1
|
Antoniades M, Srinivasan D, Wen J, Erus G, Abdulkadir A, Mamourian E, Melhem R, Hwang G, Cui Y, Govindarajan ST, Chen AA, Zhou Z, Yang Z, Chen J, Pomponio R, Sotardi S, An Y, Bilgel M, LaMontagne P, Singh A, Benzinger T, Beason-Held L, Marcus DS, Yaffe K, Launer L, Morris JC, Tosun D, Ferrucci L, Bryan RN, Resnick SM, Habes M, Wolk D, Fan Y, Nasrallah IM, Shou H, Davatzikos C. Relationship between MRI brain-age heterogeneity, cognition, genetics and Alzheimer's disease neuropathology. EBioMedicine 2024; 109:105399. [PMID: 39437659 DOI: 10.1016/j.ebiom.2024.105399] [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: 11/13/2023] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Brain ageing is highly heterogeneous, as it is driven by a variety of normal and neuropathological processes. These processes may differentially affect structural and functional brain ageing across individuals, with more pronounced ageing (older brain age) during midlife being indicative of later development of dementia. Here, we examined whether brain-ageing heterogeneity in unimpaired older adults related to neurodegeneration, different cognitive trajectories, genetic and amyloid-beta (Aβ) profiles, and to predicted progression to Alzheimer's disease (AD). METHODS Functional and structural brain age measures were obtained for resting-state functional MRI and structural MRI, respectively, in 3460 cognitively normal individuals across an age range spanning 42-85 years. Participants were categorised into four groups based on the difference between their chronological and predicted age in each modality: advanced age in both (n = 291), resilient in both (n = 260) or advanced in one/resilient in the other (n = 163/153). With the resilient group as the reference, brain-age groups were compared across neuroimaging features of neuropathology (white matter hyperintensity volume, neuronal loss measured with Neurite Orientation Dispersion and Density Imaging, AD-specific atrophy patterns measured with the Spatial Patterns of Abnormality for Recognition of Early Alzheimer's Disease index, amyloid burden using amyloid positron emission tomography (PET), progression to mild cognitive impairment and baseline and longitudinal cognitive measures (trail making task, mini mental state examination, digit symbol substitution task). FINDINGS Individuals with advanced structural and functional brain-ages had more features indicative of neurodegeneration and they had poor cognition. Individuals with a resilient brain-age in both modalities had a genetic variant that has been shown to be associated with age of onset of AD. Mixed brain-age was associated with selective cognitive deficits. INTERPRETATION The advanced group displayed evidence of increased atrophy across all neuroimaging features that was not found in either of the mixed groups. This is in line with biomarkers of preclinical AD and cerebrovascular disease. These findings suggest that the variation in structural and functional brain ageing across individuals reflects the degree of underlying neuropathological processes and may indicate the propensity to develop dementia in later life. FUNDING The National Institute on Aging, the National Institutes of Health, the Swiss National Science Foundation, the Kaiser Foundation Research Institute and the National Heart, Lung, and Blood Institute.
Collapse
Affiliation(s)
- Mathilde Antoniades
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Dhivya Srinivasan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Guray Erus
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Clinical Neuroscience, Center for Research in Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Elizabeth Mamourian
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yuhan Cui
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja Tirumalai Govindarajan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew A Chen
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zhen Zhou
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiong Chen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond Pomponio
- Department of Biostatistics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Susan Sotardi
- Department of Radiology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ashish Singh
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Lenore Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institute of Health, Baltimore, MD 21224, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mohamad Habes
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - David Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Christos Davatzikos
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Galvin JE, Kleiman MJ, Estes PW, Harris HM, Fung E. Cognivue Clarity characterizes mild cognitive impairment and Alzheimer's disease in biomarker confirmed cohorts in the Bio-Hermes Study. Sci Rep 2024; 14:24519. [PMID: 39424626 PMCID: PMC11489461 DOI: 10.1038/s41598-024-75304-5] [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: 08/07/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
The Bio-Hermes Study was a cross-sectional observational study designed to develop a database of blood-based and digital biomarkers to improve detection of Alzheimer's disease (AD) and mild cognitive impairment (MCI). We examined the ability of Cognivue Clarity to (a) detect MCI and AD in clinical diagnostics groups, (b) determine the presence of amyloid, and (c) distinguish between biomarker-confirmed groups. Bio-Hermes enrolled 887 participants who completed both Cognivue Clarity and amyloid PET scans (388 Cognitively Normal, 282 MCI, 217 Probable AD). Cognivue Clarity differentiated between Cognitively Normal, MCI, and probable AD in clinical cohorts, amyloid positive from amyloid negative individuals, and True Controls from MCI due to AD and AD in biomarker-confirmed cohorts (all p < 0.001) with large effect sizes. Cognivue Clarity correlated with amyloid PET and plasma amyloid and pTau (all p < 0.001). In biomarker confirmed groups, Cognivue Clarity had a positive likelihood ratio of 2.17, a negative likelihood ratio of 0.29, and a diagnostic odds ratio of 7.48. Cognivue Clarity detected cognitive impairment and differentiated between both clinically and biomarker defined MCI and AD groups. The use of Cognivue Clarity could assist with identification of MCI-AD or AD for inclusion into current treatment protocols or for enriching recruitment into clinical trials. Trial registration ClinicalTrials.gov (NCT04733989).
Collapse
Affiliation(s)
- James E Galvin
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, 7700 W Camino Real, Suite 200, Boca Raton, FL, 33433, USA.
- Cognivue, Inc, 7911 Rae Blvd, Victor, NY, 14564, USA.
| | - Michael J Kleiman
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, 7700 W Camino Real, Suite 200, Boca Raton, FL, 33433, USA
| | - Paul W Estes
- Cognivue, Inc, 7911 Rae Blvd, Victor, NY, 14564, USA
| | | | - Ernest Fung
- Cognivue, Inc, 7911 Rae Blvd, Victor, NY, 14564, USA
| |
Collapse
|
3
|
Yang B, Earnest T, Kumar S, Kothapalli D, Benzinger T, Gordon B, Sotiras A. Evaluation of ComBat harmonization for reducing across-tracer differences in regional amyloid PET analyses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.14.24308952. [PMID: 38947044 PMCID: PMC11213066 DOI: 10.1101/2024.06.14.24308952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Introduction Differences in amyloid positron emission tomography (PET) radiotracer pharmacokinetics and binding properties lead to discrepancies in amyloid-β uptake estimates. Harmonization of tracer-specific biases is crucial for optimal performance of downstream tasks. Here, we investigated the efficacy of ComBat, a data-driven harmonization model, for reducing tracer-specific biases in regional amyloid PET measurements from [18F]-florbetapir (FBP) and [11C]-Pittsburgh Compound-B (PiB). Methods One-hundred-thirteen head-to-head FBP-PiB scan pairs, scanned from the same subject within ninety days, were selected from the Open Access Series of Imaging Studies 3 (OASIS-3) dataset. The Centiloid scale, ComBat with no covariates, ComBat with biological covariates, and GAM-ComBat with biological covariates were used to harmonize both global and regional amyloid standardized uptake value ratios (SUVR). Variants of ComBat, including longitudinal ComBat and PEACE, were also tested. Intraclass correlation coefficient (ICC) and mean absolute error (MAE) were computed to measure the absolute agreement between tracers. Additionally, longitudinal amyloid SUVRs from an anti-amyloid drug trial were simulated using linear mixed effects modeling. Differences in rates-of-change between simulated treatment and placebo groups were tested, and change in statistical power/Type-I error after harmonization was quantified. Results In the head-to-head tracer comparison, ComBat with no covariates was the best at increasing ICC and decreasing MAE of both global summary and regional amyloid PET SUVRs between scan pairs of the same group of subjects. In the clinical trial simulation, harmonization with both Centiloid and ComBat increased statistical power of detecting true rate-of-change differences between groups and decreased false discovery rate in the absence of a treatment effect. The greatest benefit of harmonization was observed when groups exhibited differing FBP-to-PiB proportions. Conclusion ComBat outperformed the Centiloid scale in harmonizing both global and regional amyloid estimates. Additionally, ComBat improved the detection of rate-of-change differences between clinical trial groups. Our findings suggest that ComBat is a viable alternative to Centiloid for harmonizing regional amyloid PET analyses.
Collapse
Affiliation(s)
- Braden Yang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Tom Earnest
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Sayantan Kumar
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Deydeep Kothapalli
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Tammie Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Brian Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| | - Aristeidis Sotiras
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA 63110
| |
Collapse
|
4
|
Yamakuni R, Murakami T, Ukon N, Kakamu T, Toda W, Hattori K, Sekino H, Ishii S, Fukushima K, Matsuda H, Ugawa Y, Wakasugi N, Abe M, Ito H. Differential centiloid scale normalization techniques: comparison between hybrid PET/MRI and independently acquired MRI. Ann Nucl Med 2024; 38:835-846. [PMID: 38902587 DOI: 10.1007/s12149-024-01955-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVE Centiloid (CL) scales play an important role in semiquantitative analyses of amyloid-β (Aβ) PET. CLs are derived from the standardized uptake value ratio (SUVR), which needs Aβ positron emission tomography (PET) normalization processing. There are two methods to collect the T1-weighted imaging (T1WI) for normalization: (i) anatomical standardization using simultaneously acquired T1WI (PET/MRI), usually adapted to PET images from PET/MRI scanners, and (ii) T1WI from a separate examination (PET + MRI), usually adapted to PET images from PET/CT scanners. This study aimed to elucidate the correlations and differences in CLs between when using the above two T1WI collection methods. METHODS Among patients who underwent Aβ PET/MRI (using 11C-Pittuberg compound B (11C-PiB) or 18F-flutemetamol (18F-FMM)) at our institution from 2015 to 2023, we selected 49 patients who also underwent other additional MRI examinations, including T1WI for anatomic standardization within 3 years. Thirty-one of them underwent 11C-PiB PET/MRI, and 18 participants underwent 18F-FMM PET/MRI. Twenty-five of them, additional MRI acquisition parameters were identical to simultaneous MRI during PET, and 24 participants were different. After normalization using PET/MRI or PET + MRI method each, SUVR was measured using the Global Alzheimer's Association Initiative Network cerebral cortical and striatum Volume of Interest templates (VOI) and whole cerebellum VOI. Subsequently, CLs were calculated using the previously established equations for each Aβ PET tracer. RESULTS Between PET/MRI and PET + MRI methods, CLs correlated linearly in 11C-PiB PET (y = 1.00x - 0.11, R2 = 0.999), 18F-FMM PET (y = 0.97x - 0.12, 0.997), identical additional MRI acquisition (y = 1.00x + 0.33, 0.999), different acquisition (y = 0.98x - 0.43, 0.997), and entire study group (y = 1.00x - 0.24, 0.999). Wilcoxon signed-rank test revealed no significant differences: 11C-PiB (p = 0.49), 18F-FMM (0.08), and whole PET (0.46). However, significant differences were identified in identical acquisition (p = 0.04) and different acquisition (p = 0.02). Bland-Altman analysis documented only a small bias between PET/MRI and PET + MRI in 11C-PiB PET, 18F-FMM PET, identical additional MRI acquisition, different acquisition, and whole PET (- 0.05, 0.67, - 0.30, 0.78, and 0.21, respectively). CONCLUSIONS Anatomical standardizations using PET/MRI and using PET + MRI can lead to almost equivalent CL. The CL values obtained using PET/MRI or PET + MRI normalization methods are consistent and comparable in clinical studies.
Collapse
Affiliation(s)
- Ryo Yamakuni
- Department of Radiology and Nuclear Medicine, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, 960-1295, Japan.
| | - Takenobu Murakami
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Naoyuki Ukon
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Takeyasu Kakamu
- Department of Hygiene and Preventive Medicine, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Wataru Toda
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Kasumi Hattori
- Department of Neurology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hirofumi Sekino
- Department of Radiology and Nuclear Medicine, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, 960-1295, Japan
| | - Shiro Ishii
- Department of Radiology and Nuclear Medicine, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, 960-1295, Japan
| | - Kenji Fukushima
- Department of Radiology and Nuclear Medicine, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, 960-1295, Japan
| | - Hiroshi Matsuda
- Department of Bio-Functional Imaging, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Noritaka Wakasugi
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Mitsunari Abe
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroshi Ito
- Department of Radiology and Nuclear Medicine, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, 960-1295, Japan
| |
Collapse
|
5
|
Takenaka A, Nihashi T, Sakurai K, Notomi K, Ono H, Inui Y, Ito S, Arahata Y, Takeda A, Ishii K, Ishii K, Ito K, Toyama H, Nakamura A, Kato T. Interrater agreement and variability in visual reading of [18F] flutemetamol PET images. Ann Nucl Med 2024:10.1007/s12149-024-01977-7. [PMID: 39316332 DOI: 10.1007/s12149-024-01977-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE The purpose of this study was to validate the concordance of visual ratings of [18F] flutemetamol amyloid positron emission tomography (PET) images and to investigate the correlation between the agreement of each rater and the Centiloid (CL) scale. METHODS A total of 192 participants, clinically classified as cognitively normal (CN) (n = 59), mild cognitive impairment (MCI) (n = 65), Alzheimer's disease (AD) (n = 55), or non-AD dementia (n = 13), participated in this study. Three experts conducted visual ratings of the amyloid PET images for all 192 patients, assigning a confidence level to each rating on a three-point scale (certain, probable, or neither). The positive or negative determination of amyloid PET results was made by majority vote. The CL value was calculated using the CapAIBL pipeline. RESULTS Overall, 101 images were determined to be positive, and 91 images were negative. Of the 101 positive images, the three raters were in complete agreement for 92 images and in disagreement for 9 images. Of the 91 negative images, the three raters were in complete agreement for 75 images and in disagreement for 16 images. Interrater reliability among the three experts was particularly high, with both Fleiss' kappa and Conger's kappa measuring 0.83 (0.76-0.89). The CL values of the unanimous positive group were significantly greater than those of the other groups, whereas the CL values of the unanimous negative group were significantly lower than those of the other groups. Images with rater disagreement had intermediate CLs. In cases with a high confidence level, the positive or negative visual ratings were in almost complete agreement. However, as confidence levels decreased, experts' visual ratings became more variable. The lower the confidence level was, the greater the number of cases with disagreement in the visual ratings. CONCLUSION Three experts independently rated 192 amyloid PET images, achieving a high level of interrater agreement. However, in patients with intermediate amyloid accumulation, visual ratings varied. Therefore, determining positive and negative decisions in these patients should be performed with caution.
Collapse
Affiliation(s)
- Akinori Takenaka
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Nihashi
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | | | - Hokuto Ono
- Micron Inc. Imaging Service Dept., Tokyo, Japan
| | - Yoshitaka Inui
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Shinji Ito
- Department of Radiology, Anjo Kosei Hospital, Anjo, Japan
| | - Yutaka Arahata
- Department of Neurology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Akinori Takeda
- Department of Neurology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kazunari Ishii
- Department of Radiology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Kenji Ishii
- Team for Neuroimaging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kengo Ito
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Hiroshi Toyama
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Akinori Nakamura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Biomarker Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan.
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan.
| |
Collapse
|
6
|
Gebre RK, Graff-Radford J, Ramanan VK, Raghavan S, Hofrenning EI, Przybelski SA, Nguyen AT, Lesnick TG, Gunter JL, Algeciras-Schimnich A, Knopman DS, Machulda MM, Vassilaki M, Lowe VJ, Jack CR, Petersen RC, Vemuri P. Can integration of Alzheimer's plasma biomarkers with MRI, cardiovascular, genetics, and lifestyle measures improve cognition prediction? Brain Commun 2024; 6:fcae300. [PMID: 39291164 PMCID: PMC11406552 DOI: 10.1093/braincomms/fcae300] [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: 03/19/2024] [Revised: 07/13/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
There is increasing interest in Alzheimer's disease related plasma biomarkers due to their accessibility and scalability. We hypothesized that integrating plasma biomarkers with other commonly used and available participant data (MRI, cardiovascular factors, lifestyle, genetics) using machine learning (ML) models can improve individual prediction of cognitive outcomes. Further, our goal was to evaluate the heterogeneity of these predictors across different age strata. This longitudinal study included 1185 participants from the Mayo Clinic Study of Aging who had complete plasma analyte work-up at baseline. We used the Quanterix Simoa immunoassay to measure neurofilament light, Aβ1-42 and Aβ1-40 (used as Aβ42/Aβ40 ratio), glial fibrillary acidic protein, and phosphorylated tau 181 (p-tau181). Participants' brain health was evaluated through gray and white matter structural MRIs. The study also considered cardiovascular factors (hyperlipidemia, hypertension, stroke, diabetes, chronic kidney disease), lifestyle factors (area deprivation index, body mass index, cognitive and physical activities), and genetic factors (APOE, single nucleotide polymorphisms, and polygenic risk scores). An ML model was developed to predict cognitive outcomes at baseline and decline (slope). Three models were created: a base model with groups of risk factors as predictors, an enhanced model included socio-demographics, and a final enhanced model by incorporating plasma and socio-demographics into the base models. Models were explained for three age strata: younger than 65 years, 65-80 years, and older than 80 years, and further divided based on amyloid positivity status. Regardless of amyloid status the plasma biomarkers showed comparable performance (R² = 0.15) to MRI (R² = 0.18) and cardiovascular measures (R² = 0.10) when predicting cognitive decline. Inclusion of cardiovascular or MRI measures with plasma in the presence of socio-demographic improved cognitive decline prediction (R² = 0.26 and 0.27). For amyloid positive individuals Aβ42/Aβ40, glial fibrillary acidic protein and p-tau181 were the top predictors of cognitive decline while Aβ42/Aβ40 was prominent for amyloid negative participants across all age groups. Socio-demographics explained a large portion of the variance in the amyloid negative individuals while the plasma biomarkers predominantly explained the variance in amyloid positive individuals (21% to 37% from the younger to the older age group). Plasma biomarkers performed similarly to MRI and cardiovascular measures when predicting cognitive outcomes and combining them with either measure resulted in better performance. Top predictors were heterogeneous between cross-sectional and longitudinal cognition models, across age groups, and amyloid status. Multimodal approaches will enhance the usefulness of plasma biomarkers through careful considerations of a study population's socio-demographics, brain and cardiovascular health.
Collapse
Affiliation(s)
- Robel K Gebre
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Vijay K Ramanan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M Machulda
- Department of Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Maria Vassilaki
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
7
|
Leuzy A, Raket LL, Villemagne VL, Klein G, Tonietto M, Olafson E, Baker S, Saad ZS, Bullich S, Lopresti B, Bohorquez SS, Boada M, Betthauser TJ, Charil A, Collins EC, Collins JA, Cullen N, Gunn RN, Higuchi M, Hostetler E, Hutchison RM, Iaccarino L, Insel PS, Irizarry MC, Jack CR, Jagust WJ, Johnson KA, Johnson SC, Karten Y, Marquié M, Mathotaarachchi S, Mintun MA, Ossenkoppele R, Pappas I, Petersen RC, Rabinovici GD, Rosa‐Neto P, Schwarz CG, Smith R, Stephens AW, Whittington A, Carrillo MC, Pontecorvo MJ, Haeberlein SB, Dunn B, Kolb HC, Sivakumaran S, Rowe CC, Hansson O, Doré V. Harmonizing tau positron emission tomography in Alzheimer's disease: The CenTauR scale and the joint propagation model. Alzheimers Dement 2024; 20:5833-5848. [PMID: 39041435 PMCID: PMC11497758 DOI: 10.1002/alz.13908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Tau-positron emission tomography (PET) outcome data of patients with Alzheimer's disease (AD) cannot currently be meaningfully compared or combined when different tracers are used due to differences in tracer properties, instrumentation, and methods of analysis. METHODS Using head-to-head data from five cohorts with tau PET radiotracers designed to target tau deposition in AD, we tested a joint propagation model (JPM) to harmonize quantification (units termed "CenTauR" [CTR]). JPM is a statistical model that simultaneously models the relationships between head-to-head and anchor point data. JPM was compared to a linear regression approach analogous to the one used in the amyloid PET Centiloid scale. RESULTS A strong linear relationship was observed between CTR values across brain regions. Using the JPM approach, CTR estimates were similar to, but more accurate than, those derived using the linear regression approach. DISCUSSION Preliminary findings using the JPM support the development and adoption of a universal scale for tau-PET quantification. HIGHLIGHTS Tested a novel joint propagation model (JPM) to harmonize quantification of tau PET. Units of common scale are termed "CenTauRs". Tested a Centiloid-like linear regression approach. Using five cohorts with head-to-head tau PET, JPM outperformed linearregressionbased approach. Strong linear relationship was observed between CenTauRs values across brain regions.
