1
|
Boccalini C, Peretti DE, Mathoux G, Iaccarino L, Ribaldi F, Scheffler M, Perani D, Frisoni GB, Garibotto V. Early-phase 18F-Flortaucipir tau-PET as a proxy of brain metabolism in Alzheimer's disease: a comparison with 18F-FDG-PET and early-phase amyloid-PET. Eur J Nucl Med Mol Imaging 2025; 52:1958-1969. [PMID: 39849149 PMCID: PMC12014785 DOI: 10.1007/s00259-024-07063-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/29/2024] [Indexed: 01/25/2025]
Abstract
PURPOSE As dual-phase amyloid-PET can evaluate amyloid (A) and neurodegeneration (N) with a single tracer injection, dual-phase tau-PET might be able to provide both tau (T) and N. Our study aims to assess the association of early-phase tau-PET scans and 18F-fluorodeoxyglucose (FDG) PET and their comparability in discriminating Alzheimer's disease (AD) patients and differentiating neurodegenerative patterns. METHODS 58 subjects evaluated at the Geneva Memory Center underwent dual-phase 18F-Flortaucipir-PET with early-phase acquisition (eTAU) and 18F-FDG-PET within 1 year. A subsample of 36 participants also underwent dual-phase amyloid-PET (eAMY). Standardized uptake value ratios (SUVRs) were calculated to assess the correlation of eTAU and their respective 18F-FDG-PET and eAMY scans. Hypometabolism and hypoperfusion maps and their spatial overlap were also evaluated at the individual level visually and semiquantitatively. Receiver operating characteristic analyses were performed to compare the discriminative power of eTAU, FDG, and eAMY SUVR between A-/T- and A+/T + participants. RESULTS Strong positive correlations were found between eTAU and FDG SUVRs (r = 0.84, p < 0.001) and eTAU and eAMY SUVRs (r > 0.87, p < 0.001). Clusters of significant hypoperfusion with good correspondence to hypometabolism topographies were found at the individual level, independently of the underlying neurodegenerative patterns. Both eTAU and FDG SUVRs significantly distinguished A+/T + from A-/T- individuals (AUCeTAU=0.604, AUCFDG=0.748) with FDG performing better than eTAU (p = 0.04). eAMY and eTAU SUVR showed comparable discriminative power. CONCLUSION Early-phase 18F-Flortaucipir-PET can provide perfusion information closely related to brain regional glucose metabolism and perfusion measured by early-phase amyloid-PET, even if less accurate than FDG-PET as a biomarker for neurodegeneration.
Collapse
Affiliation(s)
- Cecilia Boccalini
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Debora Elisa Peretti
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gregory Mathoux
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland
| | | | | | - Max Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | - Daniela Perani
- Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| | | | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland.
- CIBM Center for Biomedical Imaging, Geneva, Switzerland.
| |
Collapse
|
2
|
Völter F, Eckenweber S, Scheifele M, Eckenweber F, Hirsch F, Franzmeier N, Kreuzer A, Griessl M, Steward A, Janowitz D, Palleis C, Bernhardt A, Vöglein J, Stockbauer A, Rauchmann BS, Schöberl F, Wlasich E, Buerger K, Wagemann O, Perneczky R, Weidinger E, Höglinger G, Levin J, Brendel M, Schönecker S. Correlation of early-phase β-amyloid positron-emission-tomography and neuropsychological testing in patients with Alzheimer's disease. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07175-5. [PMID: 40019578 DOI: 10.1007/s00259-025-07175-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE Clinical staging in individuals with Alzheimer's disease (AD) typically relies on neuropsychological testing. Recognizing the imperative for an objective measure of clinical AD staging, regional perfusion in early-phase β-amyloid-PET may aid as a cost-efficient index for the assessment of neurodegeneration severity in patients with Alzheimer's disease. METHODS Regional perfusion deficits in early-phase β-amyloid-PET as well as neuropsychological testing (max. 90 days delay) were evaluated in 82 patients with biologically defined AD according to the ATN classification. In reference to the Braak staging system patients were classified into the groups stage0, stageI-II+, stageI-IV+, stageI-VI+, and stageatypical+ according to regional perfusion deficits in regions of interest (ROIs) published by the Alzheimer's Disease Neuroimaging Initiative. Multiple regression analysis controlling for age, gender, and education was used to evaluate the association of regional z-scores on perfusion-phase PET with clinical scores for all patients and with annual decline of cognitive performance in 23 patients with follow-up data. RESULTS Patients classified as stage0 and stageI-II+ demonstrated significantly superior neuropsychological performance compared to those classified as stageI-IV+ and stageI-VI+. Lower cognitive performance was associated with decreased perfusion in early-phase β-amyloid-PET globally and regionally, with the most pronounced association identified in the left temporal lobe. Mean z-scores on early-phase PET in temporal and parietal regions offered a robust prediction of future annual decline in MMSE and sum scores of the CERAD-Plus (Consortium to Establish a Registry for Alzheimer's Disease) test battery. CONCLUSION Regional and global perfusion deficits in early-phase β-amyloid-PET can serve as an objective index of neurodegeneration severity and may act as prognostic markers of future cognitive decline in AD.
Collapse
Affiliation(s)
- Friederike Völter
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
- Department of Internal Medicine IV, University Hospital of Munich, LMU Munich, Munich, Germany.
| | - Sebastian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Fabian Hirsch
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Annika Kreuzer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Maria Griessl
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Carla Palleis
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Alexander Bernhardt
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jonathan Vöglein
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Anna Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Neuroradiology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Florian Schöberl
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Elisabeth Wlasich
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Olivia Wagemann
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Endy Weidinger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Günter Höglinger
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Johannes Levin
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Sonja Schönecker
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
3
|
Bittner T, Tonietto M, Klein G, Belusov A, Illiano V, Voyle N, Delmar P, Scelsi MA, Gobbi S, Silvestri E, Barakovic M, Napolitano A, Galli C, Abaei M, Blennow K, Barkhof F. Biomarker treatment effects in two phase 3 trials of gantenerumab. Alzheimers Dement 2025; 21:e14414. [PMID: 39887500 PMCID: PMC11848197 DOI: 10.1002/alz.14414] [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: 07/03/2024] [Revised: 10/21/2024] [Accepted: 10/27/2024] [Indexed: 02/01/2025]
Abstract
INTRODUCTION We report biomarker treatment effects in the GRADUATE I and II phase 3 studies of gantenerumab in early Alzheimer's disease (AD). METHODS Amyloid and tau positron emission tomography (PET), volumetric magnetic resonance imaging (vMRI), cerebrospinal fluid (CSF), and plasma biomarkers used to assess gantenerumab treatment related changes on neuropathology, neurodegeneration, and neuroinflammation over 116 weeks. RESULTS Gantenerumab reduced amyloid PET load, CSF biomarkers of amyloid beta (Aβ)40, total tau (t-tau), phosphorylated tau 181 (p-tau181), neurogranin, S100 calcium-binding protein B (S100B), neurofilament light (NfL), alpha-synuclein (α-syn), neuronal pentraxin-2 (NPTX2), and plasma biomarkers of t-tau, p-tau181, p-tau217, and glial fibrillary acidic protein (GFAP) while increasing plasma Aβ40, Aβ42. vMRI showed increased reduction in whole brain volume and increased ventricular expansion, while hippocampal volume was unaffected. Tau PET showed no treatment effect. DISCUSSION Robust treatment effects were observed for multiple biomarkers in GRADUATE I and II. Comparison across anti-amyloid antibodies indicates utility of p-tau and GFAP as biomarkers of amyloid plaque removal while NfL and tau PET seem unsuitable as consistent indicators of clinical efficacy. vMRI might be confounded by non-neurodegenerative brain volume changes. TRIAL REGISTRATION NUMBER (CLINICALTRIALS.GOV IDENTIFIER): NCT03444870 and NCT03443973. HIGHLIGHTS Gantenerumab significantly reduced brain amyloid load. Tau positron emission tomography showed no treatment effect in a small subset of participants. Volumetric magnetic resonance imaging showed increased whole brain volume reduction under treatment while hippocampal volume was unaffected. Robust treatment effects on cerebrospinal fluid and plasma biomarkers were found, despite lack of clinical efficacy.
Collapse
Affiliation(s)
- Tobias Bittner
- Genentech, Inc.South San FranciscoCaliforniaUSA
- F. Hoffmann‐La Roche LtdBaselSwitzerland
| | | | | | | | | | | | | | | | | | - Erica Silvestri
- F. Hoffmann‐La Roche LtdBaselSwitzerland
- A4P Consulting Ltd.SandwichUK
| | | | | | | | - Maryam Abaei
- F. Hoffmann‐La Roche LtdBaselSwitzerland
- A4P Consulting Ltd.SandwichUK
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Paris Brain InstituteICMPitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
| | - Frederik Barkhof
- Department of Radiology & Nuclear MedicineAmsterdam UMCVrije UniversiteitAmsterdamthe Netherlands
- UCL Queen Square Institute of Neurology and Centre for Medical Image Computing, Queen SquareLondonUK
| |
Collapse
|
4
|
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 2025; 52:26-38. [PMID: 38433571 DOI: 10.1017/cjn.2024.31] [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: 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
|
5
|
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
|
6
|
Meindl M, Zatcepin A, Gnörich J, Scheifele M, Zaganjori M, Groß M, Lindner S, Schaefer R, Simmet M, Roemer S, Katzdobler S, Levin J, Höglinger G, Bischof AC, Barthel H, Sabri O, Bartenstein P, Saller T, Franzmeier N, Ziegler S, Brendel M. Assessment of [ 18F]PI-2620 Tau-PET Quantification via Non-Invasive Automatized Image Derived Input Function. Eur J Nucl Med Mol Imaging 2024; 51:3252-3266. [PMID: 38717592 PMCID: PMC11368995 DOI: 10.1007/s00259-024-06741-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 05/01/2024] [Indexed: 09/03/2024]
Abstract
PURPOSE [18F]PI-2620 positron emission tomography (PET) detects misfolded tau in progressive supranuclear palsy (PSP) and Alzheimer's disease (AD). We questioned the feasibility and value of absolute [18F]PI-2620 PET quantification for assessing tau by regional distribution volumes (VT). Here, arterial input functions (AIF) represent the gold standard, but cannot be applied in routine clinical practice, whereas image-derived input functions (IDIF) represent a non-invasive alternative. We aimed to validate IDIF against AIF and we evaluated the potential to discriminate patients with PSP and AD from healthy controls by non-invasive quantification of [18F] PET. METHODS In the first part of the study, we validated AIF derived from radial artery whole blood against IDIF by investigating 20 subjects (ten controls and ten patients). IDIF were generated by manual extraction of the carotid artery using the average and the five highest (max5) voxel intensity values and by automated extraction of the carotid artery using the average and the maximum voxel intensity value. In the second part of the study, IDIF quantification using the IDIF with the closest match to the AIF was transferred to group comparison of a large independent cohort of 40 subjects (15 healthy controls, 15 PSP patients and 10 AD patients). We compared VT and VT ratios, both calculated by Logan plots, with distribution volume (DV) ratios using simplified reference tissue modelling and standardized uptake value (SUV) ratios. RESULTS AIF and IDIF showed highly correlated input curves for all applied IDIF extraction methods (0.78 < r < 0.83, all p < 0.0001; area under the curves (AUC): 0.73 < r ≤ 0.82, all p ≤ 0.0003). Regarding the VT values, correlations were mainly found between those generated by the AIF and by the IDIF methods using the maximum voxel intensity values. Lowest relative differences (RD) were observed by applying the manual method using the five highest voxel intensity values (max5) (AIF vs. IDIF manual, avg: RD = -82%; AIF vs. IDIF automated, avg: RD = -86%; AIF vs. IDIF manual, max5: RD = -6%; AIF vs. IDIF automated, max: RD = -26%). Regional VT values revealed considerable variance at group level, which was strongly reduced upon scaling by the inferior cerebellum. The resulting VT ratio values were adequate to detect group differences between patients with PSP or AD and healthy controls (HC) (PSP target region (globus pallidus): HC vs. PSP vs. AD: 1.18 vs. 1.32 vs. 1.16; AD target region (Braak region I): HC vs. PSP vs. AD: 1.00 vs. 1.00 vs. 1.22). VT ratios and DV ratios outperformed SUV ratios and VT in detecting differences between PSP and healthy controls, whereas all quantification approaches performed similarly in comparing AD and healthy controls. CONCLUSION Blood-free IDIF is a promising approach for quantification of [18F]PI-2620 PET, serving as correlating surrogate for invasive continuous arterial blood sampling. Regional [18F]PI-2620 VT show large variance, in contrast to regional [18F]PI-2620 VT ratios scaled with the inferior cerebellum, which are appropriate for discriminating PSP, AD and healthy controls. DV ratios obtained by simplified reference tissue modeling are similarly suitable for this purpose.
