1
|
Mathies FL, Heeman F, Visser PJ, den Braber A, Yaqub M, Klutmann S, Schöll M, van de Giessen E, Collij LE, Buchert R. The Early Perfusion Image Is Useful to Support the Visual Interpretation of Brain Amyloid-PET With 18F-Flutemetamol in Borderline Cases. Clin Nucl Med 2024; 49:838-846. [PMID: 39102811 DOI: 10.1097/rlu.0000000000005360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
PURPOSE Visual interpretation of brain amyloid-β (Aβ) PET can be difficult in individuals with borderline Aβ burden. Coregistration with individual MRI is recommended in these cases, which, however, is not always available. This study evaluated coregistration with the early perfusion frames acquired immediately after tracer injection to support the visual interpretation of the late Aβ-frames in PET with 18F-flutemetamol (FMM). PATIENTS AND METHODS Fifty dual-time-window FMM-PET scans of cognitively normal subjects with 0 to 60 Centiloids were included retrospectively (70.1 ± 6.9 years, 56% female, MMSE score 28.9 ± 1.3, 42% APOE ɛ4 carrier). Regional Aβ load was scored with respect to a 6-point Likert scale by 3 independent raters in the 10 regions of interest recommended for FMM reading using 3 different settings: Aβ image only, Aβ image coregistered with MRI, and Aβ image coregistered with the perfusion image. The impact of setting, within- and between-readers variability, region of interest, and Aβ-status was tested by repeated-measure analysis of variance of the Likert score. RESULTS The Centiloid scale ranged between 2 and 52 (interquartile range, 7-19). Support of visual scoring by the perfusion image resulted in the best discrimination between Aβ-positive and Aβ-negative cases, mainly by improved certainty of excluding Aβ plaques in Aβ-negative cases (P = 0.030). It also resulted in significantly higher between-rater agreement. The setting effect was most pronounced in the frontal lobe and in the posterior cingulate cortex/precuneus area (P = 0.005). CONCLUSIONS The early perfusion image is a suitable alternative to T1-weighted MRI to support the visual interpretation of the late Aβ image in FMM-PET.
Collapse
Affiliation(s)
- Franziska L Mathies
- From the Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | - Susanne Klutmann
- From the Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Ralph Buchert
- From the Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
2
|
Collij LE, Smith A, Buckley C. Degree of amyloid-β burden could be indicative of the primary etiology underlying dementia. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06875-8. [PMID: 39212682 DOI: 10.1007/s00259-024-06875-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Lyduine E Collij
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands.
| | | | | |
Collapse
|
3
|
Kim S, Wang SM, Kang DW, Um YH, Han EJ, Park SY, Ha S, Choe YS, Kim HW, Kim REY, Kim D, Lee CU, Lim HK. A Comparative Analysis of Two Automated Quantification Methods for Regional Cerebral Amyloid Retention: PET-Only and PET-and-MRI-Based Methods. Int J Mol Sci 2024; 25:7649. [PMID: 39062892 PMCID: PMC11276670 DOI: 10.3390/ijms25147649] [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: 06/15/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Accurate quantification of amyloid positron emission tomography (PET) is essential for early detection of and intervention in Alzheimer's disease (AD) but there is still a lack of studies comparing the performance of various automated methods. This study compared the PET-only method and PET-and-MRI-based method with a pre-trained deep learning segmentation model. A large sample of 1180 participants in the Catholic Aging Brain Imaging (CABI) database was analyzed to calculate the regional standardized uptake value ratio (SUVR) using both methods. The logistic regression models were employed to assess the discriminability of amyloid-positive and negative groups through 10-fold cross-validation and area under the receiver operating characteristics (AUROC) metrics. The two methods showed a high correlation in calculating SUVRs but the PET-MRI method, incorporating MRI data for anatomical accuracy, demonstrated superior performance in predicting amyloid-positivity. The parietal, frontal, and cingulate importantly contributed to the prediction. The PET-MRI method with a pre-trained deep learning model approach provides an efficient and precise method for earlier diagnosis and intervention in the AD continuum.
Collapse
Affiliation(s)
- Sunghwan Kim
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sheng-Min Wang
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Dong Woo Kang
- Department of Psychiatry, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yoo Hyun Um
- Department of Psychiatry, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Eun Ji Han
- Division of Nuclear Medicine, Department of Radiology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sonya Youngju Park
- Division of Nuclear Medicine, Department of Radiology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yeong Sim Choe
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Hye Weon Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Regina EY Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Donghyeon Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Chang Uk Lee
- Department of Psychiatry, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
4
|
Salvadó G, Horie K, Barthélemy NR, Vogel JW, Pichet Binette A, Chen CD, Aschenbrenner AJ, Gordon BA, Benzinger TLS, Holtzman DM, Morris JC, Palmqvist S, Stomrud E, Janelidze S, Ossenkoppele R, Schindler SE, Bateman RJ, Hansson O. Disease staging of Alzheimer's disease using a CSF-based biomarker model. NATURE AGING 2024; 4:694-708. [PMID: 38514824 PMCID: PMC11108782 DOI: 10.1038/s43587-024-00599-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2024]
Abstract
Biological staging of individuals with Alzheimer's disease (AD) may improve diagnostic and prognostic workup of dementia in clinical practice and the design of clinical trials. In this study, we used the Subtype and Stage Inference (SuStaIn) algorithm to establish a robust biological staging model for AD using cerebrospinal fluid (CSF) biomarkers. Our analysis involved 426 participants from BioFINDER-2 and was validated in 222 participants from the Knight Alzheimer Disease Research Center cohort. SuStaIn identified a singular biomarker sequence and revealed that five CSF biomarkers effectively constituted a reliable staging model (ordered: Aβ42/40, pT217/T217, pT205/T205, MTBR-tau243 and non-phosphorylated mid-region tau). The CSF stages (0-5) demonstrated a correlation with increased abnormalities in other AD-related biomarkers, such as Aβ-PET and tau-PET, and aligned with longitudinal biomarker changes reflective of AD progression. Higher CSF stages at baseline were associated with an elevated hazard ratio of clinical decline. This study highlights a common molecular pathway underlying AD pathophysiology across all patients, suggesting that a single CSF collection can accurately indicate the presence of AD pathologies and characterize the stage of disease progression. The proposed staging model has implications for enhancing diagnostic and prognostic assessments in both clinical practice and the design of clinical trials.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| | - Kanta Horie
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Eisai, Inc., Nutley, NJ, USA
| | - Nicolas R Barthélemy
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Science, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Charles D Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- 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
- Charles F. and Joanne 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
- Charles F. and Joanne 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
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne 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.
| |
Collapse
|
5
|
Barthélemy NR, Salvadó G, Schindler SE, He Y, Janelidze S, Collij LE, Saef B, Henson RL, Chen CD, Gordon BA, Li Y, La Joie R, Benzinger TLS, Morris JC, Mattsson-Carlgren N, Palmqvist S, Ossenkoppele R, Rabinovici GD, Stomrud E, Bateman RJ, Hansson O. Highly accurate blood test for Alzheimer's disease is similar or superior to clinical cerebrospinal fluid tests. Nat Med 2024; 30:1085-1095. [PMID: 38382645 PMCID: PMC11031399 DOI: 10.1038/s41591-024-02869-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: 10/24/2023] [Accepted: 02/12/2024] [Indexed: 02/23/2024]
Abstract
With the emergence of Alzheimer's disease (AD) disease-modifying therapies, identifying patients who could benefit from these treatments becomes critical. In this study, we evaluated whether a precise blood test could perform as well as established cerebrospinal fluid (CSF) tests in detecting amyloid-β (Aβ) plaques and tau tangles. Plasma %p-tau217 (ratio of phosporylated-tau217 to non-phosphorylated tau) was analyzed by mass spectrometry in the Swedish BioFINDER-2 cohort (n = 1,422) and the US Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC) cohort (n = 337). Matched CSF samples were analyzed with clinically used and FDA-approved automated immunoassays for Aβ42/40 and p-tau181/Aβ42. The primary and secondary outcomes were detection of brain Aβ or tau pathology, respectively, using positron emission tomography (PET) imaging as the reference standard. Main analyses were focused on individuals with cognitive impairment (mild cognitive impairment and mild dementia), which is the target population for available disease-modifying treatments. Plasma %p-tau217 was clinically equivalent to FDA-approved CSF tests in classifying Aβ PET status, with an area under the curve (AUC) for both between 0.95 and 0.97. Plasma %p-tau217 was generally superior to CSF tests in classification of tau-PET with AUCs of 0.95-0.98. In cognitively impaired subcohorts (BioFINDER-2: n = 720; Knight ADRC: n = 50), plasma %p-tau217 had an accuracy, a positive predictive value and a negative predictive value of 89-90% for Aβ PET and 87-88% for tau PET status, which was clinically equivalent to CSF tests, further improving to 95% using a two-cutoffs approach. Blood plasma %p-tau217 demonstrated performance that was clinically equivalent or superior to clinically used FDA-approved CSF tests in the detection of AD pathology. Use of high-performance blood tests in clinical practice can improve access to accurate AD diagnosis and AD-specific treatments.