Collapse
|
8
|
Daniels AJ, McDade E, Llibre-Guerra JJ, Xiong C, Perrin RJ, Ibanez L, Supnet-Bell C, Cruchaga C, Goate A, Renton AE, Benzinger TL, Gordon BA, Hassenstab J, Karch C, Popp B, Levey A, Morris J, Buckles V, Allegri RF, Chrem P, Berman SB, Chhatwal JP, Farlow MR, Fox NC, Day GS, Ikeuchi T, Jucker M, Lee JH, Levin J, Lopera F, Takada L, Sosa AL, Martins R, Mori H, Noble JM, Salloway S, Huey E, Rosa-Neto P, Sánchez-Valle R, Schofield PR, Roh JH, Bateman RJ. 15 Years of Longitudinal Genetic, Clinical, Cognitive, Imaging, and Biochemical Measures in DIAN. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.08.24311689. [PMID: 39148846 PMCID: PMC11326320 DOI: 10.1101/2024.08.08.24311689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
This manuscript describes and summarizes the Dominantly Inherited Alzheimer Network Observational Study (DIAN Obs), highlighting the wealth of longitudinal data, samples, and results from this human cohort study of brain aging and a rare monogenic form of Alzheimer's disease (AD). DIAN Obs is an international collaborative longitudinal study initiated in 2008 with support from the National Institute on Aging (NIA), designed to obtain comprehensive and uniform data on brain biology and function in individuals at risk for autosomal dominant AD (ADAD). ADAD gene mutations in the amyloid protein precursor (APP), presenilin 1 (PSEN1), or presenilin 2 (PSEN2) genes are deterministic causes of ADAD, with virtually full penetrance, and a predictable age at symptomatic onset. Data and specimens collected are derived from full clinical assessments, including neurologic and physical examinations, extensive cognitive batteries, structural and functional neuro-imaging, amyloid and tau pathological measures using positron emission tomography (PET), flurordeoxyglucose (FDG) PET, cerebrospinal fluid and blood collection (plasma, serum, and whole blood), extensive genetic and multi-omic analyses, and brain donation upon death. This comprehensive evaluation of the human nervous system is performed longitudinally in both mutation carriers and family non-carriers, providing one of the deepest and broadest evaluations of the human brain across decades and through AD progression. These extensive data sets and samples are available for researchers to address scientific questions on the human brain, aging, and AD.
Collapse
Affiliation(s)
- Alisha J. Daniels
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Eric McDade
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Chengjie Xiong
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Richard J. Perrin
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Laura Ibanez
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Carlos Cruchaga
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Alison Goate
- Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Alan E. Renton
- Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Brian A. Gordon
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Jason Hassenstab
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Celeste Karch
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Brent Popp
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Allan Levey
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA, USA
| | - John Morris
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | - Virginia Buckles
- Washington University School of Medicine, St Louis, St Louis, MO, USA
| | | | - Patricio Chrem
- Institute of Neurological Research FLENI, Buenos Aires, Argentina
| | | | - Jasmeer P. Chhatwal
- Massachusetts General and Brigham & Women’s Hospitals, Harvard Medical School, Boston MA, USA
| | | | - Nick C. Fox
- UK Dementia Research Institute at University College London, London, United Kingdom
- University College London, London, United Kingdom
| | | | - Takeshi Ikeuchi
- Brain Research Institute, Niigata University, Niigata, Japan
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | | | - Johannes Levin
- DZNE, German Center for Neurodegenerative Diseases, Munich, Germany
- Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | - Ana Luisa Sosa
- Instituto Nacional de Neurologia y Neurocirugla Innn, Mexico City, Mexico
| | - Ralph Martins
- Edith Cowan University, Western Australia, Australia
| | | | - James M. Noble
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Department of Neurology, and GH Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Edward Huey
- Brown University, Butler Hospital, Providence, RI, USA
| | - Pedro Rosa-Neto
- Centre de Recherche de L’hopital Douglas and McGill University, Montreal, Quebec
| | - Raquel Sánchez-Valle
- Hospital Clínic de Barcelona. IDIBAPS. University of Barcelona, Barcelona, Spain
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jee Hoon Roh
- Korea University, Korea University Anam Hospital, Seoul, South Korea
| | | | | |
Collapse
|
9
|
Shekari M, Vállez García D, Collij LE, Altomare D, Heeman F, Pemberton H, Roé Vellvé N, Bullich S, Buckley C, Stephens A, Farrar G, Frisoni G, Klunk WE, Barkhof F, Gispert JD. Stress testing the Centiloid: Precision and variability of PET quantification of amyloid pathology. Alzheimers Dement 2024; 20:5102-5113. [PMID: 38961808 PMCID: PMC11350134 DOI: 10.1002/alz.13883] [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: 10/05/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Assessing the potential sources of bias and variability of the Centiloid (CL) scale is fundamental for its appropriate clinical application. METHODS We included 533 participants from AMYloid imaging to Prevent Alzheimer's Disease (AMYPAD DPMS) and Alzheimer's Disease Neuroimaging Initiative (ADNI) cohorts. Thirty-two CL pipelines were created using different combinations of reference region (RR), RR and target types, and quantification spaces. Generalized estimating equations stratified by amyloid positivity were used to assess the impact of the quantification pipeline, radiotracer, age, brain atrophy, and harmonization status on CL. RESULTS RR selection and RR type impact CL the most, particularly in amyloid-negative individuals. The standard CL pipeline with the whole cerebellum as RR is robust against brain atrophy and differences in image resolution, with 95% confidence intervals below ± 3.95 CL for amyloid beta positivity cutoffs (CL < 24). DISCUSSION The standard CL pipeline is recommended for most scenarios. Confidence intervals should be considered when operationalizing CL cutoffs in clinical and research settings. HIGHLIGHTS We developed a framework for evaluating Centiloid (CL) variability to different factors. Reference region selection and delineation had the highest impact on CL values. Whole cerebellum (WCB) and whole cerebellum plus brainstem (WCB+BSTM) as reference regions yielded consistent results across tracers. The standard CL pipeline is robust against atrophy and image resolution variation. Estimated within- and between-pipeline variability (95% confidence interval) in absolute CL units.
Collapse
Grants
- Janssen Alzheimer Immunotherapy Research & Development, LLC
- 115952 Innovative Medicines Initiative 2 Joint Undertaking
- GE Healthcare
- Transition Therapeutics
- F. Hoffmann-La Roche Ltd
- Eli Lilly and Company
- Eisai Inc.
- W81XWH-12-2-0012 Department of Defense
- EuroImmun
- Biogen
- Alzheimer's Drug Discovery Foundation
- Servier
- Lumosity
- Bristol-Myers Squibb Company
- U01 AG024904 NIA NIH HHS
- Piramal Imaging
- Takeda Pharmaceutical Company
- Alzheimer's Association
- Genentech, Inc.
- Araclon Biotech
- U01 AG024904 NIH HHS
- Meso Scale Diagnostics, LLC
- Novartis Pharmaceuticals Corporation
- CereSpir, Inc.
- BioClinica, Inc.
- Johnson & Johnson Pharmaceutical Research & Development LLC
- AbbVie
- Cogstate
- Merck & Co., Inc.
- NIBIB NIH HHS
- European Union's Horizon 2020
- Pfizer Inc.
- CIHR
- Elan Pharmaceuticals, Inc.
- IXICO Ltd.
- NeuroRx Research
- Neurotrack Technologies
- Fujirebio
- Lundbeck
- National Institutes of Health
- Department of Defense
- National Institute on Aging
- National Institute of Biomedical Imaging and Bioengineering
- AbbVie
- Alzheimer's Association
- Alzheimer's Drug Discovery Foundation
- BioClinica, Inc.
- Biogen
- Bristol‐Myers Squibb Company
- Eli Lilly and Company
- F. Hoffmann‐La Roche Ltd
- Genentech, Inc.
- Fujirebio
- GE Healthcare
- Lundbeck
- Merck & Co., Inc.
- Novartis Pharmaceuticals Corporation
- Pfizer Inc.
- Servier
- Takeda Pharmaceutical Company
- Canadian Institutes of Health Research
Collapse
Affiliation(s)
- Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - David Vállez García
- Department of Radiology and Nuclear MedicineAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamNetherlands
| | - Lyduine E. Collij
- Department of Radiology and Nuclear MedicineAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamNetherlands
- Clinical Memory Research UnitClinical Sciences MalmöLund UniversityMalmöSweden
| | - Daniele Altomare
- Memory CenterDepartment of Rehabilitation and GeriatricsUniversity Hospitals and University of GenevaGenèveSwitzerland
| | - Fiona Heeman
- Department of Radiology and Nuclear MedicineAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamNetherlands
- Wallenberg Centre for Molecular and Translational MedicineUniversity of Gothenburg, The University of GothenburgGothenburgSweden
- Department of Psychiatry and NeurochemistryUniversity of GothenburgSahlgrenska University HospitalGothenburgSweden
| | - Hugh Pemberton
- GE Healthcare Life SciencesAmershamUK
- Institute of Neurology and Centre for Medical Image ComputingUniversity College LondonLondonUK
| | | | | | | | | | | | - Giovanni Frisoni
- Memory CenterDepartment of Rehabilitation and GeriatricsUniversity Hospitals and University of GenevaGenèveSwitzerland
| | | | - Frederik Barkhof
- Department of Radiology and Nuclear MedicineAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamNetherlands
- Institute of Neurology and Centre for Medical Image ComputingUniversity College LondonLondonUK
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, (CIBER‐BBN)MadridSpain
| | | |
Collapse
|
10
|
Bao YW, Wang ZJ, Shea YF, Chiu PKC, Kwan JS, Chan FHW, Mak HKF. Combined quantitative amyloid-β PET and structural MRI features improve Alzheimer's Disease classification in random forest model - A multicenter study. Acad Radiol 2024:S1076-6332(24)00426-4. [PMID: 39003227 DOI: 10.1016/j.acra.2024.06.040] [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: 12/26/2023] [Revised: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
RATIONALE AND OBJECTIVES Prior to clinical presentations of Alzheimer's Disease (AD), neuropathological changes, such as amyloid-β and brain atrophy, have accumulated at the earlier stages of the disease. The combination of such biomarkers assessed by multiple modalities commonly improves the likelihood of AD etiology. We aimed to explore the discriminative ability of Aβ PET features and whether combining Aβ PET and structural MRI features can improve the classification performance of the machine learning model in older healthy control (OHC) and mild cognitive impairment (MCI) from AD. MATERIAL AND METHODS We collected 94 AD patients, 82 MCI patients, and 85 OHC from three different cohorts. 17 global/regional Aβ features in Centiloid, 122 regional volume, and 68 regional cortical thickness were extracted as imaging features. Single or combined modality features were used to train the random forest model on the testing set. The top 10 features were sorted based on the Gini index in each binary classification. RESULTS The results showed that AUC scores were 0.81/0.86 and 0.69/0.68 using sMRI/Aβ PET features on the testing set in differentiating OHC and MCI from AD. The performance was improved while combining two-modality features with an AUC of 0.89 and an AUC of 0.71 in two classifications. Compared to sMRI features, particular Aβ PET features contributed more to differentiating AD from others. CONCLUSION Our study demonstrated the discriminative ability of Aβ PET features in differentiating AD from OHC and MCI. A combination of Aβ PET and structural MRI features can improve the RF model performance.
Collapse
Affiliation(s)
- Yi-Wen Bao
- Department of Medical Imaging Center, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China (Y-W.B.)
| | - Zuo-Jun Wang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China (Z-J.W., H.K-F.M.)
| | - Yat-Fung Shea
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Patrick Ka-Chun Chiu
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Joseph Sk Kwan
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Felix Hon-Wai Chan
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Henry Ka-Fung Mak
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China (Z-J.W., H.K-F.M.).
| |
Collapse
|
11
|
Xiong C, Luo J, Wolk DA, Shaw LM, Roberson ED, Murchison CF, Henson RL, Benzinger TLS, Bui Q, Agboola F, Grant E, Gremminger EN, Moulder KL, Geldmacher DS, Clay OJ, Babulal G, Cruchaga C, Holtzman DM, Bateman RJ, Morris JC, Schindler SE. Baseline levels and longitudinal changes in plasma Aβ42/40 among Black and white individuals. Nat Commun 2024; 15:5539. [PMID: 38956096 PMCID: PMC11219932 DOI: 10.1038/s41467-024-49859-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
Blood-based biomarkers of Alzheimer disease (AD) may facilitate testing of historically under-represented groups. The Study of Race to Understand Alzheimer Biomarkers (SORTOUT-AB) is a multi-center longitudinal study to compare AD biomarkers in participants who identify their race as either Black or white. Plasma samples from 324 Black and 1,547 white participants underwent analysis with C2N Diagnostics' PrecivityAD test for Aβ42 and Aβ40. Compared to white individuals, Black individuals had higher average plasma Aβ42/40 levels at baseline, consistent with a lower average level of amyloid pathology. Interestingly, this difference resulted from lower average levels of plasma Aβ40 in Black participants. Despite the differences, Black and white individuals had similar longitudinal rates of change in Aβ42/40, consistent with a similar rate of amyloid accumulation. Our results agree with multiple recent studies demonstrating a lower prevalence of amyloid pathology in Black individuals, and additionally suggest that amyloid accumulates consistently across both groups.
Collapse
Affiliation(s)
- Chengjie Xiong
- Division of Biostatistics, Washington University, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center Biostatistics and Qualitative Research Shared Resource, Washington University School of Medicine, St. Louis, MO, USA
| | - David A Wolk
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leslie M Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erik D Roberson
- Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles F Murchison
- Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rachel L Henson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Quoc Bui
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - Folasade Agboola
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - Elizabeth Grant
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | | | - Krista L Moulder
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David S Geldmacher
- Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Olivio J Clay
- Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ganesh Babulal
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
12
|
Kumar S, Earnest T, Yang B, Kothapalli D, Aschenbrenner AJ, Hassenstab J, Xiong C, Ances B, Morris J, Benzinger TLS, Gordon BA, Payne P, Sotiras A. Analyzing heterogeneity in Alzheimer Disease using multimodal normative modeling on imaging-based ATN biomarkers. ARXIV 2024:arXiv:2404.05748v2. [PMID: 39010871 PMCID: PMC11247918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
INTRODUCTION Previous studies have applied normative modeling on a single neuroimaging modality to investigate Alzheimer Disease (AD) heterogeneity. We employed a deep learning-based multimodal normative framework to analyze individual-level variation across ATN (amyloid-tau-neurodegeneration) imaging biomarkers. METHODS We selected cross-sectional discovery (n = 665) and replication cohorts (n = 430) with available T1-weighted MRI, amyloid and tau PET. Normative modeling estimated individual-level abnormal deviations in amyloid-positive individuals compared to amyloid-negative controls. Regional abnormality patterns were mapped at different clinical group levels to assess intra-group heterogeneity. An individual-level disease severity index (DSI) was calculated using both the spatial extent and magnitude of abnormal deviations across ATN. RESULTS Greater intra-group heterogeneity in ATN abnormality patterns was observed in more severe clinical stages of AD. Higher DSI was associated with worse cognitive function and increased risk of disease progression. DISCUSSION Subject-specific abnormality maps across ATN reveal the heterogeneous impact of AD on the brain.
Collapse
Affiliation(s)
- Sayantan Kumar
- Department of Computer Science and Engineering, Washington University in St Louis; 1 Brookings Drive, Saint Louis, MO 63130
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Tom Earnest
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Braden Yang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Deydeep Kothapalli
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Andrew J. Aschenbrenner
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Chengie Xiong
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Beau Ances
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - John Morris
- Department of Neurology, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8111, St louis, MO 63110
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Brian A. Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| | - Philip Payne
- Department of Computer Science and Engineering, Washington University in St Louis; 1 Brookings Drive, Saint Louis, MO 63130
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
| | - Aristeidis Sotiras
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine in St Louis; 660 S. Euclid Ave, Campus Box 8132, Saint Louis, MO 63110
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis; 4525 Scott Ave, Saint Louis, MO 63110
| |
Collapse
|
13
|
Babulal GM, Chen L, Murphy SA, Carr DB, Morris JC. Predicting Driving Cessation Among Cognitively Normal Older Drivers: The Role of Alzheimer Disease Biomarkers and Clinical Assessments. Neurology 2024; 102:e209426. [PMID: 38776513 PMCID: PMC11226325 DOI: 10.1212/wnl.0000000000209426] [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: 01/04/2024] [Accepted: 03/11/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES With the aging US population and increasing incidence of Alzheimer disease (AD), understanding factors contributing to driving cessation among older adults is crucial for clinicians. Driving is integral for maintaining independence and functional mobility, but the risk factors for driving cessation, particularly in the context of normal aging and preclinical AD, are not well understood. We studied a well-characterized community cohort to examine factors associated with driving cessation. METHODS This prospective, longitudinal observation study enrolled participants from the Knight Alzheimer Disease Research Center and The DRIVES Project. Participants were enrolled if they were aged 65 years or older, drove weekly, and were cognitively normal (Clinical Dementia Rating [CDR] = 0) at baseline. Participants underwent annual clinical, neurologic, and neuropsychological assessments, including β-amyloid PET imaging and CSF (Aβ42, total tau [t-Tau], and phosphorylated tau [p-Tau]) collection every 2-3 years. The primary outcome was time from baseline visit to driving cessation, accounting for death as a competing risk. The cumulative incidence function of driving cessation was estimated for each biomarker. The Fine and Gray subdistribution hazard model was used to examine the association between time to driving cessation and biomarkers adjusting for clinical and demographic covariates. RESULTS Among the 283 participants included in this study, there was a mean follow-up of 5.62 years. Driving cessation (8%) was associated with older age, female sex, progression to symptomatic AD (CDR ≥0.5), and poorer performance on a preclinical Alzheimer cognitive composite (PACC) score. Aβ PET imaging did not independently predict driving cessation, whereas CSF biomarkers, specifically t-Tau/Aβ42 (hazard ratio [HR] 2.82, 95% CI 1.23-6.44, p = 0.014) and p-Tau/Aβ42 (HR 2.91, 95% CI 1.28-6.59, p = 0.012) ratios, were independent predictors in the simple model adjusting for age, education, and sex. However, in the full model, progression to cognitive impairment based on the CDR and PACC score across each model was associated with a higher risk of driving cessation, whereas AD biomarkers were not statistically significant. DISCUSSION Female sex, CDR progression, and neuropsychological measures of cognitive functioning obtained in the clinic were strongly associated with future driving cessation. The results emphasize the need for early planning and conversations about driving retirement in the context of cognitive decline and the immense value of clinical measures in determining functional outcomes.