Collapse
Affiliation(s)
- Maria Meindl
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Artem Zatcepin
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mattes Groß
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Rebecca Schaefer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Marcel Simmet
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Roemer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany
- Department of Neurology, Technical University Munich, Munich, Germany
| | - Ann-Cathrin Bischof
- Department of Anesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Saller
- Department of Anesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Sibylle Ziegler
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
7
|
Aye WWT, Stark MR, Horne K, Livingston L, Grenfell S, Myall DJ, Pitcher TL, Almuqbel MM, Keenan RJ, Meissner WG, Dalrymple‐Alford JC, Anderson TJ, Heron CL, Melzer TR. Early-phase amyloid PET reproduces metabolic signatures of cognitive decline in Parkinson's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12601. [PMID: 38912306 PMCID: PMC11193095 DOI: 10.1002/dad2.12601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION Recent work suggests that amyloid beta (Aβ) positron emission tomography (PET) tracer uptake shortly after injection ("early phase") reflects brain metabolism and perfusion. We assessed this modality in a predominantly amyloid-negative neurodegenerative condition, Parkinson's disease (PD), and hypothesized that early-phase 18F-florbetaben (eFBB) uptake would reproduce characteristic hypometabolism and hypoperfusion patterns associated with cognitive decline in PD. METHODS One hundred fifteen PD patients across the spectrum of cognitive impairment underwent dual-phase Aβ PET, structural and arterial spin labeling (ASL) magnetic resonance imaging (MRI), and neuropsychological assessments. Multiple linear regression models compared eFBB uptake to cognitive performance and ASL MRI perfusion. RESULTS Reduced eFBB uptake was associated with cognitive performance in brain regions previously linked to hypometabolism-associated cognitive decline in PD, independent of amyloid status. Furthermore, eFBB uptake correlated with cerebral perfusion across widespread regions. DISCUSSION EFBB uptake is a potential surrogate measure for cerebral perfusion/metabolism. A dual-phase PET imaging approach may serve as a clinical tool for assessing cognitive impairment. Highlights Images taken at amyloid beta (Aβ) positron emission tomography tracer injection may reflect brain perfusion and metabolism.Parkinson's disease (PD) is a predominantly amyloid-negative condition.Early-phase florbetaben (eFBB) in PD was associated with cognitive performance.eFBB uptake reflects hypometabolism-related cognitive decline in PD.eFBB correlated with arterial spin labeling magnetic resonance imaging measured cerebral perfusion.eFBB distinguished dementia from normal cognition and mild cognitive impairment.Findings were independent of late-phase Aβ burden.Thus, eFBB may serve as a surrogate measure for brain metabolism/perfusion.
Collapse
Affiliation(s)
- William W. T. Aye
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Department of MedicineUniversity of OtagoChristchurchNew Zealand
| | - Megan R. Stark
- New Zealand Brain Research InstituteChristchurchNew Zealand
| | - Kyla‐Louise Horne
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Department of MedicineUniversity of OtagoChristchurchNew Zealand
| | | | | | | | - Toni L. Pitcher
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Department of MedicineUniversity of OtagoChristchurchNew Zealand
| | - Mustafa M. Almuqbel
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Radiology Holding Company New ZealandChristchurchNew Zealand
| | - Ross J. Keenan
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Radiology Holding Company New ZealandChristchurchNew Zealand
| | - Wassilios G. Meissner
- New Zealand Brain Research InstituteChristchurchNew Zealand
- CHU Bordeaux, Service de Neurologie des Maladies NeurodégénérativesIMNc, NS‐Park/FCRIN NetworkBordeauxFrance
- Univ. Bordeaux, CNRS, IMNBordeauxFrance
| | - John C. Dalrymple‐Alford
- New Zealand Brain Research InstituteChristchurchNew Zealand
- School of Psychology, Speech and Hearing, University of Canterbury, PsychologySpeech and Hearing Arts Road, IlamChristchurchNew Zealand
| | - Tim J. Anderson
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Department of MedicineUniversity of OtagoChristchurchNew Zealand
- Department of NeurologyCanterbury District Health BoardChristchurchNew Zealand
| | - Campbell Le Heron
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Department of MedicineUniversity of OtagoChristchurchNew Zealand
- School of Psychology, Speech and Hearing, University of Canterbury, PsychologySpeech and Hearing Arts Road, IlamChristchurchNew Zealand
- Department of NeurologyCanterbury District Health BoardChristchurchNew Zealand
| | - Tracy R. Melzer
- New Zealand Brain Research InstituteChristchurchNew Zealand
- Department of MedicineUniversity of OtagoChristchurchNew Zealand
- Radiology Holding Company New ZealandChristchurchNew Zealand
- School of Psychology, Speech and Hearing, University of Canterbury, PsychologySpeech and Hearing Arts Road, IlamChristchurchNew Zealand
| |
Collapse
|
8
|
Lopergolo D, Bianchi S, Gallus GN, Locci S, Pucci B, Leoni V, Gasparini D, Tardelli E, Chincarini A, Sestini S, Santorelli FM, Zetterberg H, De Stefano N, Mignarri A. Familial Alzheimer's disease associated with heterozygous NPC1 mutation. J Med Genet 2024; 61:332-339. [PMID: 37989569 DOI: 10.1136/jmg-2023-109219] [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: 02/14/2023] [Accepted: 10/14/2023] [Indexed: 11/23/2023]
Abstract
INTRODUCTION NPC1 mutations are responsible for Niemann-Pick disease type C (NPC), a rare autosomal recessive neurodegenerative disease. Patients harbouring heterozygous NPC1 mutations may rarely show parkinsonism or dementia. Here, we describe for the first time a large family with an apparently autosomal dominant late-onset Alzheimer's disease (AD) harbouring a novel heterozygous NPC1 mutation. METHODS All the five living siblings belonging to the family were evaluated. We performed clinical evaluation, neuropsychological tests, assessment of cerebrospinal fluid markers of amyloid deposition, tau pathology and neurodegeneration (ATN), structural neuroimaging and brain amyloid-positron emission tomography. Oxysterol serum levels were also tested. A wide next-generation sequencing panel of genes associated with neurodegenerative diseases and a whole exome sequencing analysis were performed. RESULTS We detected the novel heterozygous c.3034G>T (p.Gly1012Cys) mutation in NPC1, shared by all the siblings. No other point mutations or deletions in NPC1 or NPC2 were found. In four siblings, a diagnosis of late-onset AD was defined according to clinical characterisation and ATN biomarkers (A+, T+, N+) and serum oxysterol analysis showed increased 7-ketocholesterol and cholestane-3β,5α,6β-triol. DISCUSSION We describe a novel NPC1 heterozygous mutation harboured by different members of a family with autosomal dominant late-onset amnesic AD without NPC-associated features. A missense mutation in homozygous state in the same aminoacidic position has been previously reported in a patient with NPC with severe phenotype. The alteration of serum oxysterols in our family corroborates the pathogenic role of our NPC1 mutation. Our work, illustrating clinical and biochemical disease hallmarks associated with NPC1 heterozygosity in patients affected by AD, provides relevant insights into the pathogenetic mechanisms underlying this possible novel association.