Collapse
Affiliation(s)
- Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), 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 Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lyduine E Collij
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Benjamin Saef
- 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
| | - Charles D Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, MO, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Gil D Rabinovici
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Tracy Family Stable Isotope Labeling Quantitation (SILQ) Center, Washington University School of Medicine, St. Louis, MO, USA.
- Charles F. and Joanne Knight Alzheimer Disease Research Center (Knight ADRC), Washington University School of Medicine, St. Louis, MO, USA.
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
6
|
Jung NY, Je Y, Ham HG, Park YH, Kim TY, Go MS, Lee HI, Kim DE, Lee MJ, Seo SW, Kim EJ. Comparison of Enlarged Perivascular Spaces in Early-Onset and Late-Onset Alzheimer Disease-related Cognitive Impairment: A Single Clinic-based Study in South Korea. Alzheimer Dis Assoc Disord 2024; 38:201-204. [PMID: 38563235 PMCID: PMC11132089 DOI: 10.1097/wad.0000000000000614] [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: 08/17/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024]
Abstract
We examined whether there were differences in the presence of centrum semiovale-enlarged perivascular spaces (CSO-ePVS) and basal ganglia-ePVS (BG-ePVS) among patients with Alzheimer disease-related cognitive impairment (ADCI) based on their age of onset. Out of a total of 239 patients with cognitive impairment, 155 with positive amyloid-PET results were included. Among these, 43 had early-onset ADCI (EOADCI) and 112 had late-onset ADCI (LOADCI). Patients with LOADCI exhibited a higher prevalence of hypertension, lacunes, white matter hyperintensities, and BG-ePVS than those with EOADCI. BG-ePVS showed a significant correlation with age at the onset and the number of lacunes, whereas CSO-ePVS did not exhibit any association. The higher prevalence of BG-ePVS in patients with LOADCI might be attributable to vascular risk factors (hypertension) and cerebral small vessel disease (CSVD). These findings support the hypothesis that BG-ePVS is associated with CSVD and vascular risk factors, whereas CSO-ePVS is associated with cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan
| | - Yuri Je
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical research institute, Busan
| | - Hong-Gi Ham
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yu Hyun Park
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Tae-Yun Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical research institute, Busan
| | - Min-su Go
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical research institute, Busan
| | - Hye-In Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical research institute, Busan
| | - Da Eun Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical research institute, Busan
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical research institute, Busan
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical research institute, Busan
| |
Collapse
|
7
|
Therriault J, Schindler SE, Salvadó G, Pascoal TA, Benedet AL, Ashton NJ, Karikari TK, Apostolova L, Murray ME, Verberk I, Vogel JW, La Joie R, Gauthier S, Teunissen C, Rabinovici GD, Zetterberg H, Bateman RJ, Scheltens P, Blennow K, Sperling R, Hansson O, Jack CR, Rosa-Neto P. Biomarker-based staging of Alzheimer disease: rationale and clinical applications. Nat Rev Neurol 2024; 20:232-244. [PMID: 38429551 DOI: 10.1038/s41582-024-00942-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Disease staging, whereby the spatial extent and load of brain pathology are used to estimate the severity of Alzheimer disease (AD), is pivotal to the gold-standard neuropathological diagnosis of AD. Current in vivo diagnostic frameworks for AD are based on abnormal concentrations of amyloid-β and tau in the cerebrospinal fluid or on PET scans, and breakthroughs in molecular imaging have opened up the possibility of in vivo staging of AD. Focusing on the key principles of disease staging shared across several areas of medicine, this Review highlights the potential for in vivo staging of AD to transform our understanding of preclinical AD, refine enrolment criteria for trials of disease-modifying therapies and aid clinical decision-making in the era of anti-amyloid therapeutics. We provide a state-of-the-art review of recent biomarker-based AD staging systems and highlight their contributions to the understanding of the natural history of AD. Furthermore, we outline hypothetical frameworks to stage AD severity using more accessible fluid biomarkers. In addition, by applying amyloid PET-based staging to recently published anti-amyloid therapeutic trials, we highlight how biomarker-based disease staging frameworks could illustrate the numerous pathological changes that have already taken place in individuals with mildly symptomatic AD. Finally, we discuss challenges related to the validation and standardization of disease staging and provide a forward-looking perspective on potential clinical applications.
Collapse
Affiliation(s)
- Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andréa Lessa Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- NIHR Biomedical Research Centre, South London and Maudsley NHS Foundation, London, UK
| | - Thomas K Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Liana Apostolova
- Department of Neurology, University of Indiana School of Medicine, Indianapolis, IN, USA
| | | | - Inge Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Charlotte Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Reisa Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Juengling F, Wuest F, Schirrmacher R, Abele J, Thiel A, Soucy JP, Camicioli R, Garibotto V. PET Imaging in Dementia: Mini-Review and Canadian Perspective for Clinical Use. Can J Neurol Sci 2024:1-13. [PMID: 38433571 DOI: 10.1017/cjn.2024.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
PET imaging is increasingly recognized as an important diagnostic tool to investigate patients with cognitive disturbances of possible neurodegenerative origin. PET with 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG), assessing glucose metabolism, provides a measure of neurodegeneration and allows a precise differential diagnosis among the most common neurodegenerative diseases, such as Alzheimer's disease, frontotemporal dementia or dementia with Lewy bodies. PET tracers specific for the pathological deposits characteristic of different neurodegenerative processes, namely amyloid and tau deposits typical of Alzheimer's Disease, allow the visualization of these aggregates in vivo. [18F]FDG and amyloid PET imaging have reached a high level of clinical validity and are since 2022 investigations that can be offered to patients in standard clinical care in most of Canada.This article will briefly review and summarize the current knowledge on these diagnostic tools, their integration into diagnostic algorithms as well as perspectives for future developments.
Collapse
Affiliation(s)
- Freimut Juengling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Faculty, University of Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ralf Schirrmacher
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Jonathan Abele
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Department of Neurology and Neurosurgery, Lady Davis Institute for Medical Research, McGill University, Montréal, QC, Canada
| | - Jean-Paul Soucy
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Valentina Garibotto
- Diagnostic Department, Nuclear Medicine and Molecular Imaging Division, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
9
|
Moonen JEF, Haan R, Bos I, Teunissen C, van de Giessen E, Tomassen J, den Braber A, van der Landen SM, de Geus EJC, Legdeur N, van Harten AC, Trieu C, de Boer C, Kroeze L, Barkhof F, Visser PJ, van der Flier WM. Contributions of amyloid beta and cerebral small vessel disease in clinical decline. Alzheimers Dement 2024; 20:1868-1880. [PMID: 38146222 PMCID: PMC10984432 DOI: 10.1002/alz.13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/27/2023]
Abstract
INTRODUCTION We assessed whether co-morbid small vessel disease (SVD) has clinical predictive value in preclinical or prodromal Alzheimer's disease. METHODS In 1090 non-demented participants (65.4 ± 10.7 years) SVD was assessed with magnetic resonance imaging and amyloid beta (Aβ) with lumbar puncture and/or positron emission tomography scan (mean follow-up for cognitive function 3.1 ± 2.4 years). RESULTS Thirty-nine percent had neither Aβ nor SVD (A-V-), 21% had SVD only (A-V+), 23% Aβ only (A+V-), and 17% had both (A+V+). Pooled cohort linear mixed model analyses demonstrated that compared to A-V- (reference), A+V- had a faster rate of cognitive decline. Co-morbid SVD (A+V+) did not further increase rate of decline. Cox regression showed that dementia risk was modestly increased in A-V+ (hazard ratio [95% confidence interval: 1.8 [1.0-3.2]) and most strongly in A+ groups. Also, mortality risk was increased in A+ groups. DISCUSSION In non-demented persons Aβ was predictive of cognitive decline, dementia, and mortality. SVD modestly predicts dementia in A-, but did not increase deleterious effects in A+. HIGHLIGHTS Amyloid beta (Aβ; A) was predictive for cognitive decline, dementia, and mortality. Small vessel disease (SVD) had no additional deleterious effects in A+. SVD modestly predicted dementia in A-. Aβ should be assessed even when magnetic resonance imaging indicates vascular cognitive impairment.