Collapse
Affiliation(s)
- Ganesh M Babulal
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - Ling Chen
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - Samantha A Murphy
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - David B Carr
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - John C Morris
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
14
|
Jadick MF, Robinson T, Farrell ME, Klinger H, Buckley RF, Marshall GA, Vannini P, Rentz DM, Johnson KA, Sperling RA, Amariglio RE. Associations Between Self and Study Partner Report of Cognitive Decline With Regional Tau in a Multicohort Study. Neurology 2024; 102:e209447. [PMID: 38810211 PMCID: PMC11226320 DOI: 10.1212/wnl.0000000000209447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/04/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Self-reported cognitive decline is an early behavioral manifestation of Alzheimer disease (AD) at the preclinical stage, often believed to precede concerns reported by a study partner. Previous work shows cross-sectional associations with β-amyloid (Aβ) status and self-reported and study partner-reported cognitive decline, but less is known about their associations with tau deposition, particularly among those with preclinical AD. METHODS This cross-sectional study included participants from the Anti-Amyloid Treatment in Asymptomatic AD/Longitudinal Evaluation of Amyloid Risk and Neurodegeneration studies (N = 444) and the Harvard Aging Brain Study and affiliated studies (N = 231), which resulted in a cognitively unimpaired (CU) sample of individuals with both nonelevated (Aβ-) and elevated Aβ (Aβ+). All participants and study partners completed the Cognitive Function Index (CFI). Two regional tau composites were derived by averaging flortaucipir PET uptake in the medial temporal lobe (MTL) and neocortex (NEO). Global Aβ PET was measured in Centiloids (CLs) with Aβ+ >26 CL. We conducted multiple linear regression analyses to test associations between tau PET and CFI, covarying for amyloid, age, sex, education, and cohort. We also controlled for objective cognitive performance, measured using the Preclinical Alzheimer Cognitive Composite (PACC). RESULTS Across 675 CU participants (age = 72.3 ± 6.6 years, female = 59%, Aβ+ = 60%), greater tau was associated with greater self-CFI (MTL: β = 0.28 [0.12, 0.44], p < 0.001, and NEO: β = 0.26 [0.09, 0.42], p = 0.002) and study partner CFI (MTL: β = 0.28 [0.14, 0.41], p < 0.001, and NEO: β = 0.31 [0.17, 0.44], p < 0.001). Significant associations between both CFI measures and MTL/NEO tau PET were driven by Aβ+. Continuous Aβ showed an independent effect on CFI in addition to MTL and NEO tau for both self-CFI and study partner CFI. Self-CFI (β = 0.01 [0.001, 0.02], p = 0.03), study partner CFI (β = 0.01 [0.003, 0.02], p = 0.01), and the PACC (β = -0.02 [-0.03, -0.01], p < 0.001) were independently associated with MTL tau, but for NEO tau, PACC (β = -0.02 [-0.03, -0.01], p < 0.001) and study partner report (β = 0.01 [0.004, 0.02], p = 0.002) were associated, but not self-CFI (β = 0.01 [-0.001, 0.02], p = 0.10). DISCUSSION Both self-report and study partner report showed associations with tau in addition to Aβ. Additionally, self-report and study partner report were associated with tau above and beyond performance on a neuropsychological composite. Stratification analyses by Aβ status indicate that associations between self-reported and study partner-reported cognitive concerns with regional tau are driven by those at the preclinical stage of AD, suggesting that both are useful to collect on the early AD continuum.
Collapse
Affiliation(s)
- Michalina F Jadick
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Talia Robinson
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Michelle E Farrell
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hannah Klinger
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rachel F Buckley
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Gad A Marshall
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Patrizia Vannini
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dorene M Rentz
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Keith A Johnson
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Reisa A Sperling
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rebecca E Amariglio
- From the Department of Neurology (M.F.J., H.K., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), and Department of Radiology (M.F.J., K.A.J.), Massachusetts General Hospital, and Center for Alzheimer Research and Treatment (T.R., M.E.F., R.F.B., G.A.M., P.V., D.M.R., K.A.J., R.A.S., R.E.A.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Earnest T, Bani A, Ha SM, Hobbs DA, Kothapalli D, Yang B, Lee JJ, Benzinger TLS, Gordon BA, Sotiras A. Data-driven decomposition and staging of flortaucipir uptake in Alzheimer's disease. Alzheimers Dement 2024; 20:4002-4019. [PMID: 38683905 PMCID: PMC11180875 DOI: 10.1002/alz.13769] [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: 10/10/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Previous approaches pursuing in vivo staging of tau pathology in Alzheimer's disease (AD) have typically relied on neuropathologically defined criteria. In using predefined systems, these studies may miss spatial deposition patterns which are informative of disease progression. METHODS We selected discovery (n = 418) and replication (n = 132) cohorts with flortaucipir imaging. Non-negative matrix factorization (NMF) was applied to learn tau covariance patterns and develop a tau staging system. Flortaucipir components were also validated by comparison with amyloid burden, gray matter loss, and the expression of AD-related genes. RESULTS We found eight flortaucipir covariance patterns which were reproducible and overlapped with relevant gene expression maps. Tau stages were associated with AD severity as indexed by dementia status and neuropsychological performance. Comparisons of flortaucipir uptake with amyloid and atrophy also supported our model of tau progression. DISCUSSION Data-driven decomposition of flortaucipir uptake provides a novel framework for tau staging which complements existing systems. HIGHLIGHTS NMF reveals patterns of tau deposition in AD. Data-driven staging of flortaucipir tracks AD severity. Learned flortaucipir patterns overlap with AD-related gene expression.
Collapse
Affiliation(s)
- Tom Earnest
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Abdalla Bani
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Sung Min Ha
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Diana A. Hobbs
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Deydeep Kothapalli
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Braden Yang
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - John J. Lee
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Brian A. Gordon
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | - Aristeidis Sotiras
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St LouisSaint LouisMissouriUSA
- Institute for Informatics, Data Science & BiostatisticsWashington University School of Medicine in St LouisSaint LouisMissouriUSA
| | | |
Collapse
|
16
|
Jagust WJ, Mattay VS, Krainak DM, Wang SJ, Weidner LD, Hofling AA, Koo H, Hsieh P, Kuo PH, Farrar G, Marzella L. Quantitative Brain Amyloid PET. J Nucl Med 2024; 65:670-678. [PMID: 38514082 PMCID: PMC11064834 DOI: 10.2967/jnumed.123.265766] [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: 09/19/2023] [Revised: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Since the development of amyloid tracers for PET imaging, there has been interest in quantifying amyloid burden in the brains of patients with Alzheimer disease. Quantitative amyloid PET imaging is poised to become a valuable approach in disease staging, theranostics, monitoring, and as an outcome measure for interventional studies. Yet, there are significant challenges and hurdles to overcome before it can be implemented into widespread clinical practice. On November 17, 2022, the U.S. Food and Drug Administration, Society of Nuclear Medicine and Molecular Imaging, and Medical Imaging and Technology Alliance cosponsored a public workshop comprising experts from academia, industry, and government agencies to discuss the role of quantitative brain amyloid PET imaging in staging, prognosis, and longitudinal assessment of Alzheimer disease. The workshop discussed a range of topics, including available radiopharmaceuticals for amyloid imaging; the methodology, metrics, and analytic validity of quantitative amyloid PET imaging; its use in disease staging, prognosis, and monitoring of progression; and challenges facing the field. This report provides a high-level summary of the presentations and the discussion.
Collapse
Affiliation(s)
| | - Venkata S Mattay
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland;
| | - Daniel M Krainak
- Division of Radiological Imaging and Radiation Therapy Devices, Office of Radiological Health, Office of Product Evaluation and Quality, Centers for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland
| | - Sue-Jane Wang
- Division of Biometrics I, Office of Biostatistics, Office of Translational Sciences, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Lora D Weidner
- Division of Radiological Imaging and Radiation Therapy Devices, Office of Radiological Health, Office of Product Evaluation and Quality, Centers for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland
| | - A Alex Hofling
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | - Hayoung Koo
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| | | | | | | | - Libero Marzella
- Division of Imaging and Radiation Medicine, Office of Specialty Medicine, Office of New Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
17
|
Li Y, Yen D, Hendrix RD, Gordon BA, Dlamini S, Barthélemy NR, Aschenbrenner AJ, Henson RL, Herries EM, Volluz K, Kirmess K, Eastwood S, Meyer M, Heller M, Jarrett L, McDade E, Holtzman DM, Benzinger TL, Morris JC, Bateman RJ, Xiong C, Schindler SE. Timing of Biomarker Changes in Sporadic Alzheimer's Disease in Estimated Years from Symptom Onset. Ann Neurol 2024; 95:951-965. [PMID: 38400792 PMCID: PMC11060905 DOI: 10.1002/ana.26891] [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: 08/30/2023] [Revised: 12/26/2023] [Accepted: 01/30/2024] [Indexed: 02/26/2024]
Abstract
OBJECTIVE A clock relating amyloid positron emission tomography (PET) to time was used to estimate the timing of biomarker changes in sporadic Alzheimer disease (AD). METHODS Research participants were included who underwent cerebrospinal fluid (CSF) collection within 2 years of amyloid PET. The ages at amyloid onset and AD symptom onset were estimated for each individual. The timing of change for plasma, CSF, imaging, and cognitive measures was calculated by comparing restricted cubic splines of cross-sectional data from the amyloid PET positive and negative groups. RESULTS The amyloid PET positive sub-cohort (n = 118) had an average age of 70.4 ± 7.4 years (mean ± standard deviation) and 16% were cognitively impaired. The amyloid PET negative sub-cohort (n = 277) included individuals with low levels of amyloid plaque burden at all scans who were cognitively unimpaired at the time of the scans. Biomarker changes were detected 15-19 years before estimated symptom onset for CSF Aβ42/Aβ40, plasma Aβ42/Aβ40, CSF pT217/T217, and amyloid PET; 12-14 years before estimated symptom onset for plasma pT217/T217, CSF neurogranin, CSF SNAP-25, CSF sTREM2, plasma GFAP, and plasma NfL; and 7-9 years before estimated symptom onset for CSF pT205/T205, CSF YKL-40, hippocampal volumes, and cognitive measures. INTERPRETATION The use of an amyloid clock enabled visualization and analysis of biomarker changes as a function of estimated years from symptom onset in sporadic AD. This study demonstrates that estimated years from symptom onset based on an amyloid clock can be used as a continuous staging measure for sporadic AD and aligns with findings in autosomal dominant AD. ANN NEUROL 2024;95:951-965.
Collapse
Affiliation(s)
- Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel Yen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel D. Hendrix
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A. Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sibonginkhosi Dlamini
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicolas R. Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Rachel L. Henson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth M. Herries
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine Volluz
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | - Maren Heller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lea Jarrett
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric McDade
- 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
| | - David M. Holtzman
- 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
| | - Tammie L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- 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
| | - 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
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - 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
| |
Collapse
|
18
|
Bollack A, Collij LE, García DV, Shekari M, Altomare D, Payoux P, Dubois B, Grau‐Rivera O, Boada M, Marquié M, Nordberg A, Walker Z, Scheltens P, Schöll M, Wolz R, Schott JM, Gismondi R, Stephens A, Buckley C, Frisoni GB, Hanseeuw B, Visser PJ, Vandenberghe R, Drzezga A, Yaqub M, Boellaard R, Gispert JD, Markiewicz P, Cash DM, Farrar G, Barkhof F. Investigating reliable amyloid accumulation in Centiloids: Results from the AMYPAD Prognostic and Natural History Study. Alzheimers Dement 2024; 20:3429-3441. [PMID: 38574374 PMCID: PMC11095430 DOI: 10.1002/alz.13761] [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: 10/09/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 04/06/2024]
Abstract
INTRODUCTION To support clinical trial designs focused on early interventions, our study determined reliable early amyloid-β (Aβ) accumulation based on Centiloids (CL) in pre-dementia populations. METHODS A total of 1032 participants from the Amyloid Imaging to Prevent Alzheimer's Disease-Prognostic and Natural History Study (AMYPAD-PNHS) and Insight46 who underwent [18F]flutemetamol, [18F]florbetaben or [18F]florbetapir amyloid-PET were included. A normative strategy was used to define reliable accumulation by estimating the 95th percentile of longitudinal measurements in sub-populations (NPNHS = 101/750, NInsight46 = 35/382) expected to remain stable over time. The baseline CL threshold that optimally predicts future accumulation was investigated using precision-recall analyses. Accumulation rates were examined using linear mixed-effect models. RESULTS Reliable accumulation in the PNHS was estimated to occur at >3.0 CL/year. Baseline CL of 16 [12,19] best predicted future Aβ-accumulators. Rates of amyloid accumulation were tracer-independent, lower for APOE ε4 non-carriers, and for subjects with higher levels of education. DISCUSSION Our results support a 12-20 CL window for inclusion into early secondary prevention studies. Reliable accumulation definition warrants further investigations.
Collapse
Affiliation(s)
- Ariane Bollack
- Centre for Medical Image Computing (CMIC)Department of Medical Physics and BioengineeringUniversity College LondonLondonLondonUK
| | - Lyduine E. Collij
- Department of Radiology and Nuclear MedicineAmsterdam UMCAmsterdamThe Netherlands
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Amsterdam Neuroscience, Brain ImagingVU University AmsterdamAmsterdamThe Netherlands
| | - David Vállez García
- Department of Radiology and Nuclear MedicineAmsterdam UMCAmsterdamThe Netherlands
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- Instituto de investigaciones médicas Hospital del Mar (IMIM)BarcelonaSpain
| | - Daniele Altomare
- Neurology UnitDepartment of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Pierre Payoux
- Department of Nuclear MedicineImaging PoleToulouse University HospitalToulouseFrance
- Toulouse NeuroImaging CenterUniversité de ToulouseInsermUPSCHU PurpanPavillon BaudotPlace du Docteur Joseph BaylacToulouseFrance
| | - Bruno Dubois
- Department of NeurologySalpêtrière HospitalAP‐HPSorbonne UniversityParisFrance
| | - Oriol Grau‐Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona – Universitat Internacional de CatalunyaBarcelonaSpain
- CIBERNEDNetwork Center for Biomedical Research in Neurodegenerative DiseasesNational Institute of Health Carlos IIIMadridSpain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona – Universitat Internacional de CatalunyaBarcelonaSpain
- CIBERNEDNetwork Center for Biomedical Research in Neurodegenerative DiseasesNational Institute of Health Carlos IIIMadridSpain
| | - Agneta Nordberg
- Department of NeurobiologyCare Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska InstitutetStockholmSweden
- Theme Inflammation and Aging, Karolinska University Hospital, Karolinska InstitutetStockholmSweden
| | - Zuzana Walker
- Division of PsychiatryUniversity College LondonLondonUK
- Essex Partnership University NHS Foundation Trust, The LodgeWickfordUK
| | - Philip Scheltens
- Alzheimer Center and Department of NeurologyAmsterdam Neuroscience, VU University Medical Center, Alzheimercentrum AmsterdamAmsterdamThe Netherlands
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, The University of GothenburgGothenburgSweden
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University HospitalGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | | | - Jonathan M. Schott
- Dementia Research Centre, UCL Queen Square Institute of NeurologyLondonUK
| | | | | | | | - Giovanni B. Frisoni
- Neurology UnitDepartment of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Bernard Hanseeuw
- Department of NeurologyInstitute of Neuroscience, Université Catholique de Louvain, Cliniques Universitaires Saint‐LucBrusselsBelgium
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General HospitalBostonMassachusettsUSA
- WELBIO DepartmentWEL Research InstituteWavreBelgium
| | - Pieter Jelle Visser
- Department of Radiology and Nuclear MedicineAmsterdam UMCAmsterdamThe Netherlands
- Department of NeurobiologyCare Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska InstitutetStockholmSweden
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht UniversityMaastrichtThe Netherlands
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, LBI – KU Leuven Brain InstituteLeuvenBelgium
| | - Alexander Drzezga
- Department of Nuclear MedicineUniversity Hospital Cologne, Universitätsklinikums KölnKölnGermany
- Molecular Organization of the Brain, Institute for Neuroscience and Medicine, INM‐2), Forschungszentrum Jülich GmbHJülichGermany
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Maqsood Yaqub
- Department of Radiology and Nuclear MedicineAmsterdam UMCAmsterdamThe Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear MedicineAmsterdam UMCAmsterdamThe Netherlands
- Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos IIIMadridSpain
| | - Pawel Markiewicz
- Centre for Medical Image Computing (CMIC)Department of Medical Physics and BioengineeringUniversity College LondonLondonLondonUK
- Computer Science and Informatics, School of Engineering, London South Bank UniversityLondonUK
| | - David M. Cash
- Queen Square Institute of Neurology, University College LondonLondonUK
- UK Dementia Research Institute at University College LondonLondonUK
| | | | - Frederik Barkhof
- Centre for Medical Image Computing (CMIC)Department of Medical Physics and BioengineeringUniversity College LondonLondonLondonUK
- Department of Radiology and Nuclear MedicineAmsterdam UMCAmsterdamThe Netherlands
- Queen Square Institute of Neurology, University College LondonLondonUK
| | | |
Collapse
|
19
|
Bao YW, Wang ZJ, Guo LL, Bai GJ, Feng Y, Zhao GD. Expression of regional brain amyloid-β deposition with [18F]Flutemetamol in Centiloid scale -a multi-site study. Neuroradiology 2024:10.1007/s00234-024-03364-5. [PMID: 38676749 DOI: 10.1007/s00234-024-03364-5] [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: 12/12/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
PURPOSE The Centiloid project helps calibrate the quantitative amyloid-β (Aβ) load into a unified Centiloid (CL) scale that allows data comparison across multi-site. How the smaller regional amyloid converted into CL has not been attempted. We first aimed to express regional Aβ deposition in CL using [18F]Flutemetamol and evaluate regional Aβ deposition in CL with that in standardized uptake value ratio (SUVr). Second, we aimed to determine the presence or absence of focal Aβ deposition by measuring regional CL in equivocal cases showing negative global CL. METHODS Following the Centiloid project pipeline, Level-1 replication, Level-2 calibration, and quality control were completed to generate corresponding Centiloid conversion equations to convert SUVr into Centiloid at regional levels. In equivocal cases, the regional CL was compared with visual inspection to evaluate regional Aβ positivity. RESULTS 14 out of 16 regional conversions from [18F]Flutemetamol SUVr to Centiloid successfully passed the quality control, showing good reliability and relative variance, especially precuneus/posterior cingulate and prefrontal regions with good stability for Centiloid scaling. The absence of focal Aβ deposition could be detected by measuring regional CL, showing a high agreement rate with visual inspection. The regional Aβ positivity in the bilateral anterior cingulate cortex was most prevalent in equivocal cases. CONCLUSION The expression of regional brain Aβ deposition in CL with [18F]Flutemetamol has been attempted in this study. Equivocal cases had focal Aβ deposition that can be detected by measuring regional CL.