Collapse
Affiliation(s)
- Diego Lopergolo
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Silvia Bianchi
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Gian Nicola Gallus
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Sara Locci
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Barbara Pucci
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Neurofisiologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Desio, ASST Brianza, School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Daniele Gasparini
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Elisa Tardelli
- Unit of Nuclear Medicine, Department of Diagnostic Imaging, PO - S. Stefano, Azienda U.S.L. Toscana Centro, Prato, italy
| | | | - Stelvio Sestini
- Unit of Nuclear Medicine, Department of Diagnostic Imaging, PO - S. Stefano, Azienda U.S.L. Toscana Centro, Prato, italy
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, Calambrone, Italy
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Special Administrative Region, People's Republic of China
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Andrea Mignarri
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| |
Collapse
|
9
|
Fettahoglu A, Zhao M, Khalighi M, Vossler H, Jovin M, Davidzon G, Zeineh M, Boada F, Mormino E, Henderson VW, Moseley M, Chen KT, Zaharchuk G. Early-Frame [ 18F]Florbetaben PET/MRI for Cerebral Blood Flow Quantification in Patients with Cognitive Impairment: Comparison to an [ 15O]Water Gold Standard. J Nucl Med 2024; 65:306-312. [PMID: 38071587 PMCID: PMC10858379 DOI: 10.2967/jnumed.123.266273] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/24/2023] [Indexed: 02/03/2024] Open
Abstract
Cerebral blood flow (CBF) may be estimated from early-frame PET imaging of lipophilic tracers, such as amyloid agents, enabling measurement of this important biomarker in participants with dementia and memory decline. Although previous methods could map relative CBF, quantitative measurement in absolute units (mL/100 g/min) remained challenging and has not been evaluated against the gold standard method of [15O]water PET. The purpose of this study was to develop and validate a minimally invasive quantitative CBF imaging method combining early [18F]florbetaben (eFBB) with phase-contrast MRI using simultaneous PET/MRI. Methods: Twenty participants (11 men and 9 women; 8 cognitively normal, 9 with mild cognitive impairment, and 3 with dementia; 10 β-amyloid negative and 10 β-amyloid positive; 69 ± 9 y old) underwent [15O]water PET, phase-contract MRI, and eFBB imaging in a single session on a 3-T PET/MRI scanner. Quantitative CBF images were created from the first 2 min of brain activity after [18F]florbetaben injection combined with phase-contrast MRI measurement of total brain blood flow. These maps were compared with [15O]water CBF using concordance correlation (CC) and Bland-Altman statistics for gray matter, white matter, and individual regions derived from the automated anatomic labeling (AAL) atlas. Results: The 2 methods showed similar results in gray matter ([15O]water, 55.2 ± 14.7 mL/100 g/min; eFBB, 55.9 ± 14.2 mL/100 g/min; difference, 0.7 ± 2.4 mL/100 g/min; P = 0.2) and white matter ([15O]water, 21.4 ± 5.6 mL/100 g/min; eFBB, 21.2 ± 5.3 mL/100 g/min; difference, -0.2 ± 1.0 mL/100 g/min; P = 0.4). The intrasubject CC for AAL-derived regions was high (0.91 ± 0.04). Intersubject CC in different AAL-derived regions was similarly high, ranging from 0.86 for midfrontal regions to 0.98 for temporal regions. There were no significant differences in performance between the methods in the amyloid-positive and amyloid-negative groups as well as participants with different cognitive statuses. Conclusion: We conclude that eFBB PET/MRI can provide robust CBF measurements, highlighting the capability of simultaneous PET/MRI to provide measurements of both CBF and amyloid burden in a single imaging session in participants with memory disorders.
Collapse
Affiliation(s)
- Ates Fettahoglu
- Department of Radiology, Stanford University, Stanford, California
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Moss Zhao
- Department of Radiology, Stanford University, Stanford, California
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Mehdi Khalighi
- Department of Radiology, Stanford University, Stanford, California
| | - Hillary Vossler
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California; and
| | - Maria Jovin
- Department of Radiology, Stanford University, Stanford, California
| | - Guido Davidzon
- Department of Radiology, Stanford University, Stanford, California
| | - Michael Zeineh
- Department of Radiology, Stanford University, Stanford, California
| | - Fernando Boada
- Department of Radiology, Stanford University, Stanford, California
| | - Elizabeth Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California; and
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California; and
| | - Michael Moseley
- Department of Radiology, Stanford University, Stanford, California
| | - Kevin T Chen
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Greg Zaharchuk
- Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
10
|
Chouliaras L, O'Brien JT. The use of neuroimaging techniques in the early and differential diagnosis of dementia. Mol Psychiatry 2023; 28:4084-4097. [PMID: 37608222 PMCID: PMC10827668 DOI: 10.1038/s41380-023-02215-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
Dementia is a leading cause of disability and death worldwide. At present there is no disease modifying treatment for any of the most common types of dementia such as Alzheimer's disease (AD), Vascular dementia, Lewy Body Dementia (LBD) and Frontotemporal dementia (FTD). Early and accurate diagnosis of dementia subtype is critical to improving clinical care and developing better treatments. Structural and molecular imaging has contributed to a better understanding of the pathophysiology of neurodegenerative dementias and is increasingly being adopted into clinical practice for early and accurate diagnosis. In this review we summarise the contribution imaging has made with particular focus on multimodal magnetic resonance imaging (MRI) and positron emission tomography imaging (PET). Structural MRI is widely used in clinical practice and can help exclude reversible causes of memory problems but has relatively low sensitivity for the early and differential diagnosis of dementia subtypes. 18F-fluorodeoxyglucose PET has high sensitivity and specificity for AD and FTD, while PET with ligands for amyloid and tau can improve the differential diagnosis of AD and non-AD dementias, including recognition at prodromal stages. Dopaminergic imaging can assist with the diagnosis of LBD. The lack of a validated tracer for α-synuclein or TAR DNA-binding protein 43 (TDP-43) imaging remain notable gaps, though work is ongoing. Emerging PET tracers such as 11C-UCB-J for synaptic imaging may be sensitive early markers but overall larger longitudinal multi-centre cross diagnostic imaging studies are needed.
Collapse
Affiliation(s)
- Leonidas Chouliaras
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Specialist Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, St Margaret's Hospital, Epping, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
11
|
Wu CH, Lu YH, Lee TH, Tu CY, Fuh JL, Wang YF, Yang BH. Feasibility evaluation of middle-phase 18F-florbetaben positron emission tomography imaging using centiloid quantification and visual assessment. Quant Imaging Med Surg 2023; 13:4806-4815. [PMID: 37581034 PMCID: PMC10423384 DOI: 10.21037/qims-23-58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/18/2023] [Indexed: 08/16/2023]
Abstract
Background 18F-florbetaben (FBB) positron emission tomography (PET) scan has been widely used in research and routine clinical practice. Most studies used late-phase (scanning from 90 to 110 min after injection) FBB scans to generate beta-amyloid accumulation data. The feasibility of middle-phase scan is seldom discussed. Using the middle-phase data can shorten the patients' waiting between the injection and scan, and hospital can acquire more flexible schedule of routine scan. Methods Paired middle-phase (60-80 min) FBB scans and standard (90-110 min) FBB scans were obtained from 27 subjects (12 neurodegenerative dementia, 8 mild cognitive impairment, 3 normal control, and 4 patients not suffering from neurodegenerative dementia). Standardized uptake value ratios (SUVRs) were calculated and converted to centiloid (CL) scale to investigate the impact on image quantification. CL pipeline validation were performed to build an equation converting the middle-phase data into equivalent standard scans. Cohen's kappa of binary interpretation and brain amyloid plaque load (BAPL) score were also used to evaluate the intrareader agreement of the FBB image from the two protocols. Results The middle-phase FBB SUVR showed an excellent correlation, which provided a linear regression equation of SUVRFBB60-80 = 0.88 × SUVRFBB90-110 + 0.07, with R2=0.98. The slope of the equation indicated that there was bias between the middle and standard acquisition. This can be converted into the CL scale using CL = 174.68 × SUVR - 166.39. Cohen's kappa of binary interpretation and BAPL score were 1.0 (P<0.0001). Conclusions Our findings indicate that the middle-phase FBB protocol is feasible in clinical applications for scans that are at either end of beta-amyloid spectrum, which provides comparable semiquantitative results to standard scan. Patient's waiting time between the injection and scan can be shortened.
Collapse
Affiliation(s)
- Cheng-Han Wu
- Department of Radiology, Taipei Medical University-Shuang Ho Hospital, New Taipei
| | - Yueh-Hsun Lu
- Department of Radiology, Taipei Medical University-Shuang Ho Hospital, New Taipei
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei
- School of Medicine, National Yang Ming Chiao Tung University, Taipei
| | - Tse-Hao Lee
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei
| | - Chun-Yuan Tu
- Department of Medical Imaging and Radiological Technology, Yuanpei University of Medical Technology, Hsinchu
- Association of Medical Radiation Technologists, Taipei
| | - Jong-Ling Fuh
- School of Medicine, National Yang Ming Chiao Tung University, Taipei
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei
| | - Yuh-Feng Wang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei
- Department of Medical Imaging and Radiological Technology, Yuanpei University of Medical Technology, Hsinchu
| | - Bang-Hung Yang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei
| |
Collapse
|
12
|
Choi HJ, Seo M, Kim A, Park SH. Generation of Conventional 18F-FDG PET Images from 18F-Florbetaben PET Images Using Generative Adversarial Network: A Preliminary Study Using ADNI Dataset. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1281. [PMID: 37512092 PMCID: PMC10385186 DOI: 10.3390/medicina59071281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) (PETFDG) image can visualize neuronal injury of the brain in Alzheimer's disease. Early-phase amyloid PET image is reported to be similar to PETFDG image. This study aimed to generate PETFDG images from 18F-florbetaben PET (PETFBB) images using a generative adversarial network (GAN) and compare the generated PETFDG (PETGE-FDG) with real PETFDG (PETRE-FDG) images using the structural similarity index measure (SSIM) and the peak signal-to-noise ratio (PSNR). Materials and Methods: Using the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, 110 participants with both PETFDG and PETFBB images at baseline were included. The paired PETFDG and PETFBB images included six and four subset images, respectively. Each subset image had a 5 min acquisition time. These subsets were randomly sampled and divided into 249 paired PETFDG and PETFBB subset images for the training datasets and 95 paired subset images for the validation datasets during the deep-learning process. The deep learning model used in this study is composed of a GAN with a U-Net. The differences in the SSIM and PSNR values between the PETGE-FDG and PETRE-FDG images in the cycleGAN and pix2pix models were evaluated using the independent Student's t-test. Statistical significance was set at p ≤ 0.05. Results: The participant demographics (age, sex, or diagnosis) showed no statistically significant differences between the training (82 participants) and validation (28 participants) groups. The mean SSIM between the PETGE-FDG and PETRE-FDG images was 0.768 ± 0.135 for the cycleGAN model and 0.745 ± 0.143 for the pix2pix model. The mean PSNR was 32.4 ± 9.5 and 30.7 ± 8.0. The PETGE-FDG images of the cycleGAN model showed statistically higher mean SSIM than those of the pix2pix model (p < 0.001). The mean PSNR was also higher in the PETGE-FDG images of the cycleGAN model than those of pix2pix model (p < 0.001). Conclusions: We generated PETFDG images from PETFBB images using deep learning. The cycleGAN model generated PETGE-FDG images with a higher SSIM and PSNR values than the pix2pix model. Image-to-image translation using deep learning may be useful for generating PETFDG images. These may provide additional information for the management of Alzheimer's disease without extra image acquisition and the consequent increase in radiation exposure, inconvenience, or expenses.