Collapse
Affiliation(s)
- Justine E. F. Moonen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Renée Haan
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Isabelle Bos
- Nivel, Research Institute for Better CareUtrechtthe Netherlands
| | - Charlotte Teunissen
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Neurochemistry LaboratoryDepartment of Clinical ChemistryAmsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit AmsterdamAmsterdamthe Netherlands
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
| | - Jori Tomassen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Sophie M. van der Landen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Eco J. C. de Geus
- Department of Biological PsychologyVU UniversityAmsterdamthe Netherlands
| | - Nienke Legdeur
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Argonde C. van Harten
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Calvin Trieu
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Casper de Boer
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Lior Kroeze
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear MedicineVrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Institute of Healthcare Engineering and the Institute of Neurology, University College LondonLondonUK
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNS), Maastricht UniversityMaastrichtthe Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of NeurogeriatricsKarolinska InstitutetSolnaSweden
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmcAmsterdamthe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamthe Netherlands
| |
Collapse
|
10
|
Zukowski LA, Fino PC, Levin I, Hsieh KL, Lockhart SN, Miller ME, Laurienti PJ, Kritchevsky SB, Hugenschmidt CE. Age and beta amyloid deposition impact gait speed, stride length, and gait smoothness while transitioning from an even to an uneven walking surface in older adults. Hum Mov Sci 2024; 93:103175. [PMID: 38198920 PMCID: PMC11195422 DOI: 10.1016/j.humov.2023.103175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/13/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Capturing a measure of movement quality during a complex walking task may indicate the earliest signs of detrimental changes to the brain due to beta amyloid (Aβ) deposition and be a potential differentiator of older adults at elevated and low risk of developing Alzheimer's disease. This study aimed to determine: 1) age-related differences in gait speed, stride length, and gait smoothness while transitioning from an even to an uneven walking surface, by comparing young adults (YA) and older adults (OA), and 2) if gait speed, stride length, and gait smoothness in OA while transitioning from an even to an uneven walking surface is influenced by the amount of Aβ deposition present in an OA's brain. METHODS Participants included 56 OA (>70 years of age) and 29 YA (25-35 years of age). In OA, Aβ deposition in the brain was quantified by PET imaging. All participants completed a series of cognitive assessments, a functional mobility assessment, and self-report questionnaires. Then participants performed two sets of walking trials on a custom-built walkway containing a mixture of even and uneven surface sections, including three trials with a grass uneven surface and three trials with a rocks uneven surface. Gait data were recorded using a wireless inertial measurement unit system. Stride length, gait speed, and gait smoothness (i.e., log dimensionless lumbar jerk) in the anteroposterior (AP), mediolateral (ML), and vertical (VT) directions were calculated for each stride. Outcomes were retained for five stride locations immediately surrounding the surface transition. RESULTS OA exhibited slower gait (Grass: p < 0.001; Rocks: p = 0.006), shorter strides (Grass: p < 0.001; Rocks: p = 0.008), and smoother gait (Grass AP: p < 0.001; Rocks AP: p = 0.002; Rocks ML: p = 0.02) than YA, but they also exhibited greater reductions in gait speed and stride length than YA while transitioning to the uneven grass and rocks surfaces. Within the OA group, those with greater Aβ deposition exhibited decreases in smoothness with age (Grass AP: p = 0.02; Rocks AP: p = 0.03; Grass ML: p = 0.04; Rocks ML: p = 0.03), while those with lower Aβ deposition exhibited increasing smoothness with age (Grass AP: p = 0.01; Rocks AP: p = 0.02; Grass ML: p = 0.08; Rocks ML: p = 0.07). Better functional mobility was associated with less smooth gait (Grass ML: p = 0.02; Rocks ML: p = 0.05) and with less variable gait smoothness (Grass and Rocks AP: both p = 0.04) in the OA group. CONCLUSION These results suggest that, relative to YA, OA may be adopting more cautious, compensatory gait strategies to maintain smoothness when approaching surface transitions. However, OA with greater Aβ deposition may have limited ability to adopt compensatory gait strategies to increase the smoothness of their walking as they get older because of neuropathological changes altering the sensory integration process and causing worse dynamic balance (i.e., jerkier gait). Functional mobility, in addition to age and Aβ deposition, may be an important factor of whether or not an OA chooses to employ compensatory strategies to prioritize smoothness while walking and what type of compensatory strategy an OA chooses.
Collapse
Affiliation(s)
- Lisa A Zukowski
- Department of Physical Therapy, High Point University, High Point, NC, United States of America.
| | - Peter C Fino
- Department of Health and Kinesiology, University of Utah, Salt Lake City, UT, United States of America
| | - Ilana Levin
- Department of Physical Therapy, High Point University, High Point, NC, United States of America
| | - Katherine L Hsieh
- Department of Physical Therapy, Georgia State University, Atlanta, GA, United States of America
| | - Samuel N Lockhart
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Michael E Miller
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Paul J Laurienti
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Christina E Hugenschmidt
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
11
|
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
|
12
|
Gunter NB, Gebre RK, Graff-Radford J, Heckman MG, Jack CR, Lowe VJ, Knopman DS, Petersen RC, Ross OA, Vemuri P, Ramanan VK. Machine Learning Models of Polygenic Risk for Enhanced Prediction of Alzheimer Disease Endophenotypes. Neurol Genet 2024; 10:e200120. [PMID: 38250184 PMCID: PMC10798228 DOI: 10.1212/nxg.0000000000200120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/01/2023] [Indexed: 01/23/2024]
Abstract
Background and Objectives Alzheimer disease (AD) has a polygenic architecture, for which genome-wide association studies (GWAS) have helped elucidate sequence variants (SVs) influencing susceptibility. Polygenic risk score (PRS) approaches show promise for generating summary measures of inherited risk for clinical AD based on the effects of APOE and other GWAS hits. However, existing PRS approaches, based on traditional regression models, explain only modest variation in AD dementia risk and AD-related endophenotypes. We hypothesized that machine learning (ML) models of polygenic risk (ML-PRS) could outperform standard regression-based PRS methods and therefore have the potential for greater clinical utility. Methods We analyzed combined data from the Mayo Clinic Study of Aging (n = 1,791) and the Alzheimer's Disease Neuroimaging Initiative (n = 864). An AD PRS was computed for each participant using the top common SVs obtained from a large AD dementia GWAS. In parallel, ML models were trained using those SV genotypes, with amyloid PET burden as the primary outcome. Secondary outcomes included amyloid PET positivity and clinical diagnosis (cognitively unimpaired vs impaired). We compared performance between ML-PRS and standard PRS across 100 training sessions with different data splits. In each session, data were split into 80% training and 20% testing, and then five-fold cross-validation was used within the training set to ensure the best model was produced for testing. We also applied permutation importance techniques to assess which genetic factors contributed most to outcome prediction. Results ML-PRS models outperformed the AD PRS (r2 = 0.28 vs r2 = 0.24 in test set) in explaining variation in amyloid PET burden. Among ML approaches, methods accounting for nonlinear genetic influences were superior to linear methods. ML-PRS models were also more accurate when predicting amyloid PET positivity (area under the curve [AUC] = 0.80 vs AUC = 0.63) and the presence of cognitive impairment (AUC = 0.75 vs AUC = 0.54) compared with the standard PRS. Discussion We found that ML-PRS approaches improved upon standard PRS for prediction of AD endophenotypes, partly related to improved accounting for nonlinear effects of genetic susceptibility alleles. Further adaptations of the ML-PRS framework could help to close the gap of remaining unexplained heritability for AD and therefore facilitate more accurate presymptomatic and early-stage risk stratification for clinical decision-making.
Collapse
Affiliation(s)
- Nathaniel B Gunter
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Robel K Gebre
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Jonathan Graff-Radford
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Michael G Heckman
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Clifford R Jack
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Val J Lowe
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - David S Knopman
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Ronald C Petersen
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Owen A Ross
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Prashanthi Vemuri
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Vijay K Ramanan
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| |
Collapse
|
13
|
Duran T, Gaussoin SA, Latham LA, Rundle MM, Espeland MA, Williams BJ, Hughes TM, Craft S, Sachs BC, Bateman JR, Lockhart SN. Examining a Preclinical Alzheimer's Cognitive Composite for Telehealth Administration for Reliability Between In-Person and Remote Cognitive Testing with Neuroimaging Biomarkers. J Alzheimers Dis 2024; 99:679-691. [PMID: 38669545 PMCID: PMC11295943 DOI: 10.3233/jad-231435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Background The preclinical Alzheimer's cognitive composite (PACC) was developed for in-person administration to capture subtle cognitive decline. At the outset of the COVID-19 pandemic, cognitive testing was increasingly performed remotely by telephone or video administration. It is desirable to have a harmonized composite measurement derived from both in-person and remote assessments for identifying cognitive changes and to examine its relationship with common neuroimaging biomarkers. Objective We defined a telehealth compatible PACC (tPACC) and examined its relationship with neuroimaging biomarkers related to neurodegeneration, brain function and perfusion, white matter integrity, and amyloid-β. Methods We examined 648 participants' neuroimaging and in-person and remote cognitive testing data from the Wake Forest Alzheimer's Disease Research Center's Clinical Core cohort (observational study) to calculate a modified PACC (PACC5-RAVLT) score and tPACC scores (in-person and remote). We performed Spearman/intraclass correlation coefficient (ICC) analyses for reliability of tPACC scores and linear regression models to evaluate associations between tPACC and neuroimaging. Bland-Altman plots for agreement were constructed across cognitively normal and impaired (mild cognitive impairment and dementia) participants. Results There was a significant positive relationship between tPACCin - person and PACC5-RAVLT (Overall group: r2 = 0.94, N = 648), and tPACCin - person and tPACCremote (validation subgroup: ICC = 0.82, n = 53). Overall, tPACC showed significant associations with brain thickness/volume, gray matter perfusion, white matter free water, and amyloid-β deposition. Conclusions There is a good agreement between tPACCand PACC5-RAVLTfor cognitively normal and impaired individuals. The tPACC is associated with common neuroimaging markers of Alzheimer's disease.