Collapse
Affiliation(s)
- Yi-Wen Bao
- Department of Medical Imaging Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 1 Huanghe West Road, Huaiyin District, 223300, Huai'an, Jiangsu, China.
| | - Zuo-Jun Wang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Li-Li Guo
- Department of Medical Imaging Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 1 Huanghe West Road, Huaiyin District, 223300, Huai'an, Jiangsu, China
| | - Gen-Ji Bai
- Department of Medical Imaging Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 1 Huanghe West Road, Huaiyin District, 223300, Huai'an, Jiangsu, China
| | - Yun Feng
- Department of Medical Imaging Center, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 1 Huanghe West Road, Huaiyin District, 223300, Huai'an, Jiangsu, China
| | - Guo-Dong Zhao
- Department of General Surgery, Lianshui County People's Hospital, 223400, Huai'an, Jiang Su, China
| |
Collapse
|
20
|
Meeker KL, Luckett PH, Barthélemy NR, Hobbs DA, Chen C, Bollinger J, Ovod V, Flores S, Keefe S, Henson RL, Herries EM, McDade E, Hassenstab JJ, Xiong C, Cruchaga C, Benzinger TLS, Holtzman DM, Schindler SE, Bateman RJ, Morris JC, Gordon BA, Ances BM. Comparison of cerebrospinal fluid, plasma and neuroimaging biomarker utility in Alzheimer's disease. Brain Commun 2024; 6:fcae081. [PMID: 38505230 PMCID: PMC10950051 DOI: 10.1093/braincomms/fcae081] [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: 10/05/2023] [Revised: 02/01/2024] [Accepted: 03/14/2024] [Indexed: 03/21/2024] Open
Abstract
Alzheimer's disease biomarkers are crucial to understanding disease pathophysiology, aiding accurate diagnosis and identifying target treatments. Although the number of biomarkers continues to grow, the relative utility and uniqueness of each is poorly understood as prior work has typically calculated serial pairwise relationships on only a handful of markers at a time. The present study assessed the cross-sectional relationships among 27 Alzheimer's disease biomarkers simultaneously and determined their ability to predict meaningful clinical outcomes using machine learning. Data were obtained from 527 community-dwelling volunteers enrolled in studies at the Charles F. and Joanne Knight Alzheimer Disease Research Center at Washington University in St Louis. We used hierarchical clustering to group 27 imaging, CSF and plasma measures of amyloid beta, tau [phosphorylated tau (p-tau), total tau t-tau)], neuronal injury and inflammation drawn from MRI, PET, mass-spectrometry assays and immunoassays. Neuropsychological and genetic measures were also included. Random forest-based feature selection identified the strongest predictors of amyloid PET positivity across the entire cohort. Models also predicted cognitive impairment across the entire cohort and in amyloid PET-positive individuals. Four clusters emerged reflecting: core Alzheimer's disease pathology (amyloid and tau), neurodegeneration, AT8 antibody-associated phosphorylated tau sites and neuronal dysfunction. In the entire cohort, CSF p-tau181/Aβ40lumi and Aβ42/Aβ40lumi and mass spectrometry measurements for CSF pT217/T217, pT111/T111, pT231/T231 were the strongest predictors of amyloid PET status. Given their ability to denote individuals on an Alzheimer's disease pathological trajectory, these same markers (CSF pT217/T217, pT111/T111, p-tau/Aβ40lumi and t-tau/Aβ40lumi) were largely the best predictors of worse cognition in the entire cohort. When restricting analyses to amyloid-positive individuals, the strongest predictors of impaired cognition were tau PET, CSF t-tau/Aβ40lumi, p-tau181/Aβ40lumi, CSF pT217/217 and pT205/T205. Non-specific CSF measures of neuronal dysfunction and inflammation were poor predictors of amyloid PET and cognitive status. The current work utilized machine learning to understand the interrelationship structure and utility of a large number of biomarkers. The results demonstrate that, although the number of biomarkers has rapidly expanded, many are interrelated and few strongly predict clinical outcomes. Examining the entire corpus of available biomarkers simultaneously provides a meaningful framework to understand Alzheimer's disease pathobiological change as well as insight into which biomarkers may be most useful in Alzheimer's disease clinical practice and trials.
Collapse
Affiliation(s)
- Karin L Meeker
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Patrick H Luckett
- Department of Neurosurgery, Washington University in St Louis, St Louis, MO 63110, USA
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Diana A Hobbs
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Charles Chen
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - James Bollinger
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Vitaliy Ovod
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Shaney Flores
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Sarah Keefe
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Rachel L Henson
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Elizabeth M Herries
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Jason J Hassenstab
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63110, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Brian A Gordon
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Beau M Ances
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
- Department of Radiology, Washington University in St Louis, St Louis, MO 63110, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
21
|
Bonomi S, Lu R, Schindler SE, Bui Q, Lah JJ, Wolk D, Gleason CE, Sperling R, Roberson ED, Levey AI, Shaw L, Van Hulle C, Benzinger T, Adams M, Manzanares C, Qiu D, Hassenstab J, Moulder KL, Balls-Berry JE, Johnson K, Johnson SC, Murchison CF, Luo J, Gremminger E, Agboola F, Grant EA, Hornbeck R, Massoumzadeh P, Keefe S, Dierker D, Gray JD, Henson RL, Streitz M, Mechanic-Hamilton D, Morris JC, Xiong C. Relationships of Cognitive Measures with Cerebrospinal Fluid but Not Imaging Biomarkers of Alzheimer Disease Vary between Black and White Individuals. Ann Neurol 2024; 95:495-506. [PMID: 38038976 PMCID: PMC10922199 DOI: 10.1002/ana.26838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023]
Abstract
OBJECTIVE Biomarkers of Alzheimer disease vary between groups of self-identified Black and White individuals in some studies. This study examined whether the relationships between biomarkers or between biomarkers and cognitive measures varied by racialized groups. METHODS Cerebrospinal fluid (CSF), amyloid positron emission tomography (PET), and magnetic resonance imaging measures were harmonized across four studies of memory and aging. Spearman correlations between biomarkers and between biomarkers and cognitive measures were calculated within each racialized group, then compared between groups by standard normal tests after Fisher's Z-transformations. RESULTS The harmonized dataset included at least one biomarker measurement from 495 Black and 2,600 White participants. The mean age was similar between racialized groups. However, Black participants were less likely to have cognitive impairment (28% vs 36%) and had less abnormality of some CSF biomarkers including CSF Aβ42/40, total tau, p-tau181, and neurofilament light. CSF Aβ42/40 was negatively correlated with total tau and p-tau181 in both groups, but at a smaller magnitude in Black individuals. CSF Aβ42/40, total tau, and p-tau181 had weaker correlations with cognitive measures, especially episodic memory, in Black than White participants. Correlations of amyloid measures between CSF (Aβ42/40, Aβ42) and PET imaging were also weaker in Black than White participants. Importantly, no differences based on race were found in correlations between different imaging biomarkers, or in correlations between imaging biomarkers and cognitive measures. INTERPRETATION Relationships between CSF biomarkers but not imaging biomarkers varied by racialized groups. Imaging biomarkers performed more consistently across racialized groups in associations with cognitive measures. ANN NEUROL 2024;95:495-506.
Collapse
Affiliation(s)
- Samuele Bonomi
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Ruijin Lu
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Quoc Bui
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - James J. Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA
| | - David Wolk
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carey E. Gleason
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer’s Disease Research Center, Madison, Wisconsin, USA
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Reisa Sperling
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Erik D. Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer’s Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Allan I. Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA
| | - Leslie Shaw
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carol Van Hulle
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer’s Disease Research Center, Madison, Wisconsin, USA
| | - Tammie Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Morgann Adams
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Cecelia Manzanares
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA
| | - Deqiang Qiu
- Goizueta Alzheimer’s Disease Research Center, Emory University, Atlanta, GA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Krista L. Moulder
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Joyce E. Balls-Berry
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sterling C. Johnson
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Wisconsin Alzheimer’s Disease Research Center, Madison, Wisconsin, USA
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Charles F. Murchison
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer’s Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jingqin Luo
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Emily Gremminger
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Folasade Agboola
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Elizabeth A. Grant
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Russ Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Parinaz Massoumzadeh
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Sarah Keefe
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Donna Dierker
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Julia D. Gray
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Rachel L. Henson
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Marissa Streitz
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Dawn Mechanic-Hamilton
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| |
Collapse
|
22
|
Chen K, Ghisays V, Luo J, Chen Y, Lee W, Wu T, Reiman EM, Su Y. Harmonizing florbetapir and PiB PET measurements of cortical Aβ plaque burden using multiple regions-of-interest and machine learning techniques: An alternative to the Centiloid approach. Alzheimers Dement 2024; 20:2165-2172. [PMID: 38276892 PMCID: PMC10984485 DOI: 10.1002/alz.13677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024]
Abstract
INTRODUCTION Machine learning (ML) can optimize amyloid (Aβ) comparability among positron emission tomography (PET) radiotracers. Using multi-regional florbetapir (FBP) measures and ML, we report better Pittsburgh compound-B (PiB)/FBP harmonization of mean-cortical Aβ (mcAβ) than Centiloid. METHODS PiB-FBP pairs from 92 subjects in www.oasis-brains.org and 46 in www.gaain.org/centiloid-project were used as the training/testing sets. FreeSurfer-extracted FBP multi-regional Aβ and actual PiB mcAβ in the training set were used to train ML models generating synthetic PiB mcAβ. The correlation coefficient (R) between the synthetic/actual PiB mcAβ in the testing set was assessed. RESULTS In the testing set, the synthetic/actual PiB mcAβ correlation R = 0.985 (R2 = 0.970) using artificial neural network was significantly higher (p ≤ 6.6e-4) than the FBP/PiB correlation R = 0.927 (R2 = 0.860), improving total variance percentage (R2 ) from 86% to 97%. Other ML models such as partial least square, ensemble, and relevance vector regressions also improved R (p = 9.677e-05 /0.045/0.0017). DISCUSSION ML improved mcAβ comparability. Additional studies are needed for the generalizability to other amyloid tracers, and to tau PET. Highlights Centiloid is a calibration of the amyloid scale, not harmonization. Centiloid unifies the amyloid scale without improving inter-tracer association (R2 ). Machine learning (ML) can harmonize the amyloid scale by improving R2 . ML harmonization maps multi-regional florbetapir SUVRs to PiB mean-cortical SUVR. Artificial neural network ML increases Centiloid R2 from 86% to 97%.
Collapse
Affiliation(s)
- Kewei Chen
- Banner Alzheimer's InstitutePhoenixArizonaUSA
- Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
- School of Mathematics and Statistical SciencesCollege of Health SolutionsArizona State UniversityTempeArizonaUSA
- Department of Neurology College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Valentina Ghisays
- Banner Alzheimer's InstitutePhoenixArizonaUSA
- Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Ji Luo
- Banner Alzheimer's InstitutePhoenixArizonaUSA
- Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Yinghua Chen
- Banner Alzheimer's InstitutePhoenixArizonaUSA
- Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Wendy Lee
- Banner Alzheimer's InstitutePhoenixArizonaUSA
- Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| | - Teresa Wu
- ASU‐Mayo Center for Innovative ImagingArizona State UniversityTempeArizonaUSA
- School of Computing and Augmented IntelligenceArizona State UniversityTempeArizonaUSA
| | - Eric M. Reiman
- Banner Alzheimer's InstitutePhoenixArizonaUSA
- Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
- ASU‐Banner Neurodegenerative Disease Research CenterArizona State UniversityTempeArizonaUSA
- Department of PsychiatryUniversity of ArizonaPhoenixArizonaUSA
| | - Yi Su
- Banner Alzheimer's InstitutePhoenixArizonaUSA
- Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
- Department of Neurology College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- ASU‐Mayo Center for Innovative ImagingArizona State UniversityTempeArizonaUSA
- School of Computing and Augmented IntelligenceArizona State UniversityTempeArizonaUSA
| |
Collapse
|
23
|
Young P, Heeman F, Axelsson J, Collij LE, Hitzel A, Sanaat A, Niñerola-Baizan A, Perissinotti A, Lubberink M, Frisoni GB, Zaidi H, Barkhof F, Farrar G, Baker S, Gispert JD, Garibotto V, Rieckmann A, Schöll M. Impact of simulated reduced injected dose on the assessment of amyloid PET scans. Eur J Nucl Med Mol Imaging 2024; 51:734-748. [PMID: 37897616 PMCID: PMC10796642 DOI: 10.1007/s00259-023-06481-0] [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: 07/11/2023] [Accepted: 10/15/2023] [Indexed: 10/30/2023]
Abstract
PURPOSE To investigate the impact of reduced injected doses on the quantitative and qualitative assessment of the amyloid PET tracers [18F]flutemetamol and [18F]florbetaben. METHODS Cognitively impaired and unimpaired individuals (N = 250, 36% Aβ-positive) were included and injected with [18F]flutemetamol (N = 175) or [18F]florbetaben (N = 75). PET scans were acquired in list-mode (90-110 min post-injection) and reduced-dose images were simulated to generate images of 75, 50, 25, 12.5 and 5% of the original injected dose. Images were reconstructed using vendor-provided reconstruction tools and visually assessed for Aβ-pathology. SUVRs were calculated for a global cortical and three smaller regions using a cerebellar cortex reference tissue, and Centiloid was computed. Absolute and percentage differences in SUVR and CL were calculated between dose levels, and the ability to discriminate between Aβ- and Aβ + scans was evaluated using ROC analyses. Finally, intra-reader agreement between the reduced dose and 100% images was evaluated. RESULTS At 5% injected dose, change in SUVR was 3.72% and 3.12%, with absolute change in Centiloid 3.35CL and 4.62CL, for [18F]flutemetamol and [18F]florbetaben, respectively. At 12.5% injected dose, percentage change in SUVR and absolute change in Centiloid were < 1.5%. AUCs for discriminating Aβ- from Aβ + scans were high (AUC ≥ 0.94) across dose levels, and visual assessment showed intra-reader agreement of > 80% for both tracers. CONCLUSION This proof-of-concept study showed that for both [18F]flutemetamol and [18F]florbetaben, adequate quantitative and qualitative assessments can be obtained at 12.5% of the original injected dose. However, decisions to reduce the injected dose should be made considering the specific clinical or research circumstances.
Collapse
Affiliation(s)
- Peter Young
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden
| | - Fiona Heeman
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Jan Axelsson
- Department of Radiation Sciences, Radiation Physics, Umeå University, Umeå, Sweden
| | - Lyduine E Collij
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Anne Hitzel
- Department of Nuclear Medicine, Toulouse University Hospital, Toulouse, France
| | - Amirhossein Sanaat
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Aida Niñerola-Baizan
- Nuclear Medicine Department, Hospital Clínic Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), ISCIII, Barcelona, Spain
| | - Andrés Perissinotti
- Nuclear Medicine Department, Hospital Clínic Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), ISCIII, Barcelona, Spain
| | - Mark Lubberink
- Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
- Geneva University Neurocenter, Geneva University, Geneva, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- UCL Institute of Neurology, London, UK
| | | | - Suzanne Baker
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, United States
| | - Juan Domingo Gispert
- Barcelona βeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva; NIMTLab; Center for Biomedical Imaging (CIBM), University of Geneva, Geneva, Switzerland
| | - Anna Rieckmann
- Institute for Psychology, Universität Der Bundeswehr München, Neubiberg, Germany
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden.
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK.
- Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
24
|
Xiong C, Schindler S, Luo J, Morris J, Bateman R, Holtzman D, Cruchaga C, Babulal G, Henson R, Benzinger T, Bui Q, Agboola F, Grant E, Emily G, Moulder K, Geldmacher D, Clay O, Roberson E, Murchison C, Wolk D, Shaw L. Baseline levels and longitudinal rates of change in plasma Aβ42/40 among self-identified Black/African American and White individuals. RESEARCH SQUARE 2024:rs.3.rs-3783571. [PMID: 38260384 PMCID: PMC10802715 DOI: 10.21203/rs.3.rs-3783571/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Objective The use of blood-based biomarkers of Alzheimer disease (AD) may facilitate access to biomarker testing of groups that have been historically under-represented in research. We evaluated whether plasma Aβ42/40 has similar or different baseline levels and longitudinal rates of change in participants racialized as Black or White. Methods The Study of Race to Understand Alzheimer Biomarkers (SORTOUT-AB) is a multi-center longitudinal study to evaluate for potential differences in AD biomarkers between individuals racialized as Black or White. Plasma samples collected at three AD Research Centers (Washington University, University of Pennsylvania, and University of Alabama-Birmingham) underwent analysis with C2N Diagnostics' PrecivityAD™ blood test for Aβ42 and Aβ40. General linear mixed effects models were used to estimate the baseline levels and rates of longitudinal change for plasma Aβ measures in both racial groups. Analyses also examined whether dementia status, age, sex, education, APOE ε4 carrier status, medical comorbidities, or fasting status modified potential racial differences. Results Of the 324 Black and 1,547 White participants, there were 158 Black and 759 White participants with plasma Aβ measures from at least two longitudinal samples over a mean interval of 6.62 years. At baseline, the group of Black participants had lower levels of plasma Aβ40 but similar levels of plasma Aβ42 as compared to the group of White participants. As a result, baseline plasma Aβ42/40 levels were higher in the Black group than the White group, consistent with the Black group having lower levels of amyloid pathology. Racial differences in plasma Aβ42/40 were not modified by age, sex, education, APOE ε4 carrier status, medical conditions (hypertension and diabetes), or fasting status. Despite differences in baseline levels, the Black and White groups had a similar longitudinal rate of change in plasma Aβ42/40. Interpretation Black individuals participating in AD research studies had a higher mean level of plasma Aβ42/40, consistent with a lower level of amyloid pathology, which, if confirmed, may imply a lower proportion of Black individuals being eligible for AD clinical trials in which the presence of amyloid is a prerequisite. However, there was no significant racial difference in the rate of change in plasma Aβ42/40, suggesting that amyloid pathology accumulates similarly across racialized groups.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Quoc Bui
- Washington University School of Medicine
| | | | | | | | | | | | | | | | | | - David Wolk
- Department of Neurology, University of Pennsylvania
| | - Leslie Shaw
- Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
25
|
Zammit MD, Betthauser TJ, McVea AK, Laymon CM, Tudorascu DL, Johnson SC, Hartley SL, Converse AK, Minhas DS, Zaman SH, Ances BM, Stone CK, Mathis CA, Cohen AD, Klunk WE, Handen BL, Christian BT. Characterizing the emergence of amyloid and tau burden in Down syndrome. Alzheimers Dement 2024; 20:388-398. [PMID: 37641577 PMCID: PMC10843570 DOI: 10.1002/alz.13444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/14/2023] [Accepted: 07/23/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Almost all individuals with Down syndrome (DS) will develop neuropathological features of Alzheimer's disease (AD). Understanding AD biomarker trajectories is necessary for DS-specific clinical interventions and interpretation of drug-related changes in the disease trajectory. METHODS A total of 177 adults with DS from the Alzheimer's Biomarker Consortium-Down Syndrome (ABC-DS) underwent positron emission tomography (PET) and MR imaging. Amyloid-beta (Aβ) trajectories were modeled to provide individual-level estimates of Aβ-positive (A+) chronicity, which were compared against longitudinal tau change. RESULTS Elevated tau was observed in all NFT regions following A+ and longitudinal tau increased with respect to A+ chronicity. Tau increases in NFT regions I-III was observed 0-2.5 years following A+. Nearly all A+ individuals had tau increases in the medial temporal lobe. DISCUSSION These findings highlight the rapid accumulation of amyloid and early onset of tau relative to amyloid in DS and provide a strategy for temporally characterizing AD neuropathology progression that is specific to the DS population and independent of chronological age. HIGHLIGHTS Longitudinal amyloid trajectories reveal rapid Aβ accumulation in Down syndrome NFT stage tau was strongly associated with A+ chronicity Early longitudinal tau increases were observed 2.5-5 years after reaching A.