Collapse
Affiliation(s)
- Hyung Jin Choi
- Department of Nuclear Medicine, Ulsan University Hospital, Ulsan 44033, Republic of Korea
| | - Minjung Seo
- Department of Nuclear Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Ahro Kim
- Department of Neurology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Seol Hoon Park
- Department of Nuclear Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| |
Collapse
|
13
|
Schönecker S, Palleis C, Franzmeier N, Katzdobler S, Ferschmann C, Schuster S, Finze A, Scheifele M, Prix C, Fietzek U, Weidinger E, Nübling G, Vöglein J, Patt M, Barthel H, Sabri O, Danek A, Höglinger GU, Brendel M, Levin J. Symptomatology in 4-repeat tauopathies is associated with data-driven topology of [ 18F]-PI-2620 tau-PET signal. Neuroimage Clin 2023; 38:103402. [PMID: 37087820 PMCID: PMC10300609 DOI: 10.1016/j.nicl.2023.103402] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/05/2023] [Accepted: 04/08/2023] [Indexed: 04/25/2023]
Abstract
In recent years in vivo visualization of tau deposits has become possible with various PET radiotracers. The tau tracer [18F]PI-2620 proved high affinity both to 3-repeat/4-repeat tau in Alzheimer's disease as well as to 4-repeat tau in progressive supranuclear palsy (PSP) and corticobasal syndrome (CBS). However, to be clinically relevant, biomarkers should not only correlate with pathological changes but also with disease stage and progression. Therefore, we aimed to investigate the correlation between topology of [18F]PI-2620 uptake and symptomatology in 4-repeat tauopathies. 72 patients with possible or probable 4-repeat tauopathy, i.e. 31 patients with PSP-Richardson's syndrome (PSP-RS), 30 with amyloid-negative CBS and 11 with PSP-non-RS/CBS, underwent [18F]PI-2620-PET. Principal component analysis was performed to identify groups of similar brain regions based on 20-40 min p.i. regional standardized uptake value ratio z-scores. Correlations between component scores and the items of the PSP Rating Scale were explored. Motor signs like gait, arising from chair and postural instability showed a positive correlation with tracer uptake in mesial frontoparietal lobes and the medial superior frontal gyrus and adjacent anterior cingulate cortex. While the signs disorientation and bradyphrenia showed a positive correlation with tracer uptake in the parietooccipital junction, the signs disorientation and arising from chair were negatively correlated with tau-PET signal in the caudate nucleus and thalamus. Total PSP Rating Scale Score showed a trend towards a positive correlation with mesial frontoparietal lobes and a negative correlation with caudate nucleus and thalamus. While in CBS patients, the main finding was a negative correlation of tracer binding in the caudate nucleus and thalamus and a positive correlation of tracer binding in medial frontal cortex with gait and motor signs, in PSP-RS patients various correlations of clinical signs with tracer binding in specific cerebral regions could be detected. Our data reveal [18F]PI-2620 tau-PET topology to correlate with symptomatology in 4-repeat tauopathies. Longitudinal studies will be needed to address whether a deterioration of signs and symptoms over time can be monitored by [18F]PI-2620 in 4-repeat tauopathies and whether [18F]PI-2620 may serve as a marker of disease progression in future therapeutic trials. The detected negative correlation of tracer binding in the caudate nucleus and thalamus with the signs disorientation and arising from chair may be due to an increasing atrophy in these regions leading to partial volume effects and a relative decrease of tracer uptake in the disease course. As cerebral regions correlating with symptomatology differ depending on the clinical phenotype, a precise knowledge of clinical signs and symptoms is necessary when interpreting [18F]PI-2620 PET results.
Collapse
Affiliation(s)
- Sonja Schönecker
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Carla Palleis
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, Ludwig-Maximilians-Universität München, LMU München, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Ferschmann
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Anika Finze
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Catharina Prix
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Urban Fietzek
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Department of Neurology and Clinical Neurophysiology, Schön Klinik München Schwabing, Munich, Germany
| | - Endy Weidinger
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Georg Nübling
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Jonathan Vöglein
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; European Reference Network for Rare Neurological Diseases (ERN-RND), Munich, Germany; Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; European Reference Network for Rare Neurological Diseases (ERN-RND), Munich, Germany.
| |
Collapse
|
14
|
Völter F, Beyer L, Eckenweber F, Scheifele M, Bui N, Patt M, Barthel H, Katzdobler S, Palleis C, Franzmeier N, Levin J, Perneczky R, Rauchmann BS, Sabri O, Hong J, Cumming P, Rominger A, Shi K, Bartenstein P, Brendel M. Assessment of perfusion deficit with early phases of [ 18F]PI-2620 tau-PET versus [ 18F]flutemetamol-amyloid-PET recordings. Eur J Nucl Med Mol Imaging 2023; 50:1384-1394. [PMID: 36572740 PMCID: PMC10027797 DOI: 10.1007/s00259-022-06087-y] [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: 08/24/2022] [Accepted: 12/11/2022] [Indexed: 12/28/2022]
Abstract
PURPOSE Characteristic features of amyloid-PET (A), tau-PET (T), and FDG-PET (N) can serve for the A/T/N classification of neurodegenerative diseases. Recent studies showed that the early, perfusion-weighted phases of amyloid- or tau-PET recordings serve to detect cerebrometabolic deficits equally to FDG-PET, therefore providing a surrogate of neuronal injury. As such, two channels of diagnostic information can be obtained in the setting of a single PET scan. However, there has hitherto been no comparison of early-phase amyloid- and tau-PET as surrogates for deficits in perfusion/metabolism. Therefore, we undertook to compare [18F]flutemetamol-amyloid-PET and [18F]PI-2620 tau-PET as "one-stop shop" dual purpose tracers for the detection of neurodegenerative disease. METHODS We obtained early-phase PET recordings with [18F]PI-2620 (0.5-2.5 min p.i.) and [18F]flutemetamol (0-10 min p.i.) in 64 patients with suspected neurodegenerative disease. We contrasted global mean normalized images (SUVr) in the patients with a normal cohort of 15 volunteers without evidence of increased pathology to β-amyloid- and tau-PET examinations. Regional group differences of tracer uptake (z-scores) of 246 Brainnetome volumes of interest were calculated for both tracers, and the correlations of the z-scores were evaluated using Pearson's correlation coefficient. Lobar compartments, regions with significant neuronal injury (z-scores < - 3), and patients with different neurodegenerative disease entities (e.g., Alzheimer's disease or 4R-tauopathies) served for subgroup analysis. Additionally, we used partial regression to correlate regional perfusion alterations with clinical scores in cognition tests. RESULTS The z-scores of perfusion-weighted images of both tracers showed high correlations across the brain, especially in the frontal and parietal lobes, which were the brain regions with pronounced perfusion deficit in the patient group (R = 0.83 ± 0.08; range, 0.61-0.95). Z-scores of individual patients correlated well by region (R = 0.57 ± 0.15; range, 0.16-0.90), notably when significant perfusion deficits were present (R = 0.66 ± 0.15; range, 0.28-0.90). CONCLUSION The early perfusion phases of [18F]PI-2620 tau- and [18F]flutemetamol-amyloid-PET are roughly equivalent indices of perfusion defect indicative of regional and lobar neuronal injury in patients with various neurodegenerative diseases. As such, either tracer may serve for two diagnostic channels by assessment of amyloid/tau status and neuronal activity.
Collapse
Affiliation(s)
- Friederike Völter
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ngoc Bui
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Sabrina Katzdobler
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | - Johannes Levin
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | | | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Jimin Hong
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, Australia
| | - Axel Rominger
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
15
|
Dai Y, Fang T, Xu Y, Jiang T, Qiao J. Multi-fluorine labeled indanone derivatives as potential MRI imaging probes for β-Amyloid plaques. Chem Biol Drug Des 2023; 101:650-661. [PMID: 36301043 DOI: 10.1111/cbdd.14162] [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: 06/30/2022] [Revised: 10/01/2022] [Accepted: 10/22/2022] [Indexed: 11/29/2022]
Abstract
In order to realize the early diagnosis of Alzheimer's disease (AD), we designed and synthesized a series of multi-fluorine labeled indanone derivatives based on indanone which could target β-amyloid (Aβ). Through the in vitro staining experiment and affinity experiment, we selected 7d out, and then evaluated it through other in vivo and in vitro experiments. The staining of AD human brain adjacent sections revealed that compound 7d could bind to Aβ plaques with high affinity. In the in vitro binding assay, 7d showed a balanced affinity with Aβ1-40 (Kd = 367 ± 13) and Aβ1-42 (Kd = 384 ± 56). Also, 7d exhibited a low toxicity (LD50 > 50 mg/kg) and an excellent ability to pass through the blood-brain barrier (Log p = 3.87). The biodistribution experiment in mice showed that 7d reached the highest brain uptake after 1 h of tail vein injection and cleared after 24 h. A low concentration of 7d (1.875 mg/ml) showed a strong imaging ability (19F-weighted mode), and the imaging capability increased with the increasing of concentration. All the results showed that 7d could provide a feasible solution for the early diagnosis of AD under non-radioactive condition.
Collapse
Affiliation(s)
- Yaqian Dai
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tangni Fang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanhong Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Taoshan Jiang
- School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Jinping Qiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
16
|
Gómez-Grande A, Seiffert AP, Villarejo-Galende A, González-Sánchez M, Llamas-Velasco S, Bueno H, Gómez EJ, Tabuenca MJ, Sánchez-González P. Static first-minute-frame (FMF) PET imaging after 18F-labeled amyloid tracer injection is correlated to [ 18F]FDG PET in patients with primary progressive aphasia. Rev Esp Med Nucl Imagen Mol 2023:S2253-8089(23)00014-9. [PMID: 36758828 DOI: 10.1016/j.remnie.2023.02.001] [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: 07/27/2022] [Accepted: 10/06/2022] [Indexed: 02/10/2023]
Abstract
OBJECTIVE To study the correlation between a static PET image of the first-minute-frame (FMF) acquired with 18F-labeled amyloid-binding radiotracers and brain [18F]FDG PET in patients with primary progressive aphasia (PPA). MATERIAL AND METHODS The study cohort includes 17 patients diagnosed with PPA with the following distribution: 9 nonfluent variant PPA, 4 logopenic variant PPA, 1 semantic variant PPA, 3 unclassifiable PPA. Regional SUVRs are extracted from FMFs and their corresponding [18F]FDG PET images and Pearson's correlation coefficients are calculated. RESULTS SUVRs of both images show similar patterns of regional cerebral alterations. Intrapatient correlation analyses result in a mean coefficient of r=0.94±0.06. Regional interpatient correlation coefficients of the study cohort are greater than 0.81. Radiotracer-specific and variant-specific subcohorts show no difference in the similarity between the images. CONCLUSIONS The static FMF could be a valid alternative to dynamic early-phase amyloid PET proposed in the literature, and a neurodegeneration biomarker for the diagnosis and classification of PPA in amyloid PET studies.