Collapse
Affiliation(s)
- Tugce Duran
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Sarah A. Gaussoin
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Lauren A. Latham
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Melissa M. Rundle
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Mark A. Espeland
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Benjamin J. Williams
- Department of Neurology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Timothy M. Hughes
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Suzanne Craft
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Bonnie C. Sachs
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - James R. Bateman
- Department of Neurology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Samuel N. Lockhart
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| |
Collapse
|
14
|
Zukowski LA, Brinkerhoff SA, Levin I, Herter TM, Hetrick L, Lockhart SN, Miller ME, Laurienti PJ, Kritchevsky SB, Hugenschmidt CE. Amyloid-β Deposition Predicts Grocery Shopping Performance in Older Adults Without Cognitive Impairment. J Alzheimers Dis 2024; 100:53-75. [PMID: 38820016 DOI: 10.3233/jad-231108] [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: 06/02/2024]
Abstract
Background A screening tool sensitive to Alzheimer's disease (AD) risk factors, such as amyloid-β (Aβ) deposition, and subtle cognitive changes, best elicited by complex everyday tasks, is needed. Objective To determine if grocery shopping performance could differentiate older adults at elevated risk of developing AD (OAer), older adults at low risk of developing AD (OAlr), and young adults (YA), and if amount of Aβ deposition could predict grocery shopping performance in older adults (OA). Methods Twenty-one OAer (78±5 years), 33 OAlr (78±5 years), and 28 YA (31±3 years) performed four grocery shopping trials, with the best and worst performances analyzed. Measures included trial time, number of correct items, number of grocery note fixations, and number of fixations and percentage of time fixating on the correct shelving unit, correct brand, and correct shelf. Linear mixed effects models compared measures by performance rank (best, worst) and group (OAer, OAlr, YA), and estimated the effect of Aβ deposition on measures in OA. Results Relative to their best performance, OAer and OAlr exhibited more correct shelving unit fixations and correct brand fixations during their worst performance, while YA did not. Within OA's worst performance, greater Aβ deposition was associated with a smaller percentage of time fixating on the correct shelving unit, correct shelf, and correct brand. Within OA, greater Aβ deposition was associated with more grocery note fixations. Conclusions OA with elevated Aβ deposition may exhibit subtle working memory impairments and less efficient visual search strategies while performing a cognitively demanding everyday task.
Collapse
Affiliation(s)
- Lisa A Zukowski
- Department of Physical Therapy, High Point University, High Point, NC, USA
| | - Sarah A Brinkerhoff
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ilana Levin
- Department of Physical Therapy, High Point University, High Point, NC, USA
| | - Troy M Herter
- Department of Exercise Science, University of South Carolina, Columbia, SC, USA
| | - Lena Hetrick
- Department of Neuroscience, High Point University, High Point, NC, USA
| | - Samuel N Lockhart
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Michael E Miller
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Paul J Laurienti
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Christina E Hugenschmidt
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
15
|
Grayson JM, Short SM, Lee CJ, Park N, Marsac C, Sette A, Lindestam Arlehamn CS, Leng XI, Lockhart SN, Craft S. T cell exhaustion is associated with cognitive status and amyloid accumulation in Alzheimer's disease. Sci Rep 2023; 13:15779. [PMID: 37737298 PMCID: PMC10516910 DOI: 10.1038/s41598-023-42708-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
Studies over the last 100 years have suggested a link between inflammation, infectious disease, and Alzheimer's Disease (AD). Understanding how the immune system changes during the development of AD may facilitate new treatments. Here, we studied an aging cohort who had been assessed for AD pathology with amyloid positron emission tomography and cognitive testing, and conducted high dimensional flow cytometry on peripheral blood mononuclear and cerebrospinal fluid cells. Participants were assigned a classification of being amyloid negative cognitively normal, amyloid positive cognitively normal (APCN), or amyloid positive mild cognitive impairment (APMCI), an early stage of AD. We observed major alterations in the peripheral innate immune system including increased myeloid and plasmacytoid dendritic cells in the blood of APMCI participants. When the adaptive immune system was examined, amyloid positive participants, regardless of cognitive status, had increased CD3+ T cells. Further analyses of CD4+ and CD8+ T cells revealed that APMCI participants had an increase in more differentiated phenotype T cells, such as effector memory and effector memory CD45RA expressing (TEMRA), compared to those with normal cognition. When T cell function was measured, we observed that T cells from APCN participants had increased IFNγ+GzB- producing cells compared to the other participants. In contrast, we demonstrate that APMCI participants had a major increase in T cells that lacked cytokine production following restimulation and expressed increased levels of PD-1 and Tox, suggesting these are exhausted cells. Rejuvenation of these cells may provide a potential treatment for AD.
Collapse
Affiliation(s)
- Jason M Grayson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA.
| | - Samantha M Short
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA
| | - C Jiah Lee
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA
| | - Nuri Park
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA
| | - Caitlyn Marsac
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA
| | - Alessandro Sette
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | | | - Xiaoyan I Leng
- Department of Biostatistics and Data Science, One Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Samuel N Lockhart
- Department of Internal Medicine-Geriatrics, One Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Suzanne Craft
- Department of Internal Medicine-Geriatrics, One Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| |
Collapse
|
16
|
Salvadó G, Horie K, Barthélemy NR, Vogel JW, Binette AP, Chen CD, Aschenbrenner AJ, Gordon BA, Benzinger TL, Holtzman DM, Morris JC, Palmqvist S, Stomrud E, Janelidze S, Ossenkoppele R, Schindler SE, Bateman RJ, Hansson O. Novel CSF tau biomarkers can be used for disease staging of sporadic Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.14.23292650. [PMID: 37503281 PMCID: PMC10370223 DOI: 10.1101/2023.07.14.23292650] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Biological staging of individuals with Alzheimer's disease (AD) may improve diagnostic and prognostic work-up of dementia in clinical practice and the design of clinical trials. Here, we created a staging model using the Subtype and Stage Inference (SuStaIn) algorithm by evaluating cerebrospinal fluid (CSF) amyloid-β (Aβ) and tau biomarkers in 426 participants from BioFINDER-2, that represent the entire spectrum of AD. The model composition and main analyses were replicated in 222 participants from the Knight ADRC cohort. SuStaIn revealed in the two cohorts that the data was best explained by a single biomarker sequence (one subtype), and that five CSF biomarkers (ordered: Aβ42/40, tau phosphorylation occupancies at the residues 217 and 205 [pT217/T217 and pT205/T205], microtubule-binding region of tau containing the residue 243 [MTBR-tau243], and total tau) were sufficient to create an accurate disease staging model. Increasing CSF stages (0-5) were associated with increased abnormality in other AD-related biomarkers, such as Aβ- and tau-PET, and aligned with different phases of longitudinal biomarker changes consistent with current models of AD progression. Higher CSF stages at baseline were associated with higher hazard ratio of clinical decline. Our findings indicate that a common pathophysiologic molecular pathway develops across all AD patients, and that a single CSF collection is sufficient to reliably indicate the presence of both AD pathologies and the degree and stage of disease progression.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Kanta Horie
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Eisai Inc., Nutley, NJ, United States
| | - Nicolas R. Barthélemy
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Jacob W. Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Science, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Charles D. Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A. Gordon
- 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
- Charles F. and Joanne 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, United States
- Charles F. and Joanne 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, United States
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J. Bateman
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne 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
| |
Collapse
|
17
|
Roh HW, Son SJ, Hong CH, Moon SY, Lee SM, Seo SW, Choi SH, Kim EJ, Cho SH, Kim BC, Park S, Song S, An YS. Comparison of automated quantification of amyloid deposition between PMOD and Heuron. Sci Rep 2023; 13:9891. [PMID: 37336977 DOI: 10.1038/s41598-023-36986-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
Several programs are widely used for clinical and research purposes to automatically quantify the degree of amyloid deposition in the brain using positron emission tomography (PET) images. Given that very few studies have investigated the use of Heuron, a PET image quantification software approved for clinical use, this study aimed to compare amyloid deposition values quantified from 18F-flutemetamol PET images using PMOD and Heuron. Amyloid PET data obtained from 408 patients were analysed using each quantitative program; moreover, the standardized uptake value ratios (SUVRs) of target areas were obtained by dividing the standardized uptake value (SUV) of the target region by the SUV of cerebellar grey matter as a reference. Compared with PMOD, Heuron yielded significantly higher SUVRs for all target areas (paired sample t-test, p < 0.001), except for the PC/PCC (p = 0.986). However, the Bland-Altman plot analysis indicated that the two quantitative methods may be used interchangeably. Moreover, receiver operating characteristic curve analysis revealed no significant between-method difference in the performance of the SUVRs in evaluating the visual positivity of amyloid deposits (p = 0.948). In conclusion, Heuron and PMOD have comparable performance in quantifying the degree of amyloid deposits in PET images.