Collapse
Affiliation(s)
| | - Tobey J. Betthauser
- University of Wisconsin‐Madison Alzheimer's Disease Research CenterMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Andrew K. McVea
- University of Wisconsin‐Madison Waisman CenterMadisonWisconsinUSA
| | - Charles M. Laymon
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Dana L. Tudorascu
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Sterling C. Johnson
- University of Wisconsin‐Madison Alzheimer's Disease Research CenterMadisonWisconsinUSA
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Sigan L. Hartley
- University of Wisconsin‐Madison Waisman CenterMadisonWisconsinUSA
| | | | - Davneet S. Minhas
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Shahid H. Zaman
- Cambridge Intellectual Disability Research GroupUniversity of CambridgeCambridgeUK
| | - Beau M. Ances
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Charles K. Stone
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Chester A. Mathis
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Annie D. Cohen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - William E. Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Benjamin L. Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bradley T. Christian
- University of Wisconsin‐Madison Waisman CenterMadisonWisconsinUSA
- Department of Medical PhysicsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | |
Collapse
|
26
|
Chen Y, Su Y, Wu J, Chen K, Atri A, Caselli RJ, Reiman EM, Wang Y. Combining Blood-Based Biomarkers and Structural MRI Measurements to Distinguish Persons with and without Significant Amyloid Plaques. J Alzheimers Dis 2024; 98:1415-1426. [PMID: 38578889 DOI: 10.3233/jad-231162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Background Amyloid-β (Aβ) plaques play a pivotal role in Alzheimer's disease. The current positron emission tomography (PET) is expensive and limited in availability. In contrast, blood-based biomarkers (BBBMs) show potential for characterizing Aβ plaques more affordably. We have previously proposed an MRI-based hippocampal morphometry measure to be an indicator of Aβ plaques. Objective To develop and validate an integrated model to predict brain amyloid PET positivity combining MRI feature and plasma Aβ42/40 ratio. Methods We extracted hippocampal multivariate morphometry statistics from MR images and together with plasma Aβ42/40 trained a random forest classifier to perform a binary classification of participant brain amyloid PET positivity. We evaluated the model performance using two distinct cohorts, one from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and the other from the Banner Alzheimer's Institute (BAI), including prediction accuracy, precision, recall rate, F1 score, and AUC score. Results Results from ADNI (mean age 72.6, Aβ+ rate 49.5%) and BAI (mean age 66.2, Aβ+ rate 36.9%) datasets revealed the integrated multimodal (IMM) model's superior performance over unimodal models. The IMM model achieved prediction accuracies of 0.86 in ADNI and 0.92 in BAI, surpassing unimodal models based solely on structural MRI (0.81 and 0.87) or plasma Aβ42/40 (0.73 and 0.81) predictors. CONCLUSIONS Our IMM model, combining MRI and BBBM data, offers a highly accurate approach to predict brain amyloid PET positivity. This innovative multiplex biomarker strategy presents an accessible and cost-effective avenue for advancing Alzheimer's disease diagnostics, leveraging diverse pathologic features related to Aβ plaques and structural MRI.
Collapse
Affiliation(s)
- Yanxi Chen
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Jianfeng Wu
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| | - Kewei Chen
- College of Health Solutions, Arizona State University, Tempe, AZ, USA
| | - Alireza Atri
- Banner Alzheimer's Institute, Phoenix, AZ, USA
- Banner Sun Health Research Institute, Sun City, AZ, USA
- Department of Neurology, Center for Brain/Mind Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Yalin Wang
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
27
|
Chen G, McKay NS, Gordon BA, Liu J, Joseph-Mathurin N, Schindler SE, Hassenstab J, Aschenbrenner AJ, Wang Q, Schultz SA, Su Y, LaMontagne PJ, Keefe SJ, Massoumzadeh P, Cruchaga C, Xiong C, Morris JC, Benzinger TLS. Predicting cognitive decline: Which is more useful, baseline amyloid levels or longitudinal change? Neuroimage Clin 2023; 41:103551. [PMID: 38150745 PMCID: PMC10788301 DOI: 10.1016/j.nicl.2023.103551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023]
Abstract
The use of biomarkers for the early detection of Alzheimer's disease (AD) is crucial for developing potential therapeutic treatments. Positron Emission Tomography (PET) is a well-established tool used to detect β-amyloid (Aβ) plaques in the brain. Previous studies have shown that cross-sectional biomarkers can predict cognitive decline (Schindler et al.,2021). However, it is still unclear whether longitudinal Aβ-PET may have additional value for predicting time to cognitive impairment in AD. The current study aims to evaluate the ability of baseline- versus longitudinal rate of change in-11C-Pittsburgh compound B (PiB) Aβ-PET to predict cognitive decline. A cohort of 153 participants who previously underwent PiB-PET scans and comprehensive clinical assessments were used in this study. Our analyses revealed that baseline Aβ is significantly associated with the rate of change in cognitive composite scores, with cognition declining more rapidly when baseline PiB Aβ levels were higher. In contrast, no signification association was identified between the rate of change in PiB-PET Aβ and cognitive decline. Additionally, the ability of the rate of change in the PiB-PET measures to predict cognitive decline was significantly influenced by APOE ε4 carrier status. These results suggest that a single PiB-PET scan is sufficient to predict cognitive decline and that longitudinal measures of Aβ accumulation do not improve the prediction of cognitive decline once someone is amyloid positive.
Collapse
Affiliation(s)
- Gengsheng Chen
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nicole S McKay
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jingxia Liu
- Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Qing Wang
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Stephanie A Schultz
- Department of Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Pamela J LaMontagne
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sarah J Keefe
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Parinaz Massoumzadeh
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Divison of Biostatistics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Departments of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
28
|
Rahmani F, Brier MR, Gordon BA, McKay N, Flores S, Keefe S, Hornbeck R, Ances B, Joseph‐Mathurin N, Xiong C, Wang G, Raji CA, Libre‐Guerra JJ, Perrin RJ, McDade E, Daniels A, Karch C, Day GS, Brickman AM, Fulham M, Jack CR, la La Fougère C, Reischl G, Schofield PR, Oh H, Levin J, Vöglein J, Cash DM, Yakushev I, Ikeuchi T, Klunk WE, Morris JC, Bateman RJ, Benzinger TLS. T1 and FLAIR signal intensities are related to tau pathology in dominantly inherited Alzheimer disease. Hum Brain Mapp 2023; 44:6375-6387. [PMID: 37867465 PMCID: PMC10681640 DOI: 10.1002/hbm.26514] [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: 07/24/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
Carriers of mutations responsible for dominantly inherited Alzheimer disease provide a unique opportunity to study potential imaging biomarkers. Biomarkers based on routinely acquired clinical MR images, could supplement the extant invasive or logistically challenging) biomarker studies. We used 1104 longitudinal MR, 324 amyloid beta, and 87 tau positron emission tomography imaging sessions from 525 participants enrolled in the Dominantly Inherited Alzheimer Network Observational Study to extract novel imaging metrics representing the mean (μ) and standard deviation (σ) of standardized image intensities of T1-weighted and Fluid attenuated inversion recovery (FLAIR) MR scans. There was an exponential decrease in FLAIR-μ in mutation carriers and an increase in FLAIR and T1 signal heterogeneity (T1-σ and FLAIR-σ) as participants approached the symptom onset in both supramarginal, the right postcentral and right superior temporal gyri as well as both caudate nuclei, putamina, thalami, and amygdalae. After controlling for the effect of regional atrophy, FLAIR-μ decreased and T1-σ and FLAIR-σ increased with increasing amyloid beta and tau deposition in numerous cortical regions. In symptomatic mutation carriers and independent of the effect of regional atrophy, tau pathology demonstrated a stronger relationship with image intensity metrics, compared with amyloid pathology. We propose novel MR imaging intensity-based metrics using standard clinical T1 and FLAIR images which strongly associates with the progression of pathology in dominantly inherited Alzheimer disease. We suggest that tau pathology may be a key driver of the observed changes in this cohort of patients.
Collapse
Affiliation(s)
| | | | - Brian A. Gordon
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Nicole McKay
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Shaney Flores
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Sarah Keefe
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Russ Hornbeck
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Beau Ances
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | - Chengjie Xiong
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Guoqiao Wang
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Cyrus A. Raji
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | | | - Eric McDade
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Alisha Daniels
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Celeste Karch
- Washington University School of MedicineSt. LouisMissouriUSA
| | - Gregory S. Day
- Mayo Clinic, Department of NeurologyJacksonvilleFloridaUSA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer's Disease & the Aging Brain, and Department of Neurology College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | | | | | - Christian la La Fougère
- Department of Nuclear Medicine and Clinical Molecular ImagingUniversity Hospital TuebingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE) TuebingenTübingenGermany
- Department of Preclinical Imaging and RadiopharmacyEberhard Karls University TübingenTübingenGermany
| | - Gerald Reischl
- Department of Nuclear Medicine and Clinical Molecular ImagingUniversity Hospital TuebingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE) TuebingenTübingenGermany
- Department of Preclinical Imaging and RadiopharmacyEberhard Karls University TübingenTübingenGermany
| | - Peter R. Schofield
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Biomedical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Hwamee Oh
- Brown UniversityProvidenceRhode IslandUSA
| | - Johannes Levin
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Jonathan Vöglein
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - David M. Cash
- UK Dementia Research Institute at University College LondonLondonUK
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Igor Yakushev
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE), site MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | | | | | - John C. Morris
- Washington University School of MedicineSt. LouisMissouriUSA
| | | | | | | |
Collapse
|
29
|
Babulal GM, Chen L, Murphy SA, Doherty JM, Johnson AM, Morris JC. Neuropsychiatric Symptoms and Alzheimer Disease Biomarkers Independently Predict Progression to Incident Cognitive Impairment. Am J Geriatr Psychiatry 2023; 31:1190-1199. [PMID: 37544835 PMCID: PMC10861300 DOI: 10.1016/j.jagp.2023.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/08/2023]
Abstract
OBJECTIVES To investigate the effect of neuropsychiatric symptoms and depression symptoms, respectively, and Alzheimer disease (AD) biomarkers (cerebrospinal fluid [CSF] or Positron Emission Tomography [PET] imaging) on the progression to incident cognitive impairment among cognitively normal older adults. DESIGN Prospective, observation, longitudinal study. SETTING Knight Alzheimer Disease Research Center (ADRC) at Washington University School of Medicine. PARTICIPANTS Older adults aged 65 and above who participated in AD longitudinal studies (n = 286). MEASUREMENTS CSF and PET biomarkers, Clinical Dementia Rating (CDR), Geriatric Depression Scale (GDS), and Neuropsychiatric Inventory Questionnaire (NPI-Q). RESULTS Participants had an average follow-up of eight years, and 31 progressed from CDR 0 to CDR >0. After adjusting for sex, age, and education in the Cox proportional hazards survival models, neuropsychiatric symptoms as a time-dependent covariate was statistically significant in the three CSF (Aβ42/Aβ40, t-Tau/Aβ42, p-Tau/Aβ42) PET imaging models (HR = 1.33-1.50). The biomarkers were also significant as main effects (HR = 2.00-4.04). Change in depression symptoms was not significant in any models. The interactions between biomarkers and neuropsychiatric symptoms and depression were not statistically significant. CONCLUSIONS Changes in neuropsychiatric symptoms increase the risk of progression to cognitive impairment among healthy, cognitively normal adults, independent of AD biomarkers. Routine assessment of neuropsychiatric symptoms could provide valuable clinical information about cognitive functioning and preclinical disease state.
Collapse
Affiliation(s)
- Ganesh M Babulal
- Department of Neurology (GMB, SAM, JCM), Washington University in St. Louis, St. Louis, MO; Institute of Public Health (GMB), Washington University in St. Louis, St. Louis, MO; Department of Psychology, Faculty of Humanities (GMB), University of Johannesburg, Johannesburg, South Africa; Department of Clinical Research and Leadership (GMB), The George Washington University School of Medicine and Health Sciences, Washington, DC.
| | - Ling Chen
- Division of Biostatistics (LC), Washington University in St. Louis, St. Louis, MO
| | - Samantha A Murphy
- Department of Neurology (GMB, SAM, JCM), Washington University in St. Louis, St. Louis, MO
| | - Jason M Doherty
- Department of Neurology (GMB, SAM, JCM), Washington University in St. Louis, St. Louis, MO
| | - Ann M Johnson
- Center for Clinical Studies (AMJ), Washington University in St. Louis, St. Louis, MO
| | - John C Morris
- Department of Neurology (GMB, SAM, JCM), Washington University in St. Louis, St. Louis, MO; Hope Center for Neurological Disorders (JCM), Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
30
|
Stojanovic M, Babulal GM, Head D. Determinants of physical activity engagement in older adults. J Behav Med 2023; 46:757-769. [PMID: 36920727 PMCID: PMC10502182 DOI: 10.1007/s10865-023-00404-y] [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: 10/28/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023]
Abstract
In order to increase engagement in physical activity, it is important to determine which factors contribute to physical activity engagement in older adults. The current study examined the relative predictive ability of several potential determinants, in terms of both the concurrent level as well as longitudinal trajectories. Clinically normal adults aged 61-92 completed the Physical Activity Scale for the Elderly (n = 189 for cross-sectional models; n = 214 for longitudinal models). Potential determinants included age, gender, education, physical health, sensory health, mood, cardiovascular health, cognitive status, and biomarkers of Alzheimer disease (AD). We observed a novel finding that both concurrent physical health (p < 0.001) and change in physical health (p < 0.001) were significant predictors above and beyond other determinants. Concurrent mood predicted levels of physical activity (p = 0.035), particularly in females. These findings suggest that poor physical health and low mood might be important to consider as potential barriers to physical activity engagement in older adults.
Collapse
Affiliation(s)
- Marta Stojanovic
- Department of Psychological & Brain Sciences, Washington University in St. Louis, One Brookings Drive, St. Louis, MO, Box 1125, USA.
| | - Ganesh M Babulal
- Department of Psychology, Faculty of Humanities, University of Johannesburg, Johannesburg, South Africa
- Department of Clinical Research and Leadership, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Institute of Public Health, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Denise Head
- Department of Psychological & Brain Sciences, Washington University in St. Louis, One Brookings Drive, St. Louis, MO, Box 1125, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
31
|
Jovalekic A, Roé-Vellvé N, Koglin N, Quintana ML, Nelson A, Diemling M, Lilja J, Gómez-González JP, Doré V, Bourgeat P, Whittington A, Gunn R, Stephens AW, Bullich S. Validation of quantitative assessment of florbetaben PET scans as an adjunct to the visual assessment across 15 software methods. Eur J Nucl Med Mol Imaging 2023; 50:3276-3289. [PMID: 37300571 PMCID: PMC10542295 DOI: 10.1007/s00259-023-06279-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023]
Abstract
PURPOSE Amyloid positron emission tomography (PET) with [18F]florbetaben (FBB) is an established tool for detecting Aβ deposition in the brain in vivo based on visual assessment of PET scans. Quantitative measures are commonly used in the research context and allow continuous measurement of amyloid burden. The aim of this study was to demonstrate the robustness of FBB PET quantification. METHODS This is a retrospective analysis of FBB PET images from 589 subjects. PET scans were quantified with 15 analytical methods using nine software packages (MIMneuro, Hermes BRASS, Neurocloud, Neurology Toolkit, statistical parametric mapping (SPM8), PMOD Neuro, CapAIBL, non-negative matrix factorization (NMF), AmyloidIQ) that used several metrics to estimate Aβ load (SUVR, centiloid, amyloid load, and amyloid index). Six analytical methods reported centiloid (MIMneuro, standard centiloid, Neurology Toolkit, SPM8 (PET only), CapAIBL, NMF). All results were quality controlled. RESULTS The mean sensitivity, specificity, and accuracy were 96.1 ± 1.6%, 96.9 ± 1.0%, and 96.4 ± 1.1%, respectively, for all quantitative methods tested when compared to histopathology, where available. The mean percentage of agreement between binary quantitative assessment across all 15 methods and visual majority assessment was 92.4 ± 1.5%. Assessments of reliability, correlation analyses, and comparisons across software packages showed excellent performance and consistent results between analytical methods. CONCLUSION This study demonstrated that quantitative methods using both CE marked software and other widely available processing tools provided comparable results to visual assessments of FBB PET scans. Software quantification methods, such as centiloid analysis, can complement visual assessment of FBB PET images and could be used in the future for identification of early amyloid deposition, monitoring disease progression and treatment effectiveness.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
32
|
McKay NS, Gordon BA, Hornbeck RC, Dincer A, Flores S, Keefe SJ, Joseph-Mathurin N, Jack CR, Koeppe R, Millar PR, Ances BM, Chen CD, Daniels A, Hobbs DA, Jackson K, Koudelis D, Massoumzadeh P, McCullough A, Nickels ML, Rahmani F, Swisher L, Wang Q, Allegri RF, Berman SB, Brickman AM, Brooks WS, Cash DM, Chhatwal JP, Day GS, Farlow MR, la Fougère C, Fox NC, Fulham M, Ghetti B, Graff-Radford N, Ikeuchi T, Klunk W, Lee JH, Levin J, Martins R, Masters CL, McConathy J, Mori H, Noble JM, Reischl G, Rowe C, Salloway S, Sanchez-Valle R, Schofield PR, Shimada H, Shoji M, Su Y, Suzuki K, Vöglein J, Yakushev I, Cruchaga C, Hassenstab J, Karch C, McDade E, Perrin RJ, Xiong C, Morris JC, Bateman RJ, Benzinger TLS. Positron emission tomography and magnetic resonance imaging methods and datasets within the Dominantly Inherited Alzheimer Network (DIAN). Nat Neurosci 2023; 26:1449-1460. [PMID: 37429916 PMCID: PMC10400428 DOI: 10.1038/s41593-023-01359-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/15/2023] [Indexed: 07/12/2023]
Abstract
The Dominantly Inherited Alzheimer Network (DIAN) is an international collaboration studying autosomal dominant Alzheimer disease (ADAD). ADAD arises from mutations occurring in three genes. Offspring from ADAD families have a 50% chance of inheriting their familial mutation, so non-carrier siblings can be recruited for comparisons in case-control studies. The age of onset in ADAD is highly predictable within families, allowing researchers to estimate an individual's point in the disease trajectory. These characteristics allow candidate AD biomarker measurements to be reliably mapped during the preclinical phase. Although ADAD represents a small proportion of AD cases, understanding neuroimaging-based changes that occur during the preclinical period may provide insight into early disease stages of 'sporadic' AD also. Additionally, this study provides rich data for research in healthy aging through inclusion of the non-carrier controls. Here we introduce the neuroimaging dataset collected and describe how this resource can be used by a range of researchers.
