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - Victoria Cheung
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada
| | - Melissa Savard
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Lauri Tuominen
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada
| | - Mira Chamoun
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Adelina McCall
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada
| | - Seyda Celebi
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada
| | - Firoza Lussier
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Gassan Massarweh
- grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - Jean-Paul Soucy
- grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - David Weinshenker
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University School of Medicine, Atlanta, GA USA
| | - Christine Tardif
- grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - Zahinoor Ismail
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | - Serge Gauthier
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Alzheimer’s Disease Research Unit, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC Canada
| | - Pedro Rosa-Neto
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| |
Collapse
|
41
|
Michael HU, Youbi E, Ohadoma SC, Ramlall S, Oosthuizen F, Polyakova M. A Meta-Analytic Review of the Effect of Antiretroviral Therapy on Neurocognitive Outcomes in Adults Living with HIV-1 in Low-and Middle-Income Countries. Neuropsychol Rev 2021; 32:828-854. [PMID: 34757490 DOI: 10.1007/s11065-021-09527-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/11/2021] [Indexed: 01/29/2023]
Abstract
HIV-associated neurocognitive impairment remains a challenge even in the era of antiretroviral therapy (ART). Over 90% of people living with HIV are in low- and middle-income countries. Hence, it is not surprising that such countries bear a considerable burden of comorbidities like HIV-associated neurocognitive impairment despite an overall increase in life expectancy. The literature suggests differences in patient characteristics, clinical profile, prevalent HIV subtypes, treatment choices, pharmacogenetics, and socioeconomic factors between low- and middle-income countries compared with high-income countries. Therefore, we aimed to evaluate the effect of ART on neurocognitive outcomes in low- and middle-income countries. A comprehensive search of five databases (PubMed, CINAHL, CENTRAL, PsychInfo, Google scholar) for studies published between 1996 to 2020 was performed to identify studies that reported neurocognitive outcomes in ART-treated and ART naïve HIV positive individuals. Two independent reviewers conducted study screening, data extraction, and evaluation of the risk of bias. Pooled effect size estimates (Hedges' g) and 95% CI were computed using random-effects models. Sensitivity analysis, subgroup analysis, meta-regression, and evaluation of publication bias were also conducted. Forty studies (24 cross-sectional, 13 longitudinal studies, and two randomized controlled trials) contributed to a series of meta-analyses. We found significant small to moderate effects of antiretroviral therapy for global cognition (Hedges' g observed = 0.30; 95% CI: 0.15, 0.44; k = 25; p = 0.0003; I2 = 92.1%; tau = 0.32; Q = 305.1), executive function (Hedges' g = 0.24, 95%CI: 0.02,0.46; p-0.04; k = 8; I2 = 37.5%; tau = 0.23; Q = 11.2), and speed of information processing (Hedges' g = 0.25, 95% CI: 0.05, 0.45; k = 9; p = 0.02; I2 = 86.4%; tau = 0.21; Q = 58.9). We found no significant ART effect on attention-working memory, learning and memory, motor function, and verbal fluency. No significant effect was seen with the duration of therapy, efavirenz use, and Central Penetrating Effectiveness (CPE) of antiretroviral therapy. Subgroup analyses identified study design (between-group and within-group; cross-sectional and longitudinal) and normative scores as significant sources of heterogeneity. Meta-regression analysis indicated that nadir CD4 modified the magnitude of ART's effect on cognitive outcomes. Age, gender, and country income-group were not significant moderators. Our findings provide systematic evidence that antiretroviral therapy improves neurocognitive outcomes in the domains of global cognition, executive function and speed of information processing, of people living with HIV in low- and middle-income countries, especially those with advanced immunosuppression. However, these findings are not definitive as they are limited by the probability of publication bias, high heterogeneity, and exclusion of significant confounders. Prospero registration number: CRD42020203791.
Collapse
Affiliation(s)
- Henry Ukachukwu Michael
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Emily Youbi
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sylvester Chika Ohadoma
- Department of Pharmacology, College of Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Suvira Ramlall
- Department of Psychiatry, Nelson R. Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South, South Africa
| | - Frasia Oosthuizen
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Maryna Polyakova
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Clinic for Psychiatry and Psychotherapy, University of Leipzig, Leipzig, Germany
- LIFE-Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Clinic for Cognitive Neurology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
42
|
Garnier-Crussard A, Krolak-Salmon P. Reader Response: Frequency of Biologically Defined Alzheimer Disease in Relation to Age, Sex, APOE ε4, and Cognitive Impairment. Neurology 2021; 97:608. [DOI: 10.1212/wnl.0000000000012585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
43
|
Therriault J, Pascoal T, Gauthier S, Rosa-Neto P. Author Response: Frequency of Biologically Defined Alzheimer Disease in Relation to Age, Sex, APOE ε4, and Cognitive Impairment. Neurology 2021; 97:609. [PMID: 35100130 DOI: 10.1212/wnl.0000000000012586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
44
|
Rates of cognitive impairment in a South African cohort of people with HIV: variation by definitional criteria and lack of association with neuroimaging biomarkers. J Neurovirol 2021; 27:579-594. [PMID: 34241815 DOI: 10.1007/s13365-021-00993-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/14/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022]
Abstract
There is wide variation in the reported prevalence of cognitive impairment in people with HIV (PWH). Part of this variation may be attributable to different studies using different methods of combining neuropsychological test scores to classify participants as either cognitively impaired or unimpaired. Our aim was to determine, in a South African cohort of PWH (N = 148), (a) how much variation in reported rates was due to method used to define cognitive impairment and (b) which method correlated best with MRI biomarkers of HIV-related brain pathology. Participants completed detailed neuropsychological assessment and underwent 3 T structural MRI and diffusion tensor imaging (DTI). We used the neuropsychological data to investigate 20 different methods of determining HIV-associated cognitive impairment. We used the neuroimaging data to obtain volumes for cortical and subcortical grey matter and total white matter and DTI metrics for several white matter tracts. Applying each of the 20 methods to the cognitive dataset resulted in a wide variation (20-97%) in estimated rates of impairment. Logistic regression models showed no method was associated with HIV-related neuroimaging abnormalities as measured by structural volumes or DTI metrics. We conclude that for the population from which this sample was drawn, much of the variation in reported rates of cognitive impairment in PWH is due to the method of classification used, and that none of these methods accurately reflects biological effects of HIV in the brain. We suggest that defining HIV-associated cognitive impairment using neuropsychological test performance only is insufficient; pre-morbid functioning, co-morbidities, cognitive symptoms, and functional impairment should always be considered.
Collapse
|
45
|
Knopman DS, Jagust WJ. Alzheimer Disease Spectrum: Syndrome and Etiology From Clinical and PET Imaging Perspectives. Neurology 2020; 96:299-300. [PMID: 33361254 DOI: 10.1212/wnl.0000000000011415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- David S Knopman
- From the Department of Neurology (D.S.K.), Mayo Clinic, Rochester MN; and School of Public Health and Helen Wills Neuroscience Institute (W.J.J.), University of California Berkeley.
| | - William J Jagust
- From the Department of Neurology (D.S.K.), Mayo Clinic, Rochester MN; and School of Public Health and Helen Wills Neuroscience Institute (W.J.J.), University of California Berkeley
| |
Collapse
|