Collapse
Affiliation(s)
- Adolfo Gómez-Grande
- Department of Nuclear Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Alexander P Seiffert
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.
| | - Alberto Villarejo-Galende
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; Group of Neurodegenerative Diseases, Hospital 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Marta González-Sánchez
- Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; Group of Neurodegenerative Diseases, Hospital 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Sara Llamas-Velasco
- Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain; Group of Neurodegenerative Diseases, Hospital 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain; Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Héctor Bueno
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; Department of Cardiology and Instituto de Investigación Sanitaria (imas12), 28041 Hospital Universitario 12 de Octubre, Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Enrique J Gómez
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María José Tabuenca
- Department of Nuclear Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Patricia Sánchez-González
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
17
|
Katzdobler S, Nitschmann A, Barthel H, Bischof G, Beyer L, Marek K, Song M, Wagemann O, Palleis C, Weidinger E, Nack A, Fietzek U, Kurz C, Häckert J, Stapf T, Ferschmann C, Scheifele M, Eckenweber F, Biechele G, Franzmeier N, Dewenter A, Schönecker S, Saur D, Schroeter ML, Rumpf JJ, Rullmann M, Schildan A, Patt M, Stephens AW, van Eimeren T, Neumaier B, Drzezga A, Danek A, Classen J, Bürger K, Janowitz D, Rauchmann BS, Stöcklein S, Perneczky R, Schöberl F, Zwergal A, Höglinger GU, Bartenstein P, Villemagne V, Seibyl J, Sabri O, Levin J, Brendel M. Additive value of [ 18F]PI-2620 perfusion imaging in progressive supranuclear palsy and corticobasal syndrome. Eur J Nucl Med Mol Imaging 2023; 50:423-434. [PMID: 36102964 PMCID: PMC9816230 DOI: 10.1007/s00259-022-05964-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 09/01/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE Early after [18F]PI-2620 PET tracer administration, perfusion imaging has potential for regional assessment of neuronal injury in neurodegenerative diseases. This is while standard late-phase [18F]PI-2620 tau-PET is able to discriminate the 4-repeat tauopathies progressive supranuclear palsy and corticobasal syndrome (4RTs) from disease controls and healthy controls. Here, we investigated whether early-phase [18F]PI-2620 PET has an additive value for biomarker based evaluation of 4RTs. METHODS Seventy-eight patients with 4RTs (71 ± 7 years, 39 female), 79 patients with other neurodegenerative diseases (67 ± 12 years, 35 female) and twelve age-matched controls (69 ± 8 years, 8 female) underwent dynamic (0-60 min) [18F]PI-2620 PET imaging. Regional perfusion (0.5-2.5 min p.i.) and tau load (20-40 min p.i.) were measured in 246 predefined brain regions [standardized-uptake-value ratios (SUVr), cerebellar reference]. Regional SUVr were compared between 4RTs and controls by an ANOVA including false-discovery-rate (FDR, p < 0.01) correction. Hypoperfusion in resulting 4RT target regions was evaluated at the patient level in all patients (mean value - 2SD threshold). Additionally, perfusion and tau pattern expression levels were explored regarding their potential discriminatory value of 4RTs against other neurodegenerative disorders, including validation in an independent external dataset (n = 37), and correlated with clinical severity in 4RTs (PSP rating scale, MoCA, activities of daily living). RESULTS Patients with 4RTs had significant hypoperfusion in 21/246 brain regions, most dominant in thalamus, caudate nucleus, and anterior cingulate cortex, fitting to the topology of the 4RT disease spectrum. However, single region hypoperfusion was not specific regarding the discrimination of patients with 4RTs against patients with other neurodegenerative diseases. In contrast, perfusion pattern expression showed promise for discrimination of patients with 4RTs from other neurodegenerative diseases (AUC: 0.850). Discrimination by the combined perfusion-tau pattern expression (AUC: 0.903) exceeded that of the sole tau pattern expression (AUC: 0.864) and the discriminatory power of the combined perfusion-tau pattern expression was replicated in the external dataset (AUC: 0.917). Perfusion but not tau pattern expression was associated with PSP rating scale (R = 0.402; p = 0.0012) and activities of daily living (R = - 0.431; p = 0.0005). CONCLUSION [18F]PI-2620 perfusion imaging mirrors known topology of regional hypoperfusion in 4RTs. Single region hypoperfusion is not specific for 4RTs, but perfusion pattern expression may provide an additive value for the discrimination of 4RTs from other neurodegenerative diseases and correlates closer with clinical severity than tau pattern expression.
Collapse
Affiliation(s)
- Sabrina Katzdobler
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Alexander Nitschmann
- grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Henryk Barthel
- grid.411339.d0000 0000 8517 9062Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Gerard Bischof
- grid.411097.a0000 0000 8852 305XDepartment of Nuclear Medicine, University Hospital Cologne, Cologne, Germany ,Molecular Organization of the Brain, Institute for Neuroscience and Medicine, INM-2), Jülich, Germany
| | - Leonie Beyer
- grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Ken Marek
- grid.452597.8InviCRO, LLC, Boston, MA USA ,grid.452597.8Molecular Neuroimaging, A Division of inviCRO, New Haven, CT USA
| | - Mengmeng Song
- grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Olivia Wagemann
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Carla Palleis
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Endy Weidinger
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Anne Nack
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Urban Fietzek
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Carolin Kurz
- grid.411095.80000 0004 0477 2585Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Jan Häckert
- grid.411095.80000 0004 0477 2585Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany ,grid.7307.30000 0001 2108 9006Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany
| | - Theresa Stapf
- grid.411095.80000 0004 0477 2585Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Christian Ferschmann
- grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Maximilian Scheifele
- grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Florian Eckenweber
- grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Gloria Biechele
- grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Nicolai Franzmeier
- grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Anna Dewenter
- grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sonja Schönecker
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Dorothee Saur
- grid.9647.c0000 0004 7669 9786Department of Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | - Matthias L. Schroeter
- grid.9647.c0000 0004 7669 9786Clinic for Cognitive Neurology, University of Leipzig, Leipzig, Germany ,grid.9647.c0000 0004 7669 9786LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany ,grid.419524.f0000 0001 0041 5028Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Jost-Julian Rumpf
- grid.9647.c0000 0004 7669 9786Department of Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | - Michael Rullmann
- grid.411339.d0000 0000 8517 9062Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Andreas Schildan
- grid.411339.d0000 0000 8517 9062Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Marianne Patt
- grid.411339.d0000 0000 8517 9062Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | | | - Thilo van Eimeren
- grid.411097.a0000 0000 8852 305XDepartment of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Bernd Neumaier
- grid.411097.a0000 0000 8852 305XDepartment of Nuclear Medicine, University Hospital Cologne, Cologne, Germany ,grid.8385.60000 0001 2297 375XInstitute for Neuroscience and Medicine (INM-3), Cognitive Neuroscience, Research Centre Juelich, Juelich, Germany
| | - Alexander Drzezga
- grid.411097.a0000 0000 8852 305XDepartment of Nuclear Medicine, University Hospital Cologne, Cologne, Germany ,Molecular Organization of the Brain, Institute for Neuroscience and Medicine, INM-2), Jülich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Adrian Danek
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Joseph Classen
- grid.9647.c0000 0004 7669 9786Department of Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | - Katharina Bürger
- grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Daniel Janowitz
- grid.411095.80000 0004 0477 2585Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- grid.7307.30000 0001 2108 9006Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany ,grid.411095.80000 0004 0477 2585Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sophia Stöcklein
- grid.411095.80000 0004 0477 2585Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Robert Perneczky
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany ,grid.7307.30000 0001 2108 9006Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, BKH Augsburg, Augsburg, Germany ,grid.7445.20000 0001 2113 8111Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College, London, UK
| | - Florian Schöberl
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Andreas Zwergal
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Günter U. Höglinger
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany ,grid.10423.340000 0000 9529 9877Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Peter Bartenstein
- grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany ,grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Victor Villemagne
- grid.410678.c0000 0000 9374 3516Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medicine, Austin Health, The University of Melbourne, Melbourne, VIC Australia ,grid.21925.3d0000 0004 1936 9000Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - John Seibyl
- grid.452597.8InviCRO, LLC, Boston, MA USA ,grid.452597.8Molecular Neuroimaging, A Division of inviCRO, New Haven, CT USA
| | - Osama Sabri
- grid.411339.d0000 0000 8517 9062Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Johannes Levin
- grid.411095.80000 0004 0477 2585Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Matthias Brendel
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), Munich, Germany ,grid.411095.80000 0004 0477 2585Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | | |
Collapse
|
18
|
Yoo SW, Ha S, Oh YS, Ryu DW, Yoo JY, Lee KS, Kim JS. Caudate-anchored cognitive connectivity pursuant to orthostatic hypotension in early Parkinson's disease. Sci Rep 2022; 12:22161. [PMID: 36550284 PMCID: PMC9780335 DOI: 10.1038/s41598-022-26811-w] [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: 10/11/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
18F-Florbetaben is a tracer used to evaluate the metabolic activity of and amyloid accumulation in the brain when measured in early- and late-phase, respectively. The metabolism of neural substrates could be viewed as a network and might be an important factor in cognition. Orthostatic hypotension (OH) might play an indirect moderating role in cognition, and its latent influence could modify the inherent cognitive network. This study aimed to identify changes of cognitive connectivity according to orthostatic stress in patients with early Parkinson's disease (PD). This study included 104 early PD patients who were evaluated with a head-up tilt-test and18F-Florbetaben positron emission tomography (PET). Cognition was assessed with a comprehensive neuropsychological battery that gauged attention/working memory, language, visuospatial, memory, and executive functions. PET images were analyzed visually for amyloid deposits, and early-phase images were normalized to obtain standardized uptake ratios (SUVRs) of pre-specified subregions relevant to specific cognitive domains. The caudate nucleus was referenced and paired to these pre-specified regions. The correlations between SUVRs of these regions were assessed and stratified according to presence of orthostatic hypotension. Among the patients studied, 22 (21.2%) participants had orthostatic hypotension. Nineteen patients (18.3%) were positive for amyloid-β accumulation upon visual analysis. Moderate correlations between the caudate and pre-specified subregions were observed (Spearman's rho, range [0.331-0.545]). Cognition did not differ, but the patterns of correlation were altered when the disease was stratified by presence of orthostatic stress. In conclusion, cognition in early PD responds to hemodynamic stress by adapting its neural connections between regions relevant to cognitive functions.