Collapse
Affiliation(s)
| | | | | | - Aylin Dincer
- Washington University in St. Louis, St. Louis, MO, USA
| | - Shaney Flores
- Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah J Keefe
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | | | | | - Beau M Ances
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | - Diana A Hobbs
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | | | | | | | | | - Laura Swisher
- Washington University in St. Louis, St. Louis, MO, USA
| | - Qing Wang
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | - Adam M Brickman
- Columbia University Irving Medical Center, New York, NY, USA
| | - William S Brooks
- Neuroscience Research Australia, Sydney, New South Wales, Australia
| | - David M Cash
- UK Dementia Research Institute at University College London, London, UK
- University College London, London, UK
| | - Jasmeer P Chhatwal
- Massachusetts General and Brigham & Women's Hospitals, Harvard Medical School, Boston, MA, USA
| | | | | | - Christian la Fougère
- Department of Radiology, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Nick C Fox
- UK Dementia Research Institute at University College London, London, UK
- University College London, London, UK
| | - Michael Fulham
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | | | | | | | | | | | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Ralph Martins
- Edith Cowan University, Joondalup, Western Australia, Australia
| | | | | | | | - James M Noble
- Columbia University Irving Medical Center, New York, NY, USA
| | - Gerald Reischl
- Department of Radiology, University of Tübingen, Tübingen, Germany
| | | | | | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | | | | | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | | | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, München, Germany
| | - Igor Yakushev
- School of Medicine, Technical University of Munich, Munich, Germany
| | | | | | - Celeste Karch
- Washington University in St. Louis, St. Louis, MO, USA
| | - Eric McDade
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | | - John C Morris
- Washington University in St. Louis, St. Louis, MO, USA
| | | | | |
Collapse
|
33
|
Chen CD, McCullough A, Gordon B, Joseph-Mathurin N, Flores S, McKay NS, Hobbs DA, Hornbeck R, Fagan AM, Cruchaga C, Goate AM, Perrin RJ, Wang G, Li Y, Shi X, Xiong C, Pontecorvo MJ, Klein G, Su Y, Klunk WE, Jack C, Koeppe R, Snider BJ, Berman SB, Roberson ED, Brosch J, Surti G, Jiménez-Velázquez IZ, Galasko D, Honig LS, Brooks WS, Clarnette R, Wallon D, Dubois B, Pariente J, Pasquier F, Sanchez-Valle R, Shcherbinin S, Higgins I, Tunali I, Masters CL, van Dyck CH, Masellis M, Hsiung R, Gauthier S, Salloway S, Clifford DB, Mills S, Supnet-Bell C, McDade E, Bateman RJ, Benzinger TLS. Longitudinal head-to-head comparison of 11C-PiB and 18F-florbetapir PET in a Phase 2/3 clinical trial of anti-amyloid-β monoclonal antibodies in dominantly inherited Alzheimer's disease. Eur J Nucl Med Mol Imaging 2023; 50:2669-2682. [PMID: 37017737 PMCID: PMC10330155 DOI: 10.1007/s00259-023-06209-0] [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: 12/08/2022] [Accepted: 03/18/2023] [Indexed: 04/06/2023]
Abstract
PURPOSE Pittsburgh Compound-B (11C-PiB) and 18F-florbetapir are amyloid-β (Aβ) positron emission tomography (PET) radiotracers that have been used as endpoints in Alzheimer's disease (AD) clinical trials to evaluate the efficacy of anti-Aβ monoclonal antibodies. However, comparing drug effects between and within trials may become complicated if different Aβ radiotracers were used. To study the consequences of using different Aβ radiotracers to measure Aβ clearance, we performed a head-to-head comparison of 11C-PiB and 18F-florbetapir in a Phase 2/3 clinical trial of anti-Aβ monoclonal antibodies. METHODS Sixty-six mutation-positive participants enrolled in the gantenerumab and placebo arms of the first Dominantly Inherited Alzheimer Network Trials Unit clinical trial (DIAN-TU-001) underwent both 11C-PiB and 18F-florbetapir PET imaging at baseline and during at least one follow-up visit. For each PET scan, regional standardized uptake value ratios (SUVRs), regional Centiloids, a global cortical SUVR, and a global cortical Centiloid value were calculated. Longitudinal changes in SUVRs and Centiloids were estimated using linear mixed models. Differences in longitudinal change between PET radiotracers and between drug arms were estimated using paired and Welch two sample t-tests, respectively. Simulated clinical trials were conducted to evaluate the consequences of some research sites using 11C-PiB while other sites use 18F-florbetapir for Aβ PET imaging. RESULTS In the placebo arm, the absolute rate of longitudinal change measured by global cortical 11C-PiB SUVRs did not differ from that of global cortical 18F-florbetapir SUVRs. In the gantenerumab arm, global cortical 11C-PiB SUVRs decreased more rapidly than global cortical 18F-florbetapir SUVRs. Drug effects were statistically significant across both Aβ radiotracers. In contrast, the rates of longitudinal change measured in global cortical Centiloids did not differ between Aβ radiotracers in either the placebo or gantenerumab arms, and drug effects remained statistically significant. Regional analyses largely recapitulated these global cortical analyses. Across simulated clinical trials, type I error was higher in trials where both Aβ radiotracers were used versus trials where only one Aβ radiotracer was used. Power was lower in trials where 18F-florbetapir was primarily used versus trials where 11C-PiB was primarily used. CONCLUSION Gantenerumab treatment induces longitudinal changes in Aβ PET, and the absolute rates of these longitudinal changes differ significantly between Aβ radiotracers. These differences were not seen in the placebo arm, suggesting that Aβ-clearing treatments may pose unique challenges when attempting to compare longitudinal results across different Aβ radiotracers. Our results suggest converting Aβ PET SUVR measurements to Centiloids (both globally and regionally) can harmonize these differences without losing sensitivity to drug effects. Nonetheless, until consensus is achieved on how to harmonize drug effects across radiotracers, and since using multiple radiotracers in the same trial may increase type I error, multisite studies should consider potential variability due to different radiotracers when interpreting Aβ PET biomarker data and, if feasible, use a single radiotracer for the best results. TRIAL REGISTRATION ClinicalTrials.gov NCT01760005. Registered 31 December 2012. Retrospectively registered.
Collapse
Affiliation(s)
- Charles D Chen
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Washington University School of Medicine, 660 South Euclid, Campus Box 8225, St. Louis, MO, 63110, USA
| | - Austin McCullough
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Brian Gordon
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicole S McKay
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Diana A Hobbs
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Russ Hornbeck
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Alison M Goate
- Department of Genetics and Genomic Sciences, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - Richard J Perrin
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Guoqiao Wang
- Department of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Xinyu Shi
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Chengjie Xiong
- Department of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael J Pontecorvo
- Avid Radiopharmaceuticals, Philadelphia, PA, USA
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Yi Su
- Banner Alzheimer's Institute, Banner Health, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Clifford Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Robert Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - B Joy Snider
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Erik D Roberson
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jared Brosch
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ghulam Surti
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | - Douglas Galasko
- Department of Neurology, University of California San Diego, San Diego, CA, USA
| | | | - William S Brooks
- Prince of Wales Medical Research Institute, University of New South Wales, Sydney, NSW, Australia
| | - Roger Clarnette
- Department of Internal Medicine, University of Western Australia, Crawley, WA, Australia
| | - David Wallon
- Department of Neurology and CNR-MAJ, Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, F-76000, Rouen, France
| | - Bruno Dubois
- Sorbonne Université, AP-HP, GRC No. 21, APM, Hôpital de La Pitié-Salpêtrière, Paris, France
- Institut du Cerveau Et de La Moelle Épinière, INSERM U1127, CNRS UMR 7225, Paris, France
- Institut de La Mémoire Et de La Maladie d'Alzheimer, Département de Neurologie, Hôpital de La Pitié-Salpêtrière, Paris, France
| | - Jérémie Pariente
- Department of Neurology, Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Toulouse NeuroImaging Centre, Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Florence Pasquier
- Univ. Lille, INSERM, CHU Lille, 59000, Lille, France
- CNR-MAJ, Labex DISTALZ, LiCEND, 59000, Lille, France
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital ClínicInstitut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Fundació Clínic Per a La Recerca Biomèdica, University of Barcelona, Barcelona, Spain
| | | | | | - Ilke Tunali
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | | | - Robin Hsiung
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Serge Gauthier
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Steve Salloway
- Alpert Medical School of Brown University, Providence, RI, USA
| | - David B Clifford
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Susan Mills
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Eric McDade
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
34
|
Ferreiro AL, Choi J, Ryou J, Newcomer EP, Thompson R, Bollinger RM, Hall-Moore C, Ndao IM, Sax L, Benzinger TLS, Stark SL, Holtzman DM, Fagan AM, Schindler SE, Cruchaga C, Butt OH, Morris JC, Tarr PI, Ances BM, Dantas G. Gut microbiome composition may be an indicator of preclinical Alzheimer's disease. Sci Transl Med 2023; 15:eabo2984. [PMID: 37315112 PMCID: PMC10680783 DOI: 10.1126/scitranslmed.abo2984] [Citation(s) in RCA: 84] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/26/2023] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) pathology is thought to progress from normal cognition through preclinical disease and ultimately to symptomatic AD with cognitive impairment. Recent work suggests that the gut microbiome of symptomatic patients with AD has an altered taxonomic composition compared with that of healthy, cognitively normal control individuals. However, knowledge about changes in the gut microbiome before the onset of symptomatic AD is limited. In this cross-sectional study that accounted for clinical covariates and dietary intake, we compared the taxonomic composition and gut microbial function in a cohort of 164 cognitively normal individuals, 49 of whom showed biomarker evidence of early preclinical AD. Gut microbial taxonomic profiles of individuals with preclinical AD were distinct from those of individuals without evidence of preclinical AD. The change in gut microbiome composition correlated with β-amyloid (Aβ) and tau pathological biomarkers but not with biomarkers of neurodegeneration, suggesting that the gut microbiome may change early in the disease process. We identified specific gut bacterial taxa associated with preclinical AD. Inclusion of these microbiome features improved the accuracy, sensitivity, and specificity of machine learning classifiers for predicting preclinical AD status when tested on a subset of the cohort (65 of the 164 participants). Gut microbiome correlates of preclinical AD neuropathology may improve our understanding of AD etiology and may help to identify gut-derived markers of AD risk.
Collapse
Affiliation(s)
- Aura L. Ferreiro
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - JooHee Choi
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jian Ryou
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erin P. Newcomer
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Regina Thompson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rebecca M. Bollinger
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Hall-Moore
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - I. Malick Ndao
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laurie Sax
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tammie L. S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan L. Stark
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Omar H. Butt
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Phillip I. Tarr
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beau M. Ances
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gautam Dantas
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
35
|
Nicosia J, Aschenbrenner AJ, Balota DA, Sliwinski MJ, Tahan M, Adams S, Stout SS, Wilks H, Gordon BA, Benzinger TL, Fagan AM, Xiong C, Bateman RJ, Morris JC, Hassenstab J. Unsupervised high-frequency smartphone-based cognitive assessments are reliable, valid, and feasible in older adults at risk for Alzheimer's disease. J Int Neuropsychol Soc 2023; 29:459-471. [PMID: 36062528 PMCID: PMC9985662 DOI: 10.1017/s135561772200042x] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Smartphones have the potential for capturing subtle changes in cognition that characterize preclinical Alzheimer's disease (AD) in older adults. The Ambulatory Research in Cognition (ARC) smartphone application is based on principles from ecological momentary assessment (EMA) and administers brief tests of associative memory, processing speed, and working memory up to 4 times per day over 7 consecutive days. ARC was designed to be administered unsupervised using participants' personal devices in their everyday environments. METHODS We evaluated the reliability and validity of ARC in a sample of 268 cognitively normal older adults (ages 65-97 years) and 22 individuals with very mild dementia (ages 61-88 years). Participants completed at least one 7-day cycle of ARC testing and conventional cognitive assessments; most also completed cerebrospinal fluid, amyloid and tau positron emission tomography, and structural magnetic resonance imaging studies. RESULTS First, ARC tasks were reliable as between-person reliability across the 7-day cycle and test-retest reliabilities at 6-month and 1-year follow-ups all exceeded 0.85. Second, ARC demonstrated construct validity as evidenced by correlations with conventional cognitive measures (r = 0.53 between composite scores). Third, ARC measures correlated with AD biomarker burden at baseline to a similar degree as conventional cognitive measures. Finally, the intensive 7-day cycle indicated that ARC was feasible (86.50% approached chose to enroll), well tolerated (80.42% adherence, 4.83% dropout), and was rated favorably by older adult participants. CONCLUSIONS Overall, the results suggest that ARC is reliable and valid and represents a feasible tool for assessing cognitive changes associated with the earliest stages of AD.
Collapse
Affiliation(s)
- Jessica Nicosia
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J. Aschenbrenner
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David A. Balota
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Martin J. Sliwinski
- Department of Human Development and Family Studies, The Pennsylvania State University, University Park, PA, USA
| | - Marisol Tahan
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah Adams
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah S. Stout
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah Wilks
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A. Gordon
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M. Fagan
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J. Bateman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
36
|
Coath W, Modat M, Cardoso MJ, Markiewicz PJ, Lane CA, Parker TD, Keshavan A, Buchanan SM, Keuss SE, Harris MJ, Burgos N, Dickson J, Barnes A, Thomas DL, Beasley D, Malone IB, Wong A, Erlandsson K, Thomas BA, Schöll M, Ourselin S, Richards M, Fox NC, Schott JM, Cash DM. Operationalizing the centiloid scale for [ 18F]florbetapir PET studies on PET/MRI. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12434. [PMID: 37201176 PMCID: PMC10186069 DOI: 10.1002/dad2.12434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/03/2023] [Accepted: 02/19/2023] [Indexed: 05/20/2023]
Abstract
INTRODUCTION The Centiloid scale aims to harmonize amyloid beta (Aβ) positron emission tomography (PET) measures across different analysis methods. As Centiloids were created using PET/computerized tomography (CT) data and are influenced by scanner differences, we investigated the Centiloid transformation with data from Insight 46 acquired with PET/magnetic resonanceimaging (MRI). METHODS We transformed standardized uptake value ratios (SUVRs) from 432 florbetapir PET/MRI scans processed using whole cerebellum (WC) and white matter (WM) references, with and without partial volume correction. Gaussian-mixture-modelling-derived cutpoints for Aβ PET positivity were converted. RESULTS The Centiloid cutpoint was 14.2 for WC SUVRs. The relationship between WM and WC uptake differed between the calibration and testing datasets, producing implausibly low WM-based Centiloids. Linear adjustment produced a WM-based cutpoint of 18.1. DISCUSSION Transformation of PET/MRI florbetapir data to Centiloids is valid. However, further understanding of the effects of acquisition or biological factors on the transformation using a WM reference is needed. HIGHLIGHTS Centiloid conversion of amyloid beta positron emission tomography (PET) data aims to standardize results.Centiloid values can be influenced by differences in acquisition.We converted florbetapir PET/magnetic resonance imaging data from a large birth cohort.Whole cerebellum referenced values could be reliably transformed to Centiloids.White matter referenced values may be less generalizable between datasets.
Collapse
Affiliation(s)
- William Coath
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Marc Modat
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - M. Jorge Cardoso
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Pawel J. Markiewicz
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical EngineeringUCLLondonUK
| | | | - Thomas D. Parker
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Ashvini Keshavan
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Sarah M. Buchanan
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Sarah E. Keuss
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Matthew J. Harris
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Ninon Burgos
- Sorbonne Université, Institut du Cerveau ‐ Paris Brain Institute ‐ ICM, Inserm, CNRS, AP‐HP, Hôpital Pitié Salpêtrière, InriaAramis project‐teamParisFrance
| | - John Dickson
- Institute of Nuclear MedicineUniversity College London HospitalsLondonUK
| | - Anna Barnes
- Institute of Nuclear MedicineUniversity College London HospitalsLondonUK
| | - David L. Thomas
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
- Department of Brain Repair and RehabilitationUCL Queen Square Institute of NeurologyLondonUK
- Wellcome Centre for Human Neuroimaging, Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Daniel Beasley
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Ian B. Malone
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCLLondonUK
| | - Kjell Erlandsson
- Institute of Nuclear MedicineUniversity College London HospitalsLondonUK
| | - Benjamin A. Thomas
- Institute of Nuclear MedicineUniversity College London HospitalsLondonUK
| | - Michael Schöll
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgMölndalSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgMölndalSweden
| | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | | | - Nick C. Fox
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
- Dementia Research InstituteUCL Queen Square Institute of NeurologyLondonUK
| | | | - David M. Cash
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical EngineeringUCLLondonUK
- Dementia Research InstituteUCL Queen Square Institute of NeurologyLondonUK
| | | |
Collapse
|
37
|
Barthélemy NR, Saef B, Li Y, Gordon BA, He Y, Horie K, Stomrud E, Salvadó G, Janelidze S, Sato C, Ovod V, Henson RL, Fagan AM, Benzinger TLS, Xiong C, Morris JC, Hansson O, Bateman RJ, Schindler SE. CSF tau phosphorylation occupancies at T217 and T205 represent improved biomarkers of amyloid and tau pathology in Alzheimer's disease. NATURE AGING 2023; 3:391-401. [PMID: 37117788 PMCID: PMC10154225 DOI: 10.1038/s43587-023-00380-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/03/2023] [Indexed: 04/30/2023]
Abstract
Cerebrospinal fluid (CSF) amyloid-β peptide (Aβ)42/Aβ40 and the concentration of tau phosphorylated at site 181 (p-tau181) are well-established biomarkers of Alzheimer's disease (AD). The present study used mass spectrometry to measure concentrations of nine phosphorylated and five nonphosphorylated tau species and phosphorylation occupancies (percentage phosphorylated/nonphosphorylated) at ten sites. In the present study we show that, in 750 individuals with a median age of 71.2 years, CSF pT217/T217 predicted the presence of brain amyloid by positron emission tomography (PET) slightly better than Aβ42/Aβ40 (P = 0.02). Furthermore, for individuals with positive brain amyloid by PET (n = 263), CSF pT217/T217 was more strongly correlated with the amount of amyloid (Spearman's ρ = 0.69) than Aβ42/Aβ40 (ρ = -0.42, P < 0.0001). In two independent cohorts of participants with symptoms of AD dementia (n = 55 and n = 90), CSF pT217/T217 and pT205/T205 were better correlated with tau PET measures than CSF p-tau181 concentration. These findings suggest that CSF pT217/T217 and pT205/T205 represent improved CSF biomarkers of amyloid and tau pathology in AD.
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA.
| | - Benjamin Saef
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Gemma Salvadó
- 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
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Vitaliy Ovod
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Rachel L Henson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- 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
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- 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
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - 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.
| |
Collapse
|
38
|
Rahmani F, Jindal S, Raji CA, Wang W, Nazeri A, Perez-Carrillo GG, Miller-Thomas MM, Graner P, Marechal B, Shah A, Zimmermann M, Chen CD, Keefe S, LaMontagne P, Benzinger TLS. Validity Assessment of an Automated Brain Morphometry Tool for Patients with De Novo Memory Symptoms. AJNR Am J Neuroradiol 2023; 44:261-267. [PMID: 36797031 PMCID: PMC10187815 DOI: 10.3174/ajnr.a7790] [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: 06/30/2022] [Accepted: 01/09/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND AND PURPOSE Automated volumetric analysis of structural MR imaging allows quantitative assessment of brain atrophy in neurodegenerative disorders. We compared the brain segmentation performance of the AI-Rad Companion brain MR imaging software against an in-house FreeSurfer 7.1.1/Individual Longitudinal Participant pipeline. MATERIALS AND METHODS T1-weighted images of 45 participants with de novo memory symptoms were selected from the OASIS-4 database and analyzed through the AI-Rad Companion brain MR imaging tool and the FreeSurfer 7.1.1/Individual Longitudinal Participant pipeline. Correlation, agreement, and consistency between the 2 tools were compared among the absolute, normalized, and standardized volumes. Final reports generated by each tool were used to compare the rates of detection of abnormality and the compatibility of radiologic impressions made using each tool, compared with the clinical diagnoses. RESULTS We observed strong correlation, moderate consistency, and poor agreement between absolute volumes of the main cortical lobes and subcortical structures measured by the AI-Rad Companion brain MR imaging tool compared with FreeSurfer. The strength of the correlations increased after normalizing the measurements to the total intracranial volume. Standardized measurements differed significantly between the 2 tools, likely owing to differences in the normative data sets used to calibrate each tool. When considering the FreeSurfer 7.1.1/Individual Longitudinal Participant pipeline as a reference standard, the AI-Rad Companion brain MR imaging tool had a specificity of 90.6%-100% and a sensitivity of 64.3%-100% in detecting volumetric abnormalities. There was no difference between the rate of compatibility of radiologic and clinical impressions when using the 2 tools. CONCLUSIONS The AI-Rad Companion brain MR imaging tool reliably detects atrophy in cortical and subcortical regions implicated in the differential diagnosis of dementia.