Collapse
Affiliation(s)
- Hyun Woong Roh
- Department of Psychiatry, School of Medicine, Ajou University, Suwon, Korea
| | - Sang Joon Son
- Department of Psychiatry, School of Medicine, Ajou University, Suwon, Korea
| | - Chang Hyung Hong
- Department of Psychiatry, School of Medicine, Ajou University, Suwon, Korea
| | - So Young Moon
- Department of Neurology, School of Medicine, Ajou University, Suwon, Korea
| | - Sun Min Lee
- Department of Neurology, School of Medicine, Ajou University, Suwon, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, Korea
| | - Seong Hye Choi
- Department of Neurology, School of Medicine, Inha University, Incheon, Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Medical Research Institute, Pusan National University School of Medicine, Busan, Korea
| | - Soo Hyun Cho
- Department of Neurology, Chonnam National University Medical School, Chonnam National University Hospital, Gwangju, Korea
| | - Byeong Chae Kim
- Department of Neurology, Chonnam National University Medical School, Chonnam National University Hospital, Gwangju, Korea
| | | | | | - Young-Sil An
- Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Ajou University, 206, World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, Suwon, 16499, Korea.
| |
Collapse
|
18
|
Pemberton HG, Buckley C, Battle M, Bollack A, Patel V, Tomova P, Cooke D, Balhorn W, Hegedorn K, Lilja J, Brand C, Farrar G. Software compatibility analysis for quantitative measures of [ 18F]flutemetamol amyloid PET burden in mild cognitive impairment. EJNMMI Res 2023; 13:48. [PMID: 37225974 DOI: 10.1186/s13550-023-00994-3] [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: 09/26/2022] [Accepted: 05/05/2023] [Indexed: 05/26/2023] Open
Abstract
RATIONALE Amyloid-β (Aβ) pathology is one of the earliest detectable brain changes in Alzheimer's disease pathogenesis. In clinical practice, trained readers will visually categorise positron emission tomography (PET) scans as either Aβ positive or negative. However, adjunct quantitative analysis is becoming more widely available, where regulatory approved software can currently generate metrics such as standardised uptake value ratios (SUVr) and individual Z-scores. Therefore, it is of direct value to the imaging community to assess the compatibility of commercially available software packages. In this collaborative project, the compatibility of amyloid PET quantification was investigated across four regulatory approved software packages. In doing so, the intention is to increase visibility and understanding of clinically relevant quantitative methods. METHODS Composite SUVr using the pons as the reference region was generated from [18F]flutemetamol (GE Healthcare) PET in a retrospective cohort of 80 amnestic mild cognitive impairment (aMCI) patients (40 each male/female; mean age = 73 years, SD = 8.52). Based on previous autopsy validation work, an Aβ positivity threshold of ≥ 0.6 SUVrpons was applied. Quantitative results from MIM Software's MIMneuro, Syntermed's NeuroQ, Hermes Medical Solutions' BRASS and GE Healthcare's CortexID were analysed using intraclass correlation coefficient (ICC), percentage agreement around the Aβ positivity threshold and kappa scores. RESULTS Using an Aβ positivity threshold of ≥ 0.6 SUVrpons, 95% agreement was achieved across the four software packages. Two patients were narrowly classed as Aβ negative by one software package but positive by the others, and two patients vice versa. All kappa scores around the same Aβ positivity threshold, both combined (Fleiss') and individual software pairings (Cohen's), were ≥ 0.9 signifying "almost perfect" inter-rater reliability. Excellent reliability was found between composite SUVr measurements for all four software packages, with an average measure ICC of 0.97 and 95% confidence interval of 0.957-0.979. Correlation coefficient analysis between the two software packages reporting composite z-scores was strong (r2 = 0.98). CONCLUSION Using an optimised cortical mask, regulatory approved software packages provided highly correlated and reliable quantification of [18F]flutemetamol amyloid PET with a ≥ 0.6 SUVrpons positivity threshold. In particular, this work could be of interest to physicians performing routine clinical imaging rather than researchers performing more bespoke image analysis. Similar analysis is encouraged using other reference regions as well as the Centiloid scale, when it has been implemented by more software packages.
Collapse
Affiliation(s)
- Hugh G Pemberton
- GE Healthcare, Pollards Wood, Chalfont St Giles, Amersham, HP8 4SP, UK.
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK.
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| | | | - Mark Battle
- GE Healthcare, Pollards Wood, Chalfont St Giles, Amersham, HP8 4SP, UK
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ariane Bollack
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK
| | - Vrajesh Patel
- GE Healthcare, Pollards Wood, Chalfont St Giles, Amersham, HP8 4SP, UK
| | - Petya Tomova
- GE Healthcare, Pollards Wood, Chalfont St Giles, Amersham, HP8 4SP, UK
| | | | | | | | | | - Christine Brand
- GE Healthcare, Pollards Wood, Chalfont St Giles, Amersham, HP8 4SP, UK
| | - Gill Farrar
- GE Healthcare, Pollards Wood, Chalfont St Giles, Amersham, HP8 4SP, UK
| |
Collapse
|
19
|
Brugulat-Serrat A, Sánchez-Benavides G, Cacciaglia R, Salvadó G, Shekari M, Collij LE, Buckley C, van Berckel BNM, Perissinotti A, Niñerola-Baizán A, Milà-Alomà M, Vilor-Tejedor N, Operto G, Falcon C, Grau-Rivera O, Arenaza-Urquijo EM, Minguillón C, Fauria K, Molinuevo JL, Suárez-Calvet M, Gispert JD. APOE-ε4 modulates the association between regional amyloid deposition and cognitive performance in cognitively unimpaired middle-aged individuals. EJNMMI Res 2023; 13:18. [PMID: 36856866 PMCID: PMC9978048 DOI: 10.1186/s13550-023-00967-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/10/2023] [Indexed: 03/02/2023] Open
Abstract
PURPOSE To determine whether the APOE-ε4 allele modulates the relationship between regional β-amyloid (Aβ) accumulation and cognitive change in middle-aged cognitively unimpaired (CU) participants. METHODS The 352 CU participants (mean aged 61.1 [4.7] years) included completed two cognitive assessments (average interval 3.34 years), underwent [18F]flutemetamol Aβ positron emission tomography (PET), T1w magnetic resonance imaging (MRI), as well as APOE genotyping. Global and regional Aβ PET positivity was assessed across five regions-of-interest by visual reading (VR) and regional Centiloids. Linear regression models were developed to examine the interaction between regional and global Aβ PET positivity and APOE-ε4 status on longitudinal cognitive change assessed with the Preclinical Alzheimer's Cognitive Composite (PACC), episodic memory, and executive function, after controlling for age, sex, education, cognitive baseline scores, and hippocampal volume. RESULTS In total, 57 participants (16.2%) were VR+ of whom 41 (71.9%) were APOE-ε4 carriers. No significant APOE-ε4*global Aβ PET interactions were associated with cognitive change for any cognitive test. However, APOE-ε4 carriers who were VR+ in temporal areas (n = 19 [9.81%], p = 0.04) and in the striatum (n = 8 [4.14%], p = 0.01) exhibited a higher decline in the PACC. The temporal areas findings were replicated when regional PET positivity was determined with Centiloid values. Regionally, VR+ in the striatum was associated with higher memory decline. As for executive function, interactions between APOE-ε4 and regional VR+ were found in temporal and parietal regions, and in the striatum. CONCLUSION CU APOE-ε4 carriers with a positive Aβ PET VR in regions known to accumulate amyloid at later stages of the Alzheimer's disease (AD) continuum exhibited a steeper cognitive decline. This work supports the contention that regional VR of Aβ PET might convey prognostic information about future cognitive decline in individuals at higher risk of developing AD. CLINICALTRIALS gov Identifier: NCT02485730. Registered 20 June 2015 https://clinicaltrials.gov/ct2/show/NCT02485730 and ClinicalTrials.gov Identifier:NCT02685969. Registered 19 February 2016 https://clinicaltrials.gov/ct2/show/NCT02685969 .