Collapse
Affiliation(s)
- Sang-Won Yoo
- grid.411947.e0000 0004 0470 4224Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seunggyun Ha
- grid.411947.e0000 0004 0470 4224Division of Nuclear Medicine, Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon-Sang Oh
- grid.411947.e0000 0004 0470 4224Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong-Woo Ryu
- grid.411947.e0000 0004 0470 4224Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji-Yeon Yoo
- grid.411947.e0000 0004 0470 4224Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kwang-Soo Lee
- grid.411947.e0000 0004 0470 4224Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joong-Seok Kim
- grid.414966.80000 0004 0647 5752Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul St. Mary’s Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591 Republic of Korea
| |
Collapse
|
19
|
Carneiro CDG, Faria DDP, Coutinho AM, Ono CR, Duran FLDS, da Costa NA, Garcez AT, da Silveira PS, Forlenza OV, Brucki SMD, Nitrini R, Busatto G, Buchpiguel CA. Evaluation of 10-minute post-injection 11C-PiB PET and its correlation with 18F-FDG PET in older adults who are cognitively healthy, mildly impaired, or with probable Alzheimer's disease. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2022; 44:495-506. [PMID: 36420910 PMCID: PMC9561831 DOI: 10.47626/1516-4446-2021-2374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Positron emission tomography (PET) allows in vivo evaluation of molecular targets in neurodegenerative diseases, such as Alzheimer's disease. Mild cognitive impairment is an intermediate stage between normal cognition and Alzheimer-type dementia. In vivo fibrillar amyloid-beta can be detected in PET using [11C]-labeled Pittsburgh compound B (11C-PiB). In contrast, [18F]fluoro-2-deoxy-d-glucose (18F-FDG) is a neurodegeneration biomarker used to evaluate cerebral glucose metabolism, indicating neuronal injury and synaptic dysfunction. In addition, early cerebral uptake of amyloid-PET tracers can determine regional cerebral blood flow. The present study compared early-phase 11C-PiB and 18F-FDG in older adults without cognitive impairment, amnestic mild cognitive impairment, and clinical diagnosis of probable Alzheimer's disease. METHODS We selected 90 older adults, clinically classified as healthy controls, with amnestic mild cognitive impairment, or with probable Alzheimer's disease, who underwent an 18F-FDG PET, early-phase 11C-PiB PET and magnetic resonance imaging. All participants were also classified as amyloid-positive or -negative in late-phase 11C-PiB. The data were analyzed using statistical parametric mapping. RESULTS We found that the probable Alzheimer's disease and amnestic mild cognitive impairment group had lower early-phase 11C-PiB uptake in limbic structures than 18F-FDG uptake. The images showed significant interactions between amyloid-beta status (negative or positive). However, early-phase 11C-PiB appears to provide different information from 18F-FDG about neurodegeneration. CONCLUSIONS Our study suggests that early-phase 11C-PiB uptake correlates with 18F-FDG, irrespective of the particular amyloid-beta status. In addition, we observed distinct regional distribution patterns between both biomarkers, reinforcing the need for more robust studies to investigate the real clinical value of early-phase amyloid-PET imaging.
Collapse
Affiliation(s)
- Camila de Godoi Carneiro
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil,Centro de Investigação Translacional em Oncologia, Departamento de Radiologia e Oncologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Daniele de Paula Faria
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil,Centro de Investigação Translacional em Oncologia, Departamento de Radiologia e Oncologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Artur Martins Coutinho
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Carla Rachel Ono
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Fábio Luís de Souza Duran
- Laboratório Neuro-Imagem em Psiquiatria (LIM 21), Departamento de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Naomi Antunes da Costa
- Laboratório Neuro-Imagem em Psiquiatria (LIM 21), Departamento de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Alexandre Teles Garcez
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Paula Squarzoni da Silveira
- Laboratório Neuro-Imagem em Psiquiatria (LIM 21), Departamento de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Orestes Vicente Forlenza
- Laboratório de Neurociências (LIM 27), Departamento de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Sonia Maria Dozzi Brucki
- Departamento de Neurologia, Hospital das Clínicas, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Ricardo Nitrini
- Departamento de Neurologia, Hospital das Clínicas, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Geraldo Busatto
- Laboratório Neuro-Imagem em Psiquiatria (LIM 21), Departamento de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| | - Carlos Alberto Buchpiguel
- Laboratório de Medicina Nuclear (LIM 43), Departamento de Radiologia e Oncologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil,Correspondence: Carlos Alberto Buchpiguel, Universidade de São Paulo, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455, CEP 01255-090, São Paulo, SP, Brazil. E-mail:
| |
Collapse
|
20
|
Alzheimer’s Disease Prediction Using Attention Mechanism with Dual-Phase 18F-Florbetaben Images. Nucl Med Mol Imaging 2022; 57:61-72. [PMID: 36998590 PMCID: PMC10043070 DOI: 10.1007/s13139-022-00767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/04/2022] [Accepted: 08/02/2022] [Indexed: 10/15/2022] Open
Abstract
Abstract
Introduction
Amyloid-beta (Aβ) imaging test plays an important role in the early diagnosis and research of biomarkers of Alzheimer’s disease (AD) but a single test may produce Aβ-negative AD or Aβ-positive cognitively normal (CN). In this study, we aimed to distinguish AD from CN with dual-phase 18F-Florbetaben (FBB) via a deep learning–based attention method and evaluate the AD positivity scores compared to late-phase FBB which is currently adopted for AD diagnosis.
Materials and Methods
A total of 264 patients (74 CN and 190 AD), who underwent FBB imaging test and neuropsychological tests, were retrospectively analyzed. Early- and delay-phase FBB images were spatially normalized with an in-house FBB template. The regional standard uptake value ratios were calculated with the cerebellar region as a reference region and used as independent variables that predict the diagnostic label assigned to the raw image.
Results
AD positivity scores estimated from dual-phase FBB showed better accuracy (ACC) and area under the receiver operating characteristic curve (AUROC) for AD detection (ACC: 0.858, AUROC: 0.831) than those from delay phase FBB imaging (ACC: 0.821, AUROC: 0.794). AD positivity score estimated by dual-phase FBB (R: −0.5412) shows a higher correlation with psychological test compared to only dFBB (R: −0.2975). In the relevance analysis, we observed that LSTM uses different time and regions of early-phase FBB for each disease group for AD detection.
Conclusions
These results show that the aggregated model with dual-phase FBB with long short-term memory and attention mechanism can be used to provide a more accurate AD positivity score, which shows a closer association with AD, than the prediction with only a single phase FBB.
Collapse
|
21
|
Boccalini C, Peretti DE, Ribaldi F, Scheffler M, Stampacchia S, Tomczyk S, Rodriguez C, Montandon ML, Haller S, Giannakopoulos P, Frisoni GB, Perani D, Garibotto V. Early-phase 18F-Florbetapir and 18F-Flutemetamol images as proxies of brain metabolism in a memory clinic setting. J Nucl Med 2022; 64:jnumed.122.264256. [PMID: 35863896 PMCID: PMC9902851 DOI: 10.2967/jnumed.122.264256] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Alzheimer's disease (AD) neuropathologic changes are β-amyloid (Aβ) deposition, pathologic tau, and neurodegeneration. Dual-phase amyloid-PET might be able to evaluate Aβ deposition and neurodegeneration with a single tracer injection. Early-phase amyloid-PET scans provide a proxy for cerebral perfusion, which has shown good correlations with neural dysfunction measured through metabolic consumption, while the late frames depict amyloid distribution. Our study aims to assess the comparability between early-phase amyloid-PET scans and 18F-fluorodeoxyglucose (18F-FDG)-PET brain topography at the individual level, and their ability to discriminate patients. Methods: 166 subjects evaluated at the Geneva Memory Center, ranging from cognitively unimpaired to Mild Cognitive Impairment (MCI) and dementia, underwent early-phase amyloid-PET - using either 18F-florbetapir (eFBP) (n = 94) or 18F-flutemetamol (eFMM) (n = 72) - and 18F-FDG-PET. Aβ status was assessed. Standardized uptake value ratios (SUVR) were extracted to evaluate the correlation of eFBP/eFMM and their respective 18F-FDG-PET scans. The single-subject procedure was applied to investigate hypometabolism and hypoperfusion maps and their spatial overlap by Dice coefficient. Receiver operating characteristic analyses were performed to compare the discriminative power of eFBP/eFMM, and 18F-FDG-PET SUVR in AD-related metaROI between Aβ-negative healthy controls and cases in the AD continuum. Results: Positive correlations were found between eFBP/eFMM and 18F-FDG-PET SUVR independently of Aβ status and Aβ radiotracer (R>0.72, p<0.001). eFBP/eFMM single-subject analysis revealed clusters of significant hypoperfusion with good correspondence to hypometabolism topographies, independently of the underlying neurodegenerative patterns. Both eFBP/eFMM and 18F-FDG-PET SUVR significantly discriminated AD patients from controls in the AD-related metaROIs (AUCFBP = 0.888; AUCFMM=0.801), with 18F-FDG-PET performing slightly better, however not significantly (all p-value higher than 0.05), than others (AUCFDG=0.915 and 0.832 for subjects evaluated with 18F-FBP and 18F-FMM, respectively). Conclusion: The distribution of perfusion was comparable to that of metabolism at the single-subject level by parametric analysis, particularly in the presence of a high neurodegeneration burden. Our findings indicate that eFBP/eFMM imaging can replace 18F-FDG-PET imaging, as they reveal typical neurodegenerative patterns, or allow to exclude the presence of neurodegeneration. The finding shows cost-saving capacities of amyloid-PET and supports the routine use of the modality for individual classification in clinical practice.