Collapse
Affiliation(s)
- F Rahmani
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
- Charles F. and Joanne Knight Alzheimer Disease Research Center (F.R., S.J., C.A.R., W.W., A.N., C.D.C., T.L.S.B.), Washington University in St. Louis, St. Lous, Missouri
| | - S Jindal
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
- Charles F. and Joanne Knight Alzheimer Disease Research Center (F.R., S.J., C.A.R., W.W., A.N., C.D.C., T.L.S.B.), Washington University in St. Louis, St. Lous, Missouri
| | - C A Raji
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
- Charles F. and Joanne Knight Alzheimer Disease Research Center (F.R., S.J., C.A.R., W.W., A.N., C.D.C., T.L.S.B.), Washington University in St. Louis, St. Lous, Missouri
| | - W Wang
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
- Charles F. and Joanne Knight Alzheimer Disease Research Center (F.R., S.J., C.A.R., W.W., A.N., C.D.C., T.L.S.B.), Washington University in St. Louis, St. Lous, Missouri
| | - A Nazeri
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
- Charles F. and Joanne Knight Alzheimer Disease Research Center (F.R., S.J., C.A.R., W.W., A.N., C.D.C., T.L.S.B.), Washington University in St. Louis, St. Lous, Missouri
| | - G G Perez-Carrillo
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
| | - M M Miller-Thomas
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
| | - P Graner
- Siemens Medical Solutions (P.G., B.M., M.Z.), Malvern, Pennsylvania
- Advanced Clinical Imaging Technology (P.G., B.M., M.Z.), Siemens Healthcare, Lausanne, Switzerland
- Department of Radiology (P.G., B.M., M.Z.), Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (P.G., B.M., A.S., M.Z.), Lausanne, Switzerland
- Siemens Healthcare (P.G., B.M., M.Z.), Erlangen, Germany
| | - B Marechal
- Siemens Medical Solutions (P.G., B.M., M.Z.), Malvern, Pennsylvania
- Advanced Clinical Imaging Technology (P.G., B.M., M.Z.), Siemens Healthcare, Lausanne, Switzerland
- Department of Radiology (P.G., B.M., M.Z.), Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (P.G., B.M., A.S., M.Z.), Lausanne, Switzerland
- Siemens Healthcare (P.G., B.M., M.Z.), Erlangen, Germany
| | - A Shah
- LTS5, École Polytechnique Fédérale de Lausanne (P.G., B.M., A.S., M.Z.), Lausanne, Switzerland
| | - M Zimmermann
- Siemens Medical Solutions (P.G., B.M., M.Z.), Malvern, Pennsylvania
- Advanced Clinical Imaging Technology (P.G., B.M., M.Z.), Siemens Healthcare, Lausanne, Switzerland
- Department of Radiology (P.G., B.M., M.Z.), Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (P.G., B.M., A.S., M.Z.), Lausanne, Switzerland
- Siemens Healthcare (P.G., B.M., M.Z.), Erlangen, Germany
| | - C D Chen
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
- Charles F. and Joanne Knight Alzheimer Disease Research Center (F.R., S.J., C.A.R., W.W., A.N., C.D.C., T.L.S.B.), Washington University in St. Louis, St. Lous, Missouri
| | - S Keefe
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
| | - P LaMontagne
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
| | - T L S Benzinger
- From the Mallinckrodt Institute of Radiology, Division of Neuroradiology (F.R., S.J., C.A.R., W.W., A.N., G.G.P.-C., M.M.M.-T., C.D.C., S.K., P.L., T.L.S.B.)
- Charles F. and Joanne Knight Alzheimer Disease Research Center (F.R., S.J., C.A.R., W.W., A.N., C.D.C., T.L.S.B.), Washington University in St. Louis, St. Lous, Missouri
| |
Collapse
|
39
|
Sprung J, Laporta ML, Knopman DS, Petersen RC, Mielke MM, Jack CR, Martin DP, Hanson AC, Schroeder DR, Schulte PJ, Przybelski SA, Valencia Morales DJ, Weingarten TN, Vemuri P, Warner DO. Association of Indication for Hospitalization With Subsequent Amyloid Positron Emission Tomography and Magnetic Resonance Imaging Biomarkers. J Gerontol A Biol Sci Med Sci 2023; 78:304-313. [PMID: 35279026 PMCID: PMC9951063 DOI: 10.1093/gerona/glac064] [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: 01/09/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Hospitalization in older age is associated with accelerated cognitive decline, typically preceded by neuropathologic changes. We assess the association between indication for hospitalization and brain neurodegeneration. METHODS Included were participants from the Mayo Clinic Study of Aging, a population-based longitudinal study, with ≥1 brain imaging available in those older than 60 years of age between 2004 and 2017. Primary analyses used linear mixed-effects models to assess association of hospitalization with changes in longitudinal trajectory of cortical thinning, amyloid accumulation, and white matter hyperintensities (WMH). Additional analyses were performed with imaging outcomes dichotomized (normal vs abnormal) using Cox proportional hazards regression. RESULTS Of 2 480 participants, 1 966 had no hospitalization and 514 had ≥1 admission. Hospitalization was associated with accelerated cortical thinning (annual slope change -0.003 mm [95% confidence interval (CI) -0.005 to -0.001], p = .002), but not amyloid accumulation (0.003 [95% CI -0.001 to 0.006], p = .107), or WMH increase (0.011 cm3 [95% CI -0.001 to 0.023], p = .062). Interaction analyses assessing whether trajectory changes are dependent on admission type (medical vs surgical) found interactions for all outcomes. While surgical hospitalizations were not, medical hospitalizations were associated with accelerated cortical thinning (-0.004 mm [95% CI -0.008 to -0.001, p = .014); amyloid accumulation (0.010, [95% CI 0.002 to 0.017, p = .011), and WMH increase (0.035 cm3 [95% CI 0.012 to 0.058, p = .006). Hospitalization was not associated with developing abnormal cortical thinning (p = .407), amyloid accumulation (p = .596), or WMH/infarctions score (p = .565). CONCLUSIONS Medical hospitalizations were associated with accelerated cortical thinning, amyloid accumulation, and WMH increases. These changes were modest and did not translate to increased risk for crossing the abnormality threshold.
Collapse
Affiliation(s)
- Juraj Sprung
- Address correspondence to: Juraj Sprung, MD, PhD, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA. E-mail:
| | - Mariana L Laporta
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Michelle M Mielke
- Division of Epidemiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - David P Martin
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew C Hanson
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Darrell R Schroeder
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Phillip J Schulte
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Toby N Weingarten
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - David O Warner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
40
|
Goyal MS, Blazey T, Metcalf NV, McAvoy MP, Strain JF, Rahmani M, Durbin TJ, Xiong C, Benzinger TLS, Morris JC, Raichle ME, Vlassenko AG. Brain aerobic glycolysis and resilience in Alzheimer disease. Proc Natl Acad Sci U S A 2023; 120:e2212256120. [PMID: 36745794 PMCID: PMC9963219 DOI: 10.1073/pnas.2212256120] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 01/04/2023] [Indexed: 02/08/2023] Open
Abstract
The distribution of brain aerobic glycolysis (AG) in normal young adults correlates spatially with amyloid-beta (Aβ) deposition in individuals with symptomatic and preclinical Alzheimer disease (AD). Brain AG decreases with age, but the functional significance of this decrease with regard to the development of AD symptomatology is poorly understood. Using PET measurements of regional blood flow, oxygen consumption, and glucose utilization-from which we derive AG-we find that cognitive impairment is strongly associated with loss of the typical youthful pattern of AG. In contrast, amyloid positivity without cognitive impairment was associated with preservation of youthful brain AG, which was even higher than that seen in cognitively unimpaired, amyloid negative adults. Similar findings were not seen for blood flow nor oxygen consumption. Finally, in cognitively unimpaired adults, white matter hyperintensity burden was found to be specifically associated with decreased youthful brain AG. Our results suggest that AG may have a role in the resilience and/or response to early stages of amyloid pathology and that age-related white matter disease may impair this process.
Collapse
Affiliation(s)
- Manu S. Goyal
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
| | - Tyler Blazey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Nicholas V. Metcalf
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Mark P. McAvoy
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO63108
| | - Jeremy F. Strain
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Maryam Rahmani
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Tony J. Durbin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
| | - Tammie L.-S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
| | - Marcus E. Raichle
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO63110
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO63130
- Department of Psychology & Brain Science, Washington University School of Medicine, St. Louis, MO63130
| | - Andrei G. Vlassenko
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO63110
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO63110
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO63108
| |
Collapse
|
41
|
Smith AM, Obuchowski NA, Foster NL, Klein G, Mozley PD, Lammertsma AA, Wahl RL, Sunderland JJ, Vanderheyden JL, Benzinger TLS, Kinahan PE, Wong DF, Perlman ES, Minoshima S, Matthews D. The RSNA QIBA Profile for Amyloid PET as an Imaging Biomarker for Cerebral Amyloid Quantification. J Nucl Med 2023; 64:294-303. [PMID: 36137760 PMCID: PMC9902844 DOI: 10.2967/jnumed.122.264031] [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: 02/16/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 02/04/2023] Open
Abstract
A standardized approach to acquiring amyloid PET images increases their value as disease and drug response biomarkers. Most 18F PET amyloid brain scans often are assessed only visually (per regulatory labels), with a binary decision indicating the presence or absence of Alzheimer disease amyloid pathology. Minimizing technical variance allows precise, quantitative SUV ratios (SUVRs) for early detection of β-amyloid plaques and allows the effectiveness of antiamyloid treatments to be assessed with serial studies. Methods: The Quantitative Imaging Biomarkers Alliance amyloid PET biomarker committee developed and validated a profile to characterize and reduce the variability of SUVRs, increasing statistical power for these assessments. Results: On achieving conformance, sites can justify a claim that brain amyloid burden reflected by the SUVR is measurable to a within-subject coefficient of variation of no more than 1.94% when the same radiopharmaceutical, scanner, acquisition, and analysis protocols are used. Conclusion: This overview explains the claim, requirements, barriers, and potential future developments of the profile to achieve precision in clinical and research amyloid PET imaging.
Collapse
Affiliation(s)
- Anne M Smith
- Siemens Medical Solutions USA, Inc., Knoxville, Tennessee;
| | | | - Norman L Foster
- Department of Neurology, University of Utah, Salt Lake City, Utah
| | | | - P David Mozley
- Weill Medical College of Cornell University, New York, New York
| | - Adriaan A Lammertsma
- Amsterdam Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Richard L Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University in Saint Louis, St. Louis, Missouri
| | - John J Sunderland
- Division of Nuclear Medicine, Department of Radiology, University of Iowa, Iowa City, Iowa
| | | | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiation Oncology, Washington University in Saint Louis, St. Louis, Missouri
| | - Paul E Kinahan
- Department of Radiology, School of Medicine, University of Washington, Seattle, Washington
| | - Dean F Wong
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah; and
| | | |
Collapse
|
42
|
Millar PR, Gordon BA, Luckett PH, Benzinger TLS, Cruchaga C, Fagan AM, Hassenstab JJ, Perrin RJ, Schindler SE, Allegri RF, Day GS, Farlow MR, Mori H, Nübling G, Bateman RJ, Morris JC, Ances BM. Multimodal brain age estimates relate to Alzheimer disease biomarkers and cognition in early stages: a cross-sectional observational study. eLife 2023; 12:e81869. [PMID: 36607335 PMCID: PMC9988262 DOI: 10.7554/elife.81869] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023] Open
Abstract
Background Estimates of 'brain-predicted age' quantify apparent brain age compared to normative trajectories of neuroimaging features. The brain age gap (BAG) between predicted and chronological age is elevated in symptomatic Alzheimer disease (AD) but has not been well explored in presymptomatic AD. Prior studies have typically modeled BAG with structural MRI, but more recently other modalities, including functional connectivity (FC) and multimodal MRI, have been explored. Methods We trained three models to predict age from FC, structural (S), or multimodal MRI (S+FC) in 390 amyloid-negative cognitively normal (CN/A-) participants (18-89 years old). In independent samples of 144 CN/A-, 154 CN/A+, and 154 cognitively impaired (CI; CDR > 0) participants, we tested relationships between BAG and AD biomarkers of amyloid and tau, as well as a global cognitive composite. Results All models predicted age in the control training set, with the multimodal model outperforming the unimodal models. All three BAG estimates were significantly elevated in CI compared to controls. FC-BAG was significantly reduced in CN/A+ participants compared to CN/A-. In CI participants only, elevated S-BAG and S+FC BAG were associated with more advanced AD pathology and lower cognitive performance. Conclusions Both FC-BAG and S-BAG are elevated in CI participants. However, FC and structural MRI also capture complementary signals. Specifically, FC-BAG may capture a unique biphasic response to presymptomatic AD pathology, while S-BAG may capture pathological progression and cognitive decline in the symptomatic stage. A multimodal age-prediction model improves sensitivity to healthy age differences. Funding This work was supported by the National Institutes of Health (P01-AG026276, P01- AG03991, P30-AG066444, 5-R01-AG052550, 5-R01-AG057680, 1-R01-AG067505, 1S10RR022984-01A1, and U19-AG032438), the BrightFocus Foundation (A2022014F), and the Alzheimer's Association (SG-20-690363-DIAN).
Collapse
Affiliation(s)
- Peter R Millar
- Department of Neurology, Washington University in St. LouisSt LouisUnited States
| | - Brian A Gordon
- Department of Radiology, Washington University in St. LouisSt LouisUnited States
| | - Patrick H Luckett
- Department of Neurosurgery, Washington University in St. LouisSt LouisUnited States
| | - Tammie LS Benzinger
- Department of Radiology, Washington University in St. LouisSt LouisUnited States
- Department of Neurosurgery, Washington University in St. LouisSt LouisUnited States
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. LouisSt LouisUnited States
| | - Anne M Fagan
- Department of Neurology, Washington University in St. LouisSt LouisUnited States
| | - Jason J Hassenstab
- Department of Neurology, Washington University in St. LouisSt LouisUnited States
| | - Richard J Perrin
- Department of Neurology, Washington University in St. LouisSt LouisUnited States
- Department of Pathology and Immunology, Washington University in St. LouisSt LouisUnited States
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St. LouisSt LouisUnited States
| | - Ricardo F Allegri
- Department of Cognitive Neurology, Institute for Neurological Research (FLENI)Buenos AiresArgentina
| | - Gregory S Day
- Department of Neurology, Mayo Clinic FloridaJacksonvilleUnited States
| | - Martin R Farlow
- Department of Neurology, Indiana University School of MedicineIndianapolisUnited States
| | - Hiroshi Mori
- Department of Clinical Neuroscience, Osaka Metropolitan University Medical School, Nagaoka Sutoku UniversityOsakaJapan
| | - Georg Nübling
- Department of Neurology, Ludwig-Maximilians UniversityMunichGermany
- German Center for Neurodegenerative DiseasesMunichGermany
| | - Randall J Bateman
- Department of Neurology, Washington University in St. LouisSt LouisUnited States
| | - John C Morris
- Department of Neurology, Washington University in St. LouisSt LouisUnited States
| | - Beau M Ances
- Department of Neurology, Washington University in St. LouisSt LouisUnited States
- Department of Radiology, Washington University in St. LouisSt LouisUnited States
| |
Collapse
|
43
|
Chen CD, Ponisio MR, Lang JA, Flores S, Schindler SE, Fagan AM, Morris JC, Benzinger TL. Comparing Tau PET Visual Interpretation with Tau PET Quantification, Cerebrospinal Fluid Biomarkers, and Longitudinal Clinical Assessment. J Alzheimers Dis 2023; 93:765-777. [PMID: 37092225 PMCID: PMC10200228 DOI: 10.3233/jad-230032] [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: 03/14/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND 18F-flortaucipir PET received FDA approval to visualize aggregated neurofibrillary tangles (NFTs) in brains of adult patients with cognitive impairment being evaluated for Alzheimer's disease (AD). However, manufacturer's guidelines for visual interpretation of 18F-flortaucipir PET differ from how 18F-flortaucipir PET has been measured in research settings using standardized uptake value ratios (SUVRs). How visual interpretation relates to 18F-flortaucipir PET SUVR, cerebrospinal fluid (CSF) biomarkers, or longitudinal clinical assessment is not well understood. OBJECTIVE We compare various diagnostic methods in participants enrolled in longitudinal observational studies of aging and memory (n = 189, 23 were cognitively impaired). METHODS Participants had tau PET, Aβ PET, MRI, and clinical and cognitive evaluation within 18 months (n = 189); the majority (n = 144) also underwent lumbar puncture. Two radiologists followed manufacturer's guidelines for 18F-flortaucipir PET visual interpretation. RESULTS Visual interpretation had high agreement with SUVR (98.4%)and moderate agreement with CSF p-tau181 (86.1%). Two participants demonstrated 18F-flortaucipir uptake from meningiomas. Visual interpretation could not predict follow-up clinical assessment in 9.52% of cases. CONCLUSION Visual interpretation was highly consistent with SUVR (discordant participants had hemorrhagic infarcts or occipital-predominant AD NFT deposition) and moderately consistent with CSF p-tau181 (discordant participants had AD pathophysiology not detectable on tau PET). However, close association between AD NFT deposition and clinical onset in group-level studies does not necessarily hold at the individual level, with discrepancies arising from atypical AD, vascular dementia, or frontotemporal dementia. A better understanding of relationships across imaging, CSF biomarkers, and clinical assessment is needed to provide appropriate diagnoses for these individuals.