Collapse
Affiliation(s)
- Anna Brugulat-Serrat
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain ,grid.512357.7Global Brain Health Institute, San Francisco, CA USA
| | - Gonzalo Sánchez-Benavides
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Raffaele Cacciaglia
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Salvadó
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.4514.40000 0001 0930 2361Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Mahnaz Shekari
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra, Barcelona, Spain
| | - Lyduine E. Collij
- grid.12380.380000 0004 1754 9227Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, Amsterdam, The Netherlands
| | - Christopher Buckley
- grid.83440.3b0000000121901201Center for Medical Image Computing, and Queen Square Institute of Neurology, UCL, London, UK
| | - Bart N. M. van Berckel
- grid.12380.380000 0004 1754 9227Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, Amsterdam, The Netherlands
| | - Andrés Perissinotti
- grid.410458.c0000 0000 9635 9413Nuclear Medicine Department, Hospital Clínic, Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Biomedical Research Networking Center of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Aida Niñerola-Baizán
- grid.410458.c0000 0000 9635 9413Nuclear Medicine Department, Hospital Clínic, Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Biomedical Research Networking Center of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Milà-Alomà
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra, Barcelona, Spain
| | - Natàlia Vilor-Tejedor
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra, Barcelona, Spain ,grid.473715.30000 0004 6475 7299Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Grégory Operto
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carles Falcon
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.411142.30000 0004 1767 8811Neurologia Department, Hospital del Mar, Barcelona, Spain
| | - Oriol Grau-Rivera
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain ,grid.411142.30000 0004 1767 8811Neurologia Department, Hospital del Mar, Barcelona, Spain
| | - Eider M. Arenaza-Urquijo
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Minguillón
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Karine Fauria
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - José Luis Molinuevo
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.424580.f0000 0004 0476 7612H. Lundbeck A/S, Copenhagen, Denmark
| | - Marc Suárez-Calvet
- grid.430077.7Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005 Barcelona, Spain ,grid.411142.30000 0004 1767 8811IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain ,grid.413448.e0000 0000 9314 1427Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain ,grid.411142.30000 0004 1767 8811Neurologia Department, Hospital del Mar, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Wellington 30, 08005, Barcelona, Spain. .,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,Biomedical Research Networking Center of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.
| | | |
Collapse
|
20
|
Collij LE, Farrar G, Valléz García D, Bader I, Shekari M, Lorenzini L, Pemberton H, Altomare D, Pla S, Loor M, Markiewicz P, Yaqub M, Buckley C, Frisoni GB, Nordberg A, Payoux P, Stephens A, Gismondi R, Visser PJ, Ford L, Schmidt M, Birck C, Georges J, Mett A, Walker Z, Boada M, Drzezga A, Vandenberghe R, Hanseeuw B, Jessen F, Schöll M, Ritchie C, Lopes Alves I, Gispert JD, Barkhof F. The amyloid imaging for the prevention of Alzheimer's disease consortium: A European collaboration with global impact. Front Neurol 2023; 13:1063598. [PMID: 36761917 PMCID: PMC9907029 DOI: 10.3389/fneur.2022.1063598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/08/2022] [Indexed: 01/22/2023] Open
Abstract
Background Amyloid-β (Aβ) accumulation is considered the earliest pathological change in Alzheimer's disease (AD). The Amyloid Imaging to Prevent Alzheimer's Disease (AMYPAD) consortium is a collaborative European framework across European Federation of Pharmaceutical Industries Associations (EFPIA), academic, and 'Small and Medium-sized enterprises' (SME) partners aiming to provide evidence on the clinical utility and cost-effectiveness of Positron Emission Tomography (PET) imaging in diagnostic work-up of AD and to support clinical trial design by developing optimal quantitative methodology in an early AD population. The AMYPAD studies In the Diagnostic and Patient Management Study (DPMS), 844 participants from eight centres across three clinical subgroups (245 subjective cognitive decline, 342 mild cognitive impairment, and 258 dementia) were included. The Prognostic and Natural History Study (PNHS) recruited pre-dementia subjects across 11 European parent cohorts (PCs). Approximately 1600 unique subjects with historical and prospective data were collected within this study. PET acquisition with [18F]flutemetamol or [18F]florbetaben radiotracers was performed and quantified using the Centiloid (CL) method. Results AMYPAD has significantly contributed to the AD field by furthering our understanding of amyloid deposition in the brain and the optimal methodology to measure this process. Main contributions so far include the validation of the dual-time window acquisition protocol to derive the fully quantitative non-displaceable binding potential (BP ND ), assess the value of this metric in the context of clinical trials, improve PET-sensitivity to emerging Aβ burden and utilize its available regional information, establish the quantitative accuracy of the Centiloid method across tracers and support implementation of quantitative amyloid-PET measures in the clinical routine. Future steps The AMYPAD consortium has succeeded in recruiting and following a large number of prospective subjects and setting up a collaborative framework to integrate data across European PCs. Efforts are currently ongoing in collaboration with ARIDHIA and ADDI to harmonize, integrate, and curate all available clinical data from the PNHS PCs, which will become openly accessible to the wider scientific community.
Collapse
Affiliation(s)
- Lyduine E. Collij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands,Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands,*Correspondence: Lyduine E. Collij ✉
| | | | - David Valléz García
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands,Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Ilona Bader
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands,Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | | | - Luigi Lorenzini
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands,Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Hugh Pemberton
- Centre for Medical Image Computing, and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), Université de Genève, Geneva, Switzerland
| | - Sandra Pla
- Synapse Research Management Partners, Barcelona, Spain
| | - Mery Loor
- Synapse Research Management Partners, Barcelona, Spain
| | - Pawel Markiewicz
- Centre for Medical Image Computing, and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | | | - Giovanni B. Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), Université de Genève, Geneva, Switzerland
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center of Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Pierre Payoux
- Department of Nuclear Medicine, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Andrew Stephens
- Life Molecular Imaging GmbH, Berlin, Baden-Württemberg, Germany
| | | | - Pieter Jelle Visser
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | - Lisa Ford
- Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | | | - Anja Mett
- GE Healthcare, Amersham, United Kingdom
| | - Zuzana Walker
- Centre for Medical Image Computing, and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Mercé Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Alexander Drzezga
- Department of Psychiatry, University Hospital of Cologne, Cologne, North Rhine-Westphalia, Germany
| | - Rik Vandenberghe
- Faculty of Medicine, University Hospitals Leuven, Leuven, Brussels, Belgium
| | - Bernard Hanseeuw
- Institute of Neuroscience (IONS), Université Catholique de Louvain, Brussels, Belgium
| | - Frank Jessen
- Department of Psychiatry, University Hospital of Cologne, Cologne, North Rhine-Westphalia, Germany
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Craig Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | | | - Juan Domingo Gispert
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands,Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands,Centre for Medical Image Computing, and Queen Square Institute of Neurology, UCL, London, United Kingdom
| |
Collapse
|
21
|
Kim J, Choe YS, Park Y, Kim Y, Kim JP, Jang H, Kim HJ, Na DL, Cho SJ, Moon SH, Seo SW. Clinical outcomes of increased focal amyloid uptake in individuals with subthreshold global amyloid levels. Front Aging Neurosci 2023; 15:1124445. [PMID: 36936497 PMCID: PMC10017468 DOI: 10.3389/fnagi.2023.1124445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Background Although the standardized uptake value ratio (SUVR) method is objective and simple, cut-off optimization using global SUVR values may not reflect focal increased uptake in the cerebrum. The present study investigated clinical and neuroimaging characteristics according to focally increased β-amyloid (Aβ) uptake and global Aβ status. Methods We recruited 968 participants with cognitive continuum. All participants underwent neuropsychological tests and 498 18F-florbetaben (FBB) amyloid positron emission tomography (PET) and 470 18F-flutemetamol (FMM) PET. Each PET scan was assessed in 10 regions (left and right frontal, lateral temporal, parietal, cingulate, and striatum) with focal-quantitative SUVR-based cutoff values for each region by using an iterative outlier approach. Results A total of 62 (6.4%) subjects showed increased focal Aβ uptake with subthreshold global Aβ status [global (-) and focal (+) Aβ group, G(-)F(+) group]. The G(-)F(+) group showed worse performance in memory impairment (p < 0.001), global cognition (p = 0.009), greater hippocampal atrophy (p = 0.045), compared to those in the G(-)F(-). Participants with widespread Aβ involvement in the whole region [G(+)] showed worse neuropsychological (p < 0.001) and neuroimaging features (p < 0.001) than those with focal Aβ involvement G(-)F(+). Conclusion Our findings suggest that individuals show distinctive clinical outcomes according to focally increased Aβ uptake and global Aβ status. Thus, researchers and clinicians should pay more attention to focal increased Aβ uptake in addition to global Aβ status.