Collapse
Affiliation(s)
- Cecilia Boccalini
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Vita-Salute San Raffaele University, Milan, Italy
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Débora Elisa Peretti
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Max Scheffler
- Division of Radiology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Sara Stampacchia
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Szymon Tomczyk
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
| | - Cristelle Rodriguez
- Division of Institutional Measures, Medical Direction, University Hospitals of Geneva, Geneva, Switzerland
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marie-Louise Montandon
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Rehabilitation and Geriatrics, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Sven Haller
- CIMC–Centre d’Imagerie Médicale de Cornavin, Geneva, Switzerland
- Faculty of Medicine of University of Geneva, Geneva, Switzerland
- Division of Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Panteleimon Giannakopoulos
- Division of Institutional Measures, Medical Direction, University Hospitals of Geneva, Geneva, Switzerland
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Giovanni B. Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Daniela Perani
- Vita-Salute San Raffaele University, Milan, Italy
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland; and
- CIBM Center for Biomedical Imaging, Geneva, Switzerland
| |
Collapse
|
22
|
Matthews DC, Lukic AS, Andrews RD, Wernick MN, Strother SC, Schmidt ME. Measurement of neurodegeneration using a multivariate early frame amyloid PET classifier. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12325. [PMID: 35846158 PMCID: PMC9270637 DOI: 10.1002/trc2.12325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/28/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022]
Abstract
Introduction Amyloid measurement provides important confirmation of pathology for Alzheimer's disease (AD) clinical trials. However, many amyloid positive (Am+) early-stage subjects do not worsen clinically during a clinical trial, and a neurodegenerative measure predictive of decline could provide critical information. Studies have shown correspondence between perfusion measured by early amyloid frames post-tracer injection and fluorodeoxyglucose (FDG) positron emission tomography (PET), but with limitations in sensitivity. Multivariate machine learning approaches may offer a more sensitive means for detection of disease related changes as we have demonstrated with FDG. Methods Using summed dynamic florbetapir image frames acquired during the first 6 minutes post-injection for 107 Alzheimer's Disease Neuroimaging Initiative subjects, we applied optimized machine learning to develop and test image classifiers aimed at measuring AD progression. Early frame amyloid (EFA) classification was compared to that of an independently developed FDG PET AD progression classifier by scoring the FDG scans of the same subjects at the same time point. Score distributions and correlation with clinical endpoints were compared to those obtained from FDG. Region of interest measures were compared between EFA and FDG to further understand discrimination performance. Results The EFA classifier produced a primary pattern similar to that of the FDG classifier whose expression correlated highly with the FDG pattern (R-squared 0.71), discriminated cognitively normal (NL) amyloid negative (Am-) subjects from all Am+ groups, and that correlated in Am+ subjects with Mini-Mental State Examination, Clinical Dementia Rating Sum of Boxes, and Alzheimer's Disease Assessment Scale-13-item Cognitive subscale (R = 0.59, 0.63, 0.73) and with subsequent 24-month changes in these measures (R = 0.67, 0.73, 0.50). Discussion Our results support the ability to use EFA with a multivariate machine learning-derived classifier to obtain a sensitive measure of AD-related loss in neuronal function that correlates with FDG PET in preclinical and early prodromal stages as well as in late mild cognitive impairment and dementia. Highlights The summed initial post-injection minutes of florbetapir positron emission tomography correlate with fluorodeoxyglucose.A machine learning classifier enabled sensitive detection of early prodromal Alzheimer's disease.Early frame amyloid (EFA) classifier scores correlate with subsequent change in Mini-Mental State Examination, Clinical Dementia Rating Sum of Boxes, and Alzheimer's Disease Assessment Scale-13-item Cognitive subscale.EFA classifier effect sizes and clinical prediction outperformed region of interest standardized uptake value ratio.EFA classification may aid in stratifying patients to assess treatment effect.
Collapse
Affiliation(s)
| | | | | | | | - Stephen C. Strother
- Baycrest Hospitaland Department of Medical BiophysicsUniversity of TorontoNorth YorkOntarioCanada
| | - Mark E. Schmidt
- Janssen Research and DevelopmentDivision of Janssen PharmaceuticaBeerseBelgium
| | | |
Collapse
|
23
|
Peira E, Poggiali D, Pardini M, Barthel H, Sabri O, Morbelli S, Cagnin A, Chincarini A, Cecchin D. A comparison of advanced semi-quantitative amyloid PET analysis methods. Eur J Nucl Med Mol Imaging 2022; 49:4097-4108. [PMID: 35652962 PMCID: PMC9525368 DOI: 10.1007/s00259-022-05846-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/18/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE To date, there is no consensus on how to semi-quantitatively assess brain amyloid PET. Some approaches use late acquisition alone (e.g., ELBA, based on radiomic features), others integrate the early scan (e.g., TDr, which targets the area of maximum perfusion) and structural imaging (e.g., WMR, that compares kinetic behaviour of white and grey matter, or SI based on the kinetic characteristics of the grey matter alone). In this study SUVr, ELBA, TDr, WMR, and SI were compared. The latter - the most complete one - provided the reference measure for amyloid burden allowing to assess the efficacy and feasibility in clinical setting of the other approaches. METHODS We used data from 85 patients (aged 44-87) who underwent dual time-point PET/MRI acquisitions. The correlations with SI were computed and the methods compared with the visual assessment. Assuming SUVr, ELBA, TDr, and WMR to be independent measures, we linearly combined them to obtain more robust indices. Finally, we investigated possible associations between each quantifier and age in amyloid-negative patients. RESULTS Each quantifier exhibited excellent agreement with visual assessment and strong correlation with SI (average AUC = 0.99, ρ = 0.91). Exceptions to this were observed for subcortical regions with ELBA and WMR (ρELBA = 0.44, ρWMR = 0.70). The linear combinations showed better performances than the individual methods. Significant associations were observed between TDr, WMR, SI, and age in amyloid-negative patients (p < 0.05). CONCLUSION Among the other methods, TDr came closest to the reference with less implementation complexity. Moreover, this study suggests that combining independent approaches gives better results than the individual procedure, so efforts should focus on multi-classifier systems for amyloid PET. Finally, the ability of techniques integrating blood perfusion to depict age-related variations in amyloid load in amyloid-negative subjects demonstrates the goodness of the estimate.
Collapse
Affiliation(s)
- Enrico Peira
- INFN - National Institute of Nuclear Physics, via Dodecaneso 33, 16146, Genoa, Italy.
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health (DINOGMI), University of Genoa, Genoa, Italy.
| | - Davide Poggiali
- PNC - Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Nuclear Medicine Unit, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Annachiara Cagnin
- Neurology Unit, Department of Neurology, University Hospital of Padua, Padua, Italy
| | - Andrea Chincarini
- INFN - National Institute of Nuclear Physics, via Dodecaneso 33, 16146, Genoa, Italy
| | - Diego Cecchin
- PNC - Padua Neuroscience Center, University of Padua, Padua, Italy
- Nuclear Medicine Unit, Department of Medicine - DIMED, University Hospital of Padua, Padua, Italy
| |
Collapse
|
24
|
Correlación entre el metabolismo de la glucosa cerebral (18F-FDG) y el flujo sanguíneo cerebral con marcadores de amiloide (18F-florbetapir) en práctica clínica: evidencias preliminares. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2021.02.004] [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]
|
25
|
SimulAD: A dynamical model for personalized simulation and disease staging in Alzheimer’s disease. Neurobiol Aging 2022; 113:73-83. [DOI: 10.1016/j.neurobiolaging.2021.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 12/13/2021] [Accepted: 12/30/2021] [Indexed: 11/17/2022]
|
26
|
Imaging Clinical Subtypes and Associated Brain Networks in Alzheimer’s Disease. Brain Sci 2022; 12:brainsci12020146. [PMID: 35203910 PMCID: PMC8869882 DOI: 10.3390/brainsci12020146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) does not present uniform symptoms or a uniform rate of progression in all cases. The classification of subtypes can be based on clinical symptoms or patterns of pathological brain alterations. Imaging techniques may allow for the identification of AD subtypes and their differentiation from other neurodegenerative diseases already at an early stage. In this review, the strengths and weaknesses of current clinical imaging methods are described. These include positron emission tomography (PET) to image cerebral glucose metabolism and pathological amyloid or tau deposits. Magnetic resonance imaging (MRI) is more widely available than PET. It provides information on structural or functional changes in brain networks and their relation to AD subtypes. Amyloid PET provides a very early marker of AD but does not distinguish between AD subtypes. Regional patterns of pathology related to AD subtypes are observed with tau and glucose PET, and eventually as atrophy patterns on MRI. Structural and functional network changes occur early in AD but have not yet provided diagnostic specificity.
Collapse
|
27
|
Myoraku A, Klein G, Landau S, Tosun D. Regional uptakes from early-frame amyloid PET and 18F-FDG PET scans are comparable independent of disease state. Eur J Hybrid Imaging 2022; 6:2. [PMID: 35039928 PMCID: PMC8763988 DOI: 10.1186/s41824-021-00123-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/10/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose Positron emission tomography (PET) imaging with amyloid-beta (Aβ) tracers and 2-[18F] fluoro-2-Deoxy-d-glucose (18F-FDG) is extensively employed in Alzheimer’s disease (AD) studies as biomarkers of AD pathology and neurodegeneration. To reduce cost and additional burdens to the patient, early-frame uptake during Aβ PET scanning has been proposed as a surrogate measure of regional glucose metabolism. Considering the disease state specific impact of AD on neurovascular coupling, we investigated to what extent the information captured in the early frames of an Aβ-PET (18F-florbetapir or 18F-florbetaben) scan is comparable to that of a 18F-FDG PET scan, independent of disease state. Method A partial correlation was performed on early-frame 18F-florbetapir and 18F-FDG regional data from 100 participants. In a secondary analysis, we compared 92 18F-florbetapir and 21 18F-florbetaben early-frame Aβ scans from cognitively unimpaired and mild cognitive impairment participants to ascertain if regional early-frame information was similar across different Aβ-PET radioligands. Results The partial correlation of early-frame 18F-florbetapir with 18F-FDG was significant in all 84 brain ROIs, with correlation values ranging from 0.61 to 0.94. There were no significant differences between early-frame 18F-florbetapir and 18F-florbetaben images. Conclusion Overall, we find that the regional uptake measurements from early-frame 18F-florbetapir are strongly correlated with regional glucose metabolism as measured in ground-truth 18F-FDG PET scans, regardless of disease state. Future studies should focus on longitudinal early-frame amyloid PET imaging studies to further assess the value of early-frame imaging as a marker of brain metabolic decline.
Supplementary Information The online version contains supplementary material available at 10.1186/s41824-021-00123-0.
Collapse
Affiliation(s)
- Alison Myoraku
- Northern California Institute for Research and Education, VA Medical Center, 4150 Clement Street, 114M, San Francisco, CA, 94121, USA. .,Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA.
| | - Gregory Klein
- Roche Pharma Research and Early Development, Basel, Switzerland
| | - Susan Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720-3190, USA
| | - Duygu Tosun
- Northern California Institute for Research and Education, VA Medical Center, 4150 Clement Street, 114M, San Francisco, CA, 94121, USA.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94143, USA
| | | |
Collapse
|
28
|
Kim YK. Recent Updates on PET Imaging in Neurodegenerative Diseases. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2022; 83:453-472. [PMID: 36238518 PMCID: PMC9514517 DOI: 10.3348/jksr.2022.0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 11/23/2022]
Abstract
양전자방출단층촬영(PET)을 이용한 단백질병리의 생체영상기술은 퇴행성 치매의 질병 기전을 이해하는데 필요한 정보를 제공할 뿐 아니라, 질병의 조기 발견과 치료법 개발에서 중요한 역할을 수행하고 있다. 베타아밀로이드와 타우 PET 영상은 인체 뇌병리에 기반한 알츠하이머병 연속체에 대한 진단 바이오마커로 확립되어 조기진단과 감별진단을 용이하게 하고, 질병 예후를 예측하고 있다. 또한, 치매치료제 개발에서 예후 및 대리 바이오마커로의 역할이 커지고 있다. 이 종설에서는 치매를 유발하는 알츠하이머병 및 기타 퇴행성 뇌질환에서 베타아밀로이드와 타우 단백질의 뇌축적을 영상화하는 PET의 최근 임상적 적용과 최근 동향을 살펴보고, 잠재적 유용성을 소개하고자 한다.