Collapse
Affiliation(s)
- Charles D. Chen
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Maria Rosana Ponisio
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan A. Lang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Anne M. Fagan
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - John C. Morris
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
44
|
Long JM, Coble DW, Xiong C, Schindler SE, Perrin RJ, Gordon BA, Benzinger TLS, Grant E, Fagan AM, Harari O, Cruchaga C, Holtzman DM, Morris JC. Preclinical Alzheimer's disease biomarkers accurately predict cognitive and neuropathological outcomes. Brain 2022; 145:4506-4518. [PMID: 35867858 PMCID: PMC10200309 DOI: 10.1093/brain/awac250] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/30/2022] [Accepted: 07/20/2022] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease biomarkers are widely accepted as surrogate markers of underlying neuropathological changes. However, few studies have evaluated whether preclinical Alzheimer's disease biomarkers predict Alzheimer's neuropathology at autopsy. We sought to determine whether amyloid PET imaging or CSF biomarkers accurately predict cognitive outcomes and Alzheimer's disease neuropathological findings. This study included 720 participants, 42-91 years of age, who were enrolled in longitudinal studies of memory and aging in the Washington University Knight Alzheimer Disease Research Center and were cognitively normal at baseline, underwent amyloid PET imaging and/or CSF collection within 1 year of baseline clinical assessment, and had subsequent clinical follow-up. Cognitive status was assessed longitudinally by Clinical Dementia Rating®. Biomarker status was assessed using predefined cut-offs for amyloid PET imaging or CSF p-tau181/amyloid-β42. Subsequently, 57 participants died and underwent neuropathologic examination. Alzheimer's disease neuropathological changes were assessed using standard criteria. We assessed the predictive value of Alzheimer's disease biomarker status on progression to cognitive impairment and for presence of Alzheimer's disease neuropathological changes. Among cognitively normal participants with positive biomarkers, 34.4% developed cognitive impairment (Clinical Dementia Rating > 0) as compared to 8.4% of those with negative biomarkers. Cox proportional hazards modelling indicated that preclinical Alzheimer's disease biomarker status, APOE ɛ4 carrier status, polygenic risk score and centred age influenced risk of developing cognitive impairment. Among autopsied participants, 90.9% of biomarker-positive participants and 8.6% of biomarker-negative participants had Alzheimer's disease neuropathological changes. Sensitivity was 87.0%, specificity 94.1%, positive predictive value 90.9% and negative predictive value 91.4% for detection of Alzheimer's disease neuropathological changes by preclinical biomarkers. Single CSF and amyloid PET baseline biomarkers were also predictive of Alzheimer's disease neuropathological changes, as well as Thal phase and Braak stage of pathology at autopsy. Biomarker-negative participants who developed cognitive impairment were more likely to exhibit non-Alzheimer's disease pathology at autopsy. The detection of preclinical Alzheimer's disease biomarkers is strongly predictive of future cognitive impairment and accurately predicts presence of Alzheimer's disease neuropathology at autopsy.
Collapse
Affiliation(s)
- Justin M Long
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Dean W Coble
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Chengjie Xiong
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Suzanne E Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Richard J Perrin
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Brian A Gordon
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Elizabeth Grant
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Division of Biostatistics, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Anne M Fagan
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Oscar Harari
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Carlos Cruchaga
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - David M Holtzman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| |
Collapse
|
45
|
Shah J, Gao F, Li B, Ghisays V, Luo J, Chen Y, Lee W, Zhou Y, Benzinger TL, Reiman EM, Chen K, Su Y, Wu T. Deep residual inception encoder-decoder network for amyloid PET harmonization. Alzheimers Dement 2022; 18:2448-2457. [PMID: 35142053 PMCID: PMC9360199 DOI: 10.1002/alz.12564] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Multiple positron emission tomography (PET) tracers are available for amyloid imaging, posing a significant challenge to consensus interpretation and quantitative analysis. We accordingly developed and validated a deep learning model as a harmonization strategy. METHOD A Residual Inception Encoder-Decoder Neural Network was developed to harmonize images between amyloid PET image pairs made with Pittsburgh Compound-B and florbetapir tracers. The model was trained using a dataset with 92 subjects with 10-fold cross validation and its generalizability was further examined using an independent external dataset of 46 subjects. RESULTS Significantly stronger between-tracer correlations (P < .001) were observed after harmonization for both global amyloid burden indices and voxel-wise measurements in the training cohort and the external testing cohort. DISCUSSION We proposed and validated a novel encoder-decoder based deep model to harmonize amyloid PET imaging data from different tracers. Further investigation is ongoing to improve the model and apply to additional tracers.
Collapse
Affiliation(s)
- Jay Shah
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
| | - Fei Gao
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
| | - Baoxin Li
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
| | - Valentina Ghisays
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Ji Luo
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Yinghua Chen
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Wendy Lee
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Yuxiang Zhou
- Department of RadiologyMayo Clinic at Arizona5777 E Mayo BlvdPhoenixArizona85054USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University School of Medicine in St. Louis510 South Kingshighway BoulevardSt. LouisMissouri63110USA
| | - Eric M. Reiman
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Kewei Chen
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Yi Su
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
- Banner Alzheimer's Institute901 E. Willetta StreetPhoenixArizona85006USA
| | - Teresa Wu
- ASU‐Mayo Center for Innovative ImagingArizona State University699 S. Mill Ave.TempeArizona85287USA
- School of Computing and Augmented IntelligenceArizona State University699 S. Mill Ave.TempeArizona85287USA
| |
Collapse
|
46
|
Xiong C, Luo J, Schindler SE, Fagan AM, Benzinger T, Hassenstab J, Balls-Berry JE, Agboola F, Grant E, Moulder KL, Morris JC. Racial differences in longitudinal Alzheimer's disease biomarkers among cognitively normal adults. Alzheimers Dement 2022; 18:2570-2581. [PMID: 35218143 PMCID: PMC9402805 DOI: 10.1002/alz.12608] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/28/2021] [Accepted: 01/01/2022] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Longitudinal changes in Alzheimer's disease (AD) biomarkers, including cerebrospinal fluid (CSF) analytes, amyloid uptakes from positron emission tomography (PET), structural outcomes from magnetic resonance imaging (MRI), and cognition, have not been compared between Blacks and Whites. METHODS A total of 179 Blacks and 1180 Whites who were cognitively normal at baseline and had longitudinal data from at least one biomarker modality were analyzed for the annual rates of change. RESULTS CSF amyloid beta (Aβ)42/Aβ40 declined more slowly (P = .0390), and amyloid (PET) accumulated more slowly (P = .0157), in Blacks than Whites. CSF Aβ42 changed in opposite directions over time between Blacks and Whites (P = .0039). The annual increase in CSF total tau and phosphorylated tau181 for Blacks was about half of that for Whites. DISCUSSION Longitudinal racial differences in amyloid biomarkers are observed. It will be important to comprehensively and prospectively examine the effects of apolipoprotein E genotype and sociocultural factors on these differences.
Collapse
Affiliation(s)
- Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jingqin Luo
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Suzanne E. Schindler
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M. Fagan
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joyce E. Balls-Berry
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Folasade Agboola
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth Grant
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Krista L. Moulder
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
47
|
Dincer A, Chen CD, McKay NS, Koenig LN, McCullough A, Flores S, Keefe SJ, Schultz SA, Feldman RL, Joseph-Mathurin N, Hornbeck RC, Cruchaga C, Schindler SE, Holtzman DM, Morris JC, Fagan AM, Benzinger TLS, Gordon BA. APOE ε4 genotype, amyloid-β, and sex interact to predict tau in regions of high APOE mRNA expression. Sci Transl Med 2022; 14:eabl7646. [PMID: 36383681 PMCID: PMC9912474 DOI: 10.1126/scitranslmed.abl7646] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The apolipoprotein E (APOE) ε4 allele is strongly linked with cerebral β-amyloidosis, but its relationship with tauopathy is less established. We investigated the relationship between APOE ε4 carrier status, regional amyloid-β (Aβ), magnetic resonance imaging (MRI) volumetrics, tau positron emission tomography (PET), APOE messenger RNA (mRNA) expression maps, and cerebrospinal fluid phosphorylated tau (CSF ptau181). Three hundred fifty participants underwent imaging, and 270 had ptau181. We used computational models to evaluate the main effect of APOE ε4 carrier status on regional neuroimaging values and then the interaction of ε4 status and global Aβ on regional tau PET and brain volumes as well as CSF ptau181. Separately, we also examined the additional interactive influence of sex. We found that, for the same degree of Aβ burden, APOE ε4 carriers showed greater tau PET signal relative to noncarriers in temporal regions, but no interaction was present for MRI volumes or CSF ptau181. This potentiation of tau aggregation irrespective of sex occurred in brain regions with high APOE mRNA expression, suggesting local vulnerabilities to tauopathy. There were greater effects of APOE genotype in females, although the interactive sex effects did not strongly mirror mRNA expression. Pathology is not homogeneously expressed throughout the brain but mirrors underlying biological patterns such as gene expression.
Collapse
Affiliation(s)
- Aylin Dincer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nicole S McKay
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Lauren N Koenig
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sarah J Keefe
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stephanie A Schultz
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rebecca L Feldman
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Suzanne E Schindler
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - David M Holtzman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Anne M Fagan
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tammie LS Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Psychological & Brain Sciences, Washington University, Saint Louis, MO, USA
| |
Collapse
|
48
|
Bourgeat P, Doré V, Burnham SC, Benzinger T, Tosun D, Li S, Goyal M, LaMontagne P, Jin L, Rowe CC, Weiner MW, Morris JC, Masters CL, Fripp J, Villemagne VL. β-amyloid PET harmonisation across longitudinal studies: Application to AIBL, ADNI and OASIS3. Neuroimage 2022; 262:119527. [PMID: 35917917 PMCID: PMC9550562 DOI: 10.1016/j.neuroimage.2022.119527] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 10/31/2022] Open
Abstract
INTRODUCTION The Centiloid scale was developed to harmonise the quantification of β-amyloid (Aβ) PET images across tracers, scanners, and processing pipelines. However, several groups have reported differences across tracers and scanners even after centiloid conversion. In this study, we aim to evaluate the impact of different pre and post-processing harmonisation steps on the robustness of longitudinal Centiloid data across three large international cohort studies. METHODS All Aβ PET data in AIBL (N = 3315), ADNI (N = 3442) and OASIS3 (N = 1398) were quantified using the MRI-based Centiloid standard SPM pipeline and the PET-only pipeline CapAIBL. SUVR were converted into Centiloids using each tracer's respective transform. Global Aβ burden from pre-defined target cortical regions in Centiloid units were quantified for both raw PET scans and PET scans smoothed to a uniform 8 mm full width half maximum (FWHM) effective smoothness. For Florbetapir, we assessed the performance of using both the standard Whole Cerebellum (WCb) and a composite white matter (WM)+WCb reference region. Additionally, our recently proposed quantification based on Non-negative Matrix Factorisation (NMF) was applied to all spatially and SUVR normalised images. Correlation with clinical severity measured by the Mini-Mental State Examination (MMSE) and effect size, as well as tracer agreement in 11C-PiB-18F-Florbetapir pairs and longitudinal consistency were evaluated. RESULTS The smoothing to a uniform resolution partially reduced longitudinal variability, but did not improve inter-tracer agreement, effect size or correlation with MMSE. Using a Composite reference region for 18F-Florbetapir improved inter-tracer agreement, effect size, correlation with MMSE, and longitudinal consistency. The best results were however obtained when using the NMF method which outperformed all other quantification approaches in all metrics used. CONCLUSIONS FWHM smoothing has limited impact on longitudinal consistency or outliers. A Composite reference region including subcortical WM should be used for computing both cross-sectional and longitudinal Florbetapir Centiloid. NMF improves Centiloid quantification on all metrics examined.
Collapse
Affiliation(s)
| | - Vincent Doré
- CSIRO Health and Biosecurity, Brisbane, Australia; Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia
| | | | | | - Duygu Tosun
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA,; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Shenpeng Li
- CSIRO Health and Biosecurity, Brisbane, Australia
| | - Manu Goyal
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, USA
| | - Liang Jin
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Michael W Weiner
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA,; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - John C Morris
- Washington University in St. Louis, St. Louis, MO, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Brisbane, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia; Department of Psychiatry, The University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
49
|
Rezai AR, Ranjan M, Haut MW, Carpenter J, D’Haese PF, Mehta RI, Najib U, Wang P, Claassen DO, Chazen JL, Krishna V, Deib G, Zibly Z, Hodder SL, Wilhelmsen KC, Finomore V, Konrad PE, Kaplitt M, _ _. Focused ultrasound–mediated blood-brain barrier opening in Alzheimer’s disease: long-term safety, imaging, and cognitive outcomes. J Neurosurg 2022:1-9. [DOI: 10.3171/2022.9.jns221565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE
MRI-guided low-intensity focused ultrasound (FUS) has been shown to reversibly open the blood-brain barrier (BBB), with the potential to deliver therapeutic agents noninvasively to target brain regions in patients with Alzheimer’s disease (AD) and other neurodegenerative conditions. Previously, the authors reported the short-term safety and feasibility of FUS BBB opening of the hippocampus and entorhinal cortex (EC) in patients with AD. Given the need to treat larger brain regions beyond the hippocampus and EC, brain volumes and locations treated with FUS have now expanded. To evaluate any potential adverse consequences of BBB opening on disease progression, the authors report safety, imaging, and clinical outcomes among participants with mild AD at 6–12 months after FUS treatment targeted to the hippocampus, frontal lobe, and parietal lobe.
METHODS
In this open-label trial, participants with mild AD underwent MRI-guided FUS sonication to open the BBB in β-amyloid positive regions of the hippocampus, EC, frontal lobe, and parietal lobe. Participants underwent 3 separate FUS treatment sessions performed 2 weeks apart. Outcome assessments included safety, imaging, neurological, cognitive, and florbetaben β-amyloid PET.
RESULTS
Ten participants (range 55–76 years old) completed 30 separate FUS treatments at 2 participating institutions, with 6–12 months of follow-up. All participants had immediate BBB opening after FUS and BBB closure within 24–48 hours. All FUS treatments were well tolerated, with no serious adverse events related to the procedure. All 10 participants had a minimum of 6 months of follow-up, and 7 participants had a follow-up out to 1 year. Changes in the Alzheimer’s Disease Assessment Scale–cognitive and Mini-Mental State Examination scores were comparable to those in controls from the Alzheimer’s Disease Neuroimaging Initiative. PET scans demonstrated an average β-amyloid plaque of 14% in the Centiloid scale in the FUS-treated regions.
CONCLUSIONS
This study is the largest cohort of participants with mild AD who received FUS treatment, and has the longest follow-up to date. Safety was demonstrated in conjunction with reversible and repeated BBB opening in multiple cortical and deep brain locations, with a concomitant reduction of β-amyloid. There was no apparent cognitive worsening beyond expectations up to 1 year after FUS treatment, suggesting that the BBB opening treatment in multiple brain regions did not adversely influence AD progression. Further studies are needed to determine the clinical significance of these findings. FUS offers a unique opportunity to decrease amyloid plaque burden as well as the potential to deliver targeted therapeutics to multiple brain regions in patients with neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | - Marc W. Haut
- Behavioral Medicine and Psychiatry,
- Neurology, and
| | - Jeffrey Carpenter
- Neuroradiology, WVU Rockefeller Neuroscience Institute, Morgantown, West Virginia
| | | | - Rashi I. Mehta
- Neuroradiology, WVU Rockefeller Neuroscience Institute, Morgantown, West Virginia
| | | | - Peng Wang
- Neuroradiology, WVU Rockefeller Neuroscience Institute, Morgantown, West Virginia
| | | | | | - Vibhor Krishna
- Department of Neurosurgery, University of North Carolina, Chapel Hill, North Carolina
| | - Gerard Deib
- Neuroradiology, WVU Rockefeller Neuroscience Institute, Morgantown, West Virginia
| | - Zion Zibly
- Department of Neurosurgery, Sheba Medical Center, Ramat Gan, Israel; and
| | - Sally L. Hodder
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown, West Virginia
| | | | | | | | - Michael Kaplitt
- Neurological Surgery, Weill Cornell Medical College, New York, New York
| | | |
Collapse
|
50
|
Morris JC, Weiner M, Xiong C, Beckett L, Coble D, Saito N, Aisen PS, Allegri R, Benzinger TLS, Berman SB, Cairns NJ, Carrillo MC, Chui HC, Chhatwal JP, Cruchaga C, Fagan AM, Farlow M, Fox NC, Ghetti B, Goate AM, Gordon BA, Graff-Radford N, Day GS, Hassenstab J, Ikeuchi T, Jack CR, Jagust WJ, Jucker M, Levin J, Massoumzadeh P, Masters CL, Martins R, McDade E, Mori H, Noble JM, Petersen RC, Ringman JM, Salloway S, Saykin AJ, Schofield PR, Shaw LM, Toga AW, Trojanowski JQ, Vöglein J, Weninger S, Bateman RJ, Buckles VD. Autosomal dominant and sporadic late onset Alzheimer's disease share a common in vivo pathophysiology. Brain 2022; 145:3594-3607. [PMID: 35580594 PMCID: PMC9989348 DOI: 10.1093/brain/awac181] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
The extent to which the pathophysiology of autosomal dominant Alzheimer's disease corresponds to the pathophysiology of 'sporadic' late onset Alzheimer's disease is unknown, thus limiting the extrapolation of study findings and clinical trial results in autosomal dominant Alzheimer's disease to late onset Alzheimer's disease. We compared brain MRI and amyloid PET data, as well as CSF concentrations of amyloid-β42, amyloid-β40, tau and tau phosphorylated at position 181, in 292 carriers of pathogenic variants for Alzheimer's disease from the Dominantly Inherited Alzheimer Network, with corresponding data from 559 participants from the Alzheimer's Disease Neuroimaging Initiative. Imaging data and CSF samples were reprocessed as appropriate to guarantee uniform pipelines and assays. Data analyses yielded rates of change before and after symptomatic onset of Alzheimer's disease, allowing the alignment of the ∼30-year age difference between the cohorts on a clinically meaningful anchor point, namely the participant age at symptomatic onset. Biomarker profiles were similar for both autosomal dominant Alzheimer's disease and late onset Alzheimer's disease. Both groups demonstrated accelerated rates of decline in cognitive performance and in regional brain volume loss after symptomatic onset. Although amyloid burden accumulation as determined by PET was greater after symptomatic onset in autosomal dominant Alzheimer's disease than in late onset Alzheimer's disease participants, CSF assays of amyloid-β42, amyloid-β40, tau and p-tau181 were largely overlapping in both groups. Rates of change in cognitive performance and hippocampal volume loss after symptomatic onset were more aggressive for autosomal dominant Alzheimer's disease participants. These findings suggest a similar pathophysiology of autosomal dominant Alzheimer's disease and late onset Alzheimer's disease, supporting a shared pathobiological construct.
Collapse
Affiliation(s)
- John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Weiner
- Department of Radiology, University of California at San Francisco, San Francisco, CA, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Laurel Beckett
- Department of Public Health Sciences, School of Medicine, University of California; Davis, Davis, CA, USA
| | - Dean Coble
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Naomi Saito
- Department of Public Health Sciences, School of Medicine, University of California; Davis, Davis, CA, USA
| | - Paul S Aisen
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ricardo Allegri
- Department of Cognitive Neurology, Neuropsychology and Neuropsychiatry, Institute for Neurological Research (FLENI), Buenos Aires, Argentina
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah B Berman
- Department of Neurology and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nigel J Cairns
- College of Medicine and Health and the Living Systems Institute, University of Exeter, Exeter, UK
| | | | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Martin Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nick C Fox
- Department of Neurodegenerative Disease and UK Dementia Research Institute, UCL Institute of Neurology, London, UK
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Mathias Jucker
- Cell Biology of Neurological Diseases Group, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Johannes Levin
- DZNE Munich, Munich Cluster of Systems Neurology (SyNergy) and Ludwig-Maximilians-Universität, Munich, Germany
| | - Parinaz Massoumzadeh
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Colin L Masters
- Florey Institute, University of Melbourne, Melbourne, Australia
| | - Ralph Martins
- Sir James McCusker Alzheimer’s Disease Research Unit, Edith Cowan University, Nedlands, Australia
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hiroshi Mori
- Department of Neuroscience, Osaka City University Medical School, Osaka City, Japan
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | | | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephen Salloway
- Department of Neurology, Butler Hospital and Alpert Medical School of Brown University, Providence, RI, 02906, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter R Schofield
- Neuroscience Research Australia and School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases (DZNE) and Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Virginia D Buckles
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|