Collapse
Affiliation(s)
- Jaeho Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Yeong Sim Choe
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Yuhyun Park
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Yeshin Kim
- Department of Neurology, Kangwon National University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
- Samsung Medical Center, Stem Cell and Regenerative Medicine Institute, Seoul, Republic of Korea
| | - Soo-Jin Cho
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- *Correspondence: Seung Hwan Moon,
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
- Sang Won Seo,
| |
Collapse
|
22
|
Collij LE, Salvadó G, de Wilde A, Altomare D, Shekari M, Gispert JD, Bullich S, Stephens A, Barkhof F, Scheltens P, Bouwman F, van der Flier WM. Quantification of [
18
F]florbetaben amyloid‐PET imaging in a mixed memory clinic population: The ABIDE project. Alzheimers Dement 2022. [DOI: 10.1002/alz.12886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Lyduine E. Collij
- Department of Radiology and Nuclear Medicine Amsterdam University Medical Center Amsterdam Neuroscience Amsterdam The Netherlands
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC) Pasqual Maragall Foundation Barcelona Spain
- Clinical Memory Research Unit Department of Clinical Sciences Lund University Malmö Sweden
| | - Arno de Wilde
- Department of Neurology Alzheimer Center Amsterdam Amsterdam Neuroscience Vrije Universiteit Amsterdam Amsterdam UMC Amsterdam The Netherlands
| | - Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE) University of Geneva Geneva Switzerland
- Memory Center Geneva University Hospitals Geneva Switzerland
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC) Pasqual Maragall Foundation Barcelona Spain
- IMIM (Hospital del Mar Medical Research Institute) Barcelona Spain
- Pompeu Fabra University Barcelona Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC) Pasqual Maragall Foundation Barcelona Spain
- IMIM (Hospital del Mar Medical Research Institute) Barcelona Spain
- Centro de Investigación Biomédica en Red de Bioingeniería Biomateriales y Nanomedicina (CIBER‐BBN) Madrid Spain
| | | | | | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine Amsterdam University Medical Center Amsterdam Neuroscience Amsterdam The Netherlands
- Centre for Medical Image Computing and Queen Square Institute of Neurology UCL London UK
| | - Philip Scheltens
- Department of Neurology Alzheimer Center Amsterdam Amsterdam Neuroscience Vrije Universiteit Amsterdam Amsterdam UMC Amsterdam The Netherlands
| | - Femke Bouwman
- Department of Neurology Alzheimer Center Amsterdam Amsterdam Neuroscience Vrije Universiteit Amsterdam Amsterdam UMC Amsterdam The Netherlands
| | - Wiesje M. van der Flier
- Department of Neurology Alzheimer Center Amsterdam Amsterdam Neuroscience Vrije Universiteit Amsterdam Amsterdam UMC Amsterdam The Netherlands
- Department of Epidemiology & Data Science Amsterdam Neuroscience Vrije Universiteit Amsterdam Amsterdam UMC Amsterdam The Netherlands
| |
Collapse
|
23
|
Pemberton HG, Collij LE, Heeman F, Bollack A, Shekari M, Salvadó G, Alves IL, Garcia DV, Battle M, Buckley C, Stephens AW, Bullich S, Garibotto V, Barkhof F, Gispert JD, Farrar G. Quantification of amyloid PET for future clinical use: a state-of-the-art review. Eur J Nucl Med Mol Imaging 2022; 49:3508-3528. [PMID: 35389071 PMCID: PMC9308604 DOI: 10.1007/s00259-022-05784-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Amyloid-β (Aβ) pathology is one of the earliest detectable brain changes in Alzheimer's disease (AD) pathogenesis. The overall load and spatial distribution of brain Aβ can be determined in vivo using positron emission tomography (PET), for which three fluorine-18 labelled radiotracers have been approved for clinical use. In clinical practice, trained readers will categorise scans as either Aβ positive or negative, based on visual inspection. Diagnostic decisions are often based on these reads and patient selection for clinical trials is increasingly guided by amyloid status. However, tracer deposition in the grey matter as a function of amyloid load is an inherently continuous process, which is not sufficiently appreciated through binary cut-offs alone. State-of-the-art methods for amyloid PET quantification can generate tracer-independent measures of Aβ burden. Recent research has shown the ability of these quantitative measures to highlight pathological changes at the earliest stages of the AD continuum and generate more sensitive thresholds, as well as improving diagnostic confidence around established binary cut-offs. With the recent FDA approval of aducanumab and more candidate drugs on the horizon, early identification of amyloid burden using quantitative measures is critical for enrolling appropriate subjects to help establish the optimal window for therapeutic intervention and secondary prevention. In addition, quantitative amyloid measurements are used for treatment response monitoring in clinical trials. In clinical settings, large multi-centre studies have shown that amyloid PET results change both diagnosis and patient management and that quantification can accurately predict rates of cognitive decline. Whether these changes in management reflect an improvement in clinical outcomes is yet to be determined and further validation work is required to establish the utility of quantification for supporting treatment endpoint decisions. In this state-of-the-art review, several tools and measures available for amyloid PET quantification are summarised and discussed. Use of these methods is growing both clinically and in the research domain. Concurrently, there is a duty of care to the wider dementia community to increase visibility and understanding of these methods.
Collapse
Affiliation(s)
- Hugh G Pemberton
- GE Healthcare, Amersham, UK.
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK.
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Lyduine E Collij
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Fiona Heeman
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ariane Bollack
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Isadora Lopes Alves
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Brain Research Center, Amsterdam, The Netherlands
| | - David Vallez Garcia
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mark Battle
- GE Healthcare, Amersham, UK
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | | | | | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
- NIMTLab, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Frederik Barkhof
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Bioengineering, University College London, London, UK
- UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Radiology and Nuclear Medicine, Amsterdam Neurocience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | | |
Collapse
|
24
|
Lorenzini L, Ansems LT, Lopes Alves I, Ingala S, Vállez García D, Tomassen J, Sudre C, Salvadó G, Shekari M, Operto G, Brugulat-Serrat A, Sánchez-Benavides G, ten Kate M, Tijms B, Wink AM, Mutsaerts HJMM, den Braber A, Visser PJ, van Berckel BNM, Gispert JD, Barkhof F, Collij LE. Regional associations of white matter hyperintensities and early cortical amyloid pathology. Brain Commun 2022; 4:fcac150. [PMID: 35783557 PMCID: PMC9246276 DOI: 10.1093/braincomms/fcac150] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/11/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
White matter hyperintensities (WMHs) have a heterogeneous aetiology, associated with both vascular risk factors and amyloidosis due to Alzheimer's disease. While spatial distribution of both amyloid and WM lesions carry important information for the underlying pathogenic mechanisms, the regional relationship between these two pathologies and their joint contribution to early cognitive deterioration remains largely unexplored. We included 662 non-demented participants from three Amyloid Imaging to Prevent Alzheimer's disease (AMYPAD)-affiliated cohorts: EPAD-LCS (N = 176), ALFA+ (N = 310), and EMIF-AD PreclinAD Twin60++ (N = 176). Using PET imaging, cortical amyloid burden was assessed regionally within early accumulating regions (medial orbitofrontal, precuneus, and cuneus) and globally, using the Centiloid method. Regional WMH volume was computed using Bayesian Model Selection. Global associations between WMH, amyloid, and cardiovascular risk scores (Framingham and CAIDE) were assessed using linear models. Partial least square (PLS) regression was used to identify regional associations. Models were adjusted for age, sex, and APOE-e4 status. Individual PLS scores were then related to cognitive performance in 4 domains (attention, memory, executive functioning, and language). While no significant global association was found, the PLS model yielded two components of interest. In the first PLS component, a fronto-parietal WMH pattern was associated with medial orbitofrontal-precuneal amyloid, vascular risk, and age. Component 2 showed a posterior WMH pattern associated with precuneus-cuneus amyloid, less related to age or vascular risk. Component 1 was associated with lower performance in all cognitive domains, while component 2 only with worse memory. In a large pre-dementia population, we observed two distinct patterns of regional associations between WMH and amyloid burden, and demonstrated their joint influence on cognitive processes. These two components could reflect the existence of vascular-dependent and -independent manifestations of WMH-amyloid regional association that might be related to distinct primary pathophysiology.
Collapse
Affiliation(s)
- Luigi Lorenzini
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Loes T Ansems
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Isadora Lopes Alves
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Silvia Ingala
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - David Vállez García
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Jori Tomassen
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Carole Sudre
- Centre for Medical Image Computing (CMIC), Departments of Medical Physics & Biomedical Engineering and Computer Science, University College London, UK
- MRC Unit for Lifelong Health and Ageing - University CollegeLondon, UK
- School of Biomedical Engineering, King’s College LondonUK
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Gregory Operto
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Anna Brugulat-Serrat
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Madrid, Spain
- Atlantic Fellow for Equity in Brain Health at the University of California San Francisco, SanFrancisco, California, USA
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Mara ten Kate
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Betty Tijms
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alle Meije Wink
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Henk J M M Mutsaerts
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Anouk den Braber
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department. of Biological Psychology, Vrije Universiteit Amsterdam, Neuroscience Amsterdam, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Bart N M van Berckel
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales Y Nanomedicina, Madrid, Spain
| | - Frederik Barkhof
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Lyduine E Collij
- Dept. of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Collij LE, Salvadó G, Wottschel V, Mastenbroek SE, Schoenmakers P, Heeman F, Aksman L, Wink AM, Berckel BNM, van de Flier WM, Scheltens P, Visser PJ, Barkhof F, Haller S, Gispert JD, Lopes Alves I. Spatial-Temporal Patterns of β-Amyloid Accumulation: A Subtype and Stage Inference Model Analysis. Neurology 2022; 98:e1692-e1703. [PMID: 35292558 PMCID: PMC9071373 DOI: 10.1212/wnl.0000000000200148] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND OBJECTIVES β-amyloid (Aβ) staging models assume a single spatial-temporal progression of amyloid accumulation. We assessed evidence for Aβ accumulation subtypes by applying the data-driven Subtype and Stage Inference (SuStaIn) model to amyloid-PET data. METHODS Amyloid-PET data of 3,010 participants were pooled from 6 cohorts (ALFA+, EMIF-AD, ABIDE, OASIS, and ADNI). Standardized uptake value ratios were calculated for 17 regions. We applied the SuStaIn algorithm to identify consistent subtypes in the pooled dataset based on the cross-validation information criterion and the most probable subtype/stage classification per scan. The effects of demographics and risk factors on subtype assignment were assessed using multinomial logistic regression. RESULTS Participants were mostly cognitively unimpaired (n = 1890 [62.8%]), had a mean age of 68.72 (SD 9.1) years, 42.1% were APOE ε4 carriers, and 51.8% were female. A 1-subtype model recovered the traditional amyloid accumulation trajectory, but SuStaIn identified 3 optimal subtypes, referred to as frontal, parietal, and occipital based on the first regions to show abnormality. Of the 788 (26.2%) with strong subtype assignment (>50% probability), the majority was assigned to frontal (n = 415 [52.5%]), followed by parietal (n = 199 [25.3%]) and occipital subtypes (n = 175 [22.2%]). Significant differences across subtypes included distinct proportions of APOE ε4 carriers (frontal 61.8%, parietal 57.1%, occipital 49.4%), participants with dementia (frontal 19.7%, parietal 19.1%, occipital 31.0%), and lower age for the parietal subtype (frontal/occipital 72.1 years, parietal 69.3 years). Higher amyloid (Centiloid) and CSF p-tau burden was observed for the frontal subtype; parietal and occipital subtypes did not differ. At follow-up, most participants (81.1%) maintained baseline subtype assignment and 25.6% progressed to a later stage. DISCUSSION Whereas a 1-trajectory model recovers the established pattern of amyloid accumulation, SuStaIn determined that 3 subtypes were optimal, showing distinct associations with Alzheimer disease risk factors. Further analyses to determine clinical utility are warranted.
Collapse
Affiliation(s)
- Lyduine E Collij
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Gemma Salvadó
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Viktor Wottschel
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Sophie E Mastenbroek
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Pierre Schoenmakers
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Fiona Heeman
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Leon Aksman
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Alle Meije Wink
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Bart N M Berckel
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Wiesje M van de Flier
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Philip Scheltens
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Pieter Jelle Visser
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Frederik Barkhof
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Sven Haller
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Juan Domingo Gispert
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| | - Isadora Lopes Alves
- From the Department of Radiology and Nuclear Medicine (L.E.C., V.W., S.E.M., P.S., F.H., A.M.W., B.N.M.B., F.B., I.L.A.), Alzheimer Center and Department of Neurology (W.M.v.d.F., P.S., P.J.V.), and Department of Epidemiology & Data Science (W.M.v.d.F.), Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; Barcelonaβeta Brain Research Center (BBRC) (G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (G.S., J.D.G.), Barcelona, Spain; Stevens Neuroimaging and Informatics Institute (L.A.), Keck School of Medicine, University of Southern California, Los Angeles; Centre for Medical Image Computing and Queen Square Institute of Neurology (F.B.), UCL, UK; Faculty of Medicine of the University of Geneva (S.H.); CIMC-Centre d'Imagerie Médicale de Cornavin (S.H.), Genève, Switzerland; Department of Surgical Sciences, Radiology (S.H.), Uppsala University, Sweden; Department of Radiology (S.H.), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; and Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) (J.D.G.), Madrid, Spain
| |
Collapse
|
26
|
Lee J, Ha S, Kim REY, Lee M, Kim D, Lim HK. Development of Amyloid PET Analysis Pipeline Using Deep Learning-Based Brain MRI Segmentation—A Comparative Validation Study. Diagnostics (Basel) 2022; 12:diagnostics12030623. [PMID: 35328176 PMCID: PMC8947654 DOI: 10.3390/diagnostics12030623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 02/04/2023] Open
Abstract
Amyloid positron emission tomography (PET) scan is clinically essential for the non-invasive assessment of the presence and spatial distribution of amyloid-beta deposition in subjects with cognitive impairment suspected to have been a result of Alzheimer’s disease. Quantitative assessment can enhance the interpretation reliability of PET scan; however, its clinical application has been limited due to the complexity of preprocessing. This study introduces a novel deep-learning-based approach for SUVR quantification that simplifies the preprocessing step and significantly reduces the analysis time. Using two heterogeneous amyloid ligands, our proposed method successfully distinguished standardized uptake value ratio (SUVR) between amyloidosis-positive and negative groups. The proposed method’s intra-class correlation coefficients were 0.97 and 0.99 against PETSurfer and PMOD, respectively. The difference of global SUVRs between the proposed method and PETSurfer or PMOD were 0.04 and −0.02, which are clinically acceptable. The AUC-ROC exceeded 0.95 for three tools in the amyloid positive assessment. Moreover, the proposed method had the fastest processing time and had a low registration failure rate (1%). In conclusion, our proposed method calculates SUVR that is consistent with PETSurfer and PMOD, and has advantages of fast processing time and low registration failure rate. Therefore, PET quantification provided by our proposed method can be used in clinical practice.
Collapse
Affiliation(s)
- Jiyeon Lee
- Research Institute, Neurophet Inc., Seoul 06234, Korea; (J.L.); (R.E.Y.K.); (M.L.)
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Regina E. Y. Kim
- Research Institute, Neurophet Inc., Seoul 06234, Korea; (J.L.); (R.E.Y.K.); (M.L.)
| | - Minho Lee
- Research Institute, Neurophet Inc., Seoul 06234, Korea; (J.L.); (R.E.Y.K.); (M.L.)
| | - Donghyeon Kim
- Research Institute, Neurophet Inc., Seoul 06234, Korea; (J.L.); (R.E.Y.K.); (M.L.)
- Correspondence: (D.K.); (H.K.L.); Tel.: +82-10-9361-3781 (D.K.); +82-10-3797-6315 (H.K.L.)
| | - Hyun Kook Lim
- Department of Psychiatry, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea
- Correspondence: (D.K.); (H.K.L.); Tel.: +82-10-9361-3781 (D.K.); +82-10-3797-6315 (H.K.L.)
| |
Collapse
|
27
|
PET imaging in dementia. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00089-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
28
|
Müller EG, Stokke C, Stokmo HL, Edwin TH, Knapskog AB, Revheim ME. Evaluation of semi-quantitative measures of 18F-flutemetamol PET for the clinical diagnosis of Alzheimer's disease. Quant Imaging Med Surg 2022; 12:493-509. [PMID: 34993096 DOI: 10.21037/qims-21-188] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/06/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND 18F-flutemetamol positron emission tomography (PET) is used to assess cortical amyloid-β burden in patients with cognitive impairment to support a clinical diagnosis. Visual classification is the most widely used method in clinical practice although semi-quantification is beneficial to obtain an objective and continuous measure of the Aβ burden. The aims were: first to evaluate the correspondence between standardized uptake value ratios (SUVRs) from three different software, Centiloids and visual classification, second to estimate thresholds for supporting visual classification and last to assess differences in semi-quantitative measures between clinical diagnoses. METHODS This observational study included 195 patients with cognitive impairment who underwent 18F-flutemetamol PET. PET images were semi-quantified with SyngoVia, CortexID suite, and PMOD. Receiver operating characteristics curves were used to compare visual classification with composite SUVR normalized to pons (SUVRpons) and cerebellar cortex (SUVRcer), and Centiloids. We explored correlations and differences between semi-quantitative measures as well as differences in SUVR between two clinical diagnosis groups: Alzheimer's disease-group and non-Alzheimer's disease-group. RESULTS PET images from 191 patients were semi-quantified with SyngoVia and CortexID and 86 PET-magnetic resonance imaging pairs with PMOD. All receiver operating characteristics curves showed a high area under the curve (>0.98). Thresholds for a visually positive PET was for SUVRcer: 1.87 (SyngoVia) and 1.64 (CortexID) and for SUVRpons: 0.54 (SyngoVia) and 0.55 (CortexID). The threshold on the Centiloid scale was 39.6 Centiloids. All semi-quantitative measures showed a very high correlation between different software and normalization methods. Composite SUVRcer was significantly different between SyngoVia and PMOD, SyngoVia and CortexID but not between PMOD and CortexID. Composite SUVRpons were significantly different between all three software. There were significant differences in the mean rank of SUVRpons, SUVRcer, and Centiloid between Alzheimer's disease-group and non-Alzheimer's disease-group. CONCLUSIONS SUVR from different software performed equally well in discriminating visually positive and negative 18F-Flutemetamol PET images. Thresholds should be considered software-specific and cautiously be applied across software without preceding validation to categorize scans as positive or negative. SUVR and Centiloid may be used alongside a thorough clinical evaluation to support a clinical diagnosis.
Collapse
Affiliation(s)
- Ebba Gløersen Müller
- Division of Radiology and Nuclear Medicine, Department of Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Caroline Stokke
- Division of Radiology and Nuclear Medicine, Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| | - Henning Langen Stokmo
- Division of Radiology and Nuclear Medicine, Department of Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trine Holt Edwin
- Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Geriatric Medicine, The memory clinic, Oslo University Hospital, Oslo, Norway
| | - Anne-Brita Knapskog
- Department of Geriatric Medicine, The memory clinic, Oslo University Hospital, Oslo, Norway
| | - Mona-Elisabeth Revheim
- Division of Radiology and Nuclear Medicine, Department of Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
29
|
Alongi P, Chiaravalloti A, Berti V, Vellani C, Trifirò G, Puccini G, Carli G, Chincarini A, Morbelli S, Perani D, Sestini S. Amyloid PET in the diagnostic workup of neurodegenerative disease. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00428-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|