Collapse
Affiliation(s)
- Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| |
Collapse
|
29
|
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
|
30
|
Wakasugi N, Hanakawa T. It Is Time to Study Overlapping Molecular and Circuit Pathophysiologies in Alzheimer's and Lewy Body Disease Spectra. Front Syst Neurosci 2021; 15:777706. [PMID: 34867224 PMCID: PMC8637125 DOI: 10.3389/fnsys.2021.777706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/28/2021] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia due to neurodegeneration and is characterized by extracellular senile plaques composed of amyloid β1 - 42 (Aβ) as well as intracellular neurofibrillary tangles consisting of phosphorylated tau (p-tau). Dementia with Lewy bodies constitutes a continuous spectrum with Parkinson's disease, collectively termed Lewy body disease (LBD). LBD is characterized by intracellular Lewy bodies containing α-synuclein (α-syn). The core clinical features of AD and LBD spectra are distinct, but the two spectra share common cognitive and behavioral symptoms. The accumulation of pathological proteins, which acquire pathogenicity through conformational changes, has long been investigated on a protein-by-protein basis. However, recent evidence suggests that interactions among these molecules may be critical to pathogenesis. For example, Aβ/tau promotes α-syn pathology, and α-syn modulates p-tau pathology. Furthermore, clinical evidence suggests that these interactions may explain the overlapping pathology between AD and LBD in molecular imaging and post-mortem studies. Additionally, a recent hypothesis points to a common mechanism of prion-like progression of these pathological proteins, via neural circuits, in both AD and LBD. This suggests a need for understanding connectomics and their alterations in AD and LBD from both pathological and functional perspectives. In AD, reduced connectivity in the default mode network is considered a hallmark of the disease. In LBD, previous studies have emphasized abnormalities in the basal ganglia and sensorimotor networks; however, these account for movement disorders only. Knowledge about network abnormalities common to AD and LBD is scarce because few previous neuroimaging studies investigated AD and LBD as a comprehensive cohort. In this paper, we review research on the distribution and interactions of pathological proteins in the brain in AD and LBD, after briefly summarizing their clinical and neuropsychological manifestations. We also describe the brain functional and connectivity changes following abnormal protein accumulation in AD and LBD. Finally, we argue for the necessity of neuroimaging studies that examine AD and LBD cases as a continuous spectrum especially from the proteinopathy and neurocircuitopathy viewpoints. The findings from such a unified AD and Parkinson's disease (PD) cohort study should provide a new comprehensive perspective and key data for guiding disease modification therapies targeting the pathological proteins in AD and LBD.
Collapse
Affiliation(s)
- Noritaka Wakasugi
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Hanakawa
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Integrated Neuroanatomy and Neuroimaging, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
31
|
Sepehrizadeh T, Jong I, DeVeer M, Malhotra A. PET/MRI in paediatric disease. Eur J Radiol 2021; 144:109987. [PMID: 34649143 DOI: 10.1016/j.ejrad.2021.109987] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Nuclear medicine and molecular imaging have a small but growing role in the management of paediatric and neonatal diseases. During the past decade, combined PET/MRI has emerged as a clinically important hybrid imaging modality in paediatric medicine due to diagnostic advantages and reduced radiation exposure compared to alternative techniques. The applications for nuclear medicine, radiopharmaceuticals and combined PET/MRI in paediatric diagnosis is broadly similar to adults, however there are some key differences. There are a variety of clinical applications for PET/MRI imaging in children including, but not limited to, oncology, neurology, cardiovascular, infection and chronic inflammatory diseases, and in renal-urological disorders. In this article, we review the applications of PET/MRI in paediatric and neonatal imaging, its current role, advantages and disadvantages over other hybrid imaging techniques such as PET/CT, and its future applications. Overall, PET/MRI is a powerful imaging technology in diagnostic medicine and paediatric diseases. Higher soft tissue contrasts and lower radiation dose of the MRI makes it the superior technology compared to other conventional techniques such as PET/CT or scintigraphy. However, this relatively new hybrid imaging has also some limitations. MRI based attenuation correction remains a challenge and although methodologies have improved significantly in the last decades, most remain under development.
Collapse
Affiliation(s)
| | - Ian Jong
- Department of diagnostic imaging, Monash Health, Melbourne, Australia
| | - Michael DeVeer
- Monash Biomedical Imaging, Monash University, Melbourne, Australia
| | - Atul Malhotra
- Monash Newborn, Monash Children's Hospital, Melbourne, Australia; Department of Paediatrics, Monash University, Melbourne, Australia
| |
Collapse
|
32
|
Kwon SJ, Ha S, Yoo SW, Shin NY, O JH, Yoo IR, Kim JS. Comparison of early F-18 Florbetaben PET/CT to Tc-99m ECD SPECT using voxel, regional, and network analysis. Sci Rep 2021; 11:16738. [PMID: 34408171 PMCID: PMC8373880 DOI: 10.1038/s41598-021-95808-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/30/2021] [Indexed: 11/09/2022] Open
Abstract
This study aimed to validate early-phase F-18 Florbetaben positron emission tomography (eFBB PET) as a brain perfusion test and determine the optimal reference region. A total of 27 patients with early Parkinson's disease with Tc-99m ethyl cysteinate dimer single photon emission tomography (ECD SPECT) and FBB PET were included. Six reference regions, including whole brain (GN), pons, central white matter (CWM), whole cerebellum (WC), WC with brain stem (WC + B), and cerebellar grey matter (CG), were applied to obtain SUVR using cortex volume-of-interest (VOI). Reference regions of WC (r 0.886), WC + B (r 0.897), and CG (r 0.904) had highest correlation values of cortex-VOI SUVR between both perfusion images (all p < 0.001). Early-phase FBB PET had a significant linear correlation of CG-normalized SUVR of the cortex, basal ganglia, thalamus, and midbrain with ECD SPECT in voxel-wise analysis (FDR adjusted-p < 0.05). Early-phase FBB PET extracts more ICNS than ECD SPECT, as 9 ICNS and 4 ICNs, respectively. Both eFBB PET and ECD SPECT well discriminated PD from DLB (Area-under-curve of receiver-operating-characteristics, 0.911 for eFBB PET, 0.922 for ECD SPECT). Our findings suggest that eFBB PET is a reliable perfusion test based on a high correlation with ECD SPECT using cerebellum-based normalization methods.
Collapse
Affiliation(s)
- Soo Jin Kwon
- Division of Nuclear Medicine, Department of Radiology, Seoul 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, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Sang-Won Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Na-Young Shin
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joo Hyun O
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Ie Ryung Yoo
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Joong-Seok Kim
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
33
|
Yoo SW, Ha S, Yoon H, Yoo JY, Lee KS, Kim JS. Paradoxical Cerebral Perfusion in Parkinson's Disease Patients with Orthostatic Hypotension: A Dual-Phase 18F-Florbetaben Positron Emission Tomography Study. JOURNAL OF PARKINSONS DISEASE 2021; 11:1335-1344. [PMID: 34024782 DOI: 10.3233/jpd-212596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Orthostatic hypotension (OH) may antedate Parkinson's disease (PD) or be found in early stages of the disease. OH may induce a PD brain to chronic hypotensive insults. 18F-Florbetaben (18F-FBB) tracer has a high first-pass influx rate and can be used with positron emission tomography (PET) as a surrogate marker for early- and late-phase evaluation of cerebral perfusion and cerebral amyloidosis, respectively. OBJECTIVE In this study, we evaluated whether 18F-FBB uptake in the early- and late-phases of PD was related to OH. This study manipulated the imaging modality to illustrate the physiology of cerebral flow with OH in PD (PD + OH). METHODS A group of 73 early-stage PD patients was evaluated with a head-up tilt-test and 18F-FBB PET imaging. The cognitive status was assessed by a comprehensive battery of neuropsychological tests. PET images were normalized, and both early- and late-phase standardized uptake value ratios (SUVRs) of pre-specified regions were obtained. The associations between regional SUVRs and OH and cognitive status were analyzed. RESULTS Twenty (27.4%) participants had OH. Thirteen (17.8%) patients were interpreted as having amyloid pathology based on regional 18F-FBB uptake. Early-phase SUVRs were higher in specific brain regions of PD + OH patients than those without OH. However, late-phase SUVRs did not differ between the groups. The early-phase SUVRs were not influenced by amyloid burden or by interaction between amyloid and orthostatic hypotension. Cognitive functions were not disparate when PD + OH patients were contrasted with non-OH patients in this study. CONCLUSION Cerebral blood flow was elevated in patients with early PD + OH. This finding suggests augmented cerebral perfusion in PD + OH might be a compensatory regulation in response to chronic OH.
Collapse
Affiliation(s)
- Sang-Won Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyukjin Yoon
- Division of Nuclear Medicine, Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji-Yeon Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kwang-Soo Lee
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joong-Seok Kim
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
34
|
Seiffert AP, Gómez-Grande A, Villarejo-Galende A, González-Sánchez M, Bueno H, Gómez EJ, Sánchez-González P. High Correlation of Static First-Minute-Frame (FMF) PET Imaging after 18F-Labeled Amyloid Tracer Injection with [ 18F]FDG PET Imaging. SENSORS (BASEL, SWITZERLAND) 2021; 21:5182. [PMID: 34372416 PMCID: PMC8348394 DOI: 10.3390/s21155182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 01/17/2023]
Abstract
Dynamic early-phase PET images acquired with radiotracers binding to fibrillar amyloid-beta (Aβ) have shown to correlate with [18F]fluorodeoxyglucose (FDG) PET images and provide perfusion-like information. Perfusion information of static PET scans acquired during the first minute after radiotracer injection (FMF, first-minute-frame) is compared to [18F]FDG PET images. FMFs of 60 patients acquired with [18F]florbetapir (FBP), [18F]flutemetamol (FMM), and [18F]florbetaben (FBB) are compared to [18F]FDG PET images. Regional standardized uptake value ratios (SUVR) are directly compared and intrapatient Pearson's correlation coefficients are calculated to evaluate the correlation of FMFs to their corresponding [18F]FDG PET images. Additionally, regional interpatient correlations are calculated. The intensity profiles of mean SUVRs among the study cohort (r = 0.98, p < 0.001) and intrapatient analyses show strong correlations between FMFs and [18F]FDG PET images (r = 0.93 ± 0.05). Regional VOI-based analyses also result in high correlation coefficients. The FMF shows similar information to the cerebral metabolic patterns obtained by [18F]FDG PET imaging. Therefore, it could be an alternative to the dynamic imaging of early phase amyloid PET and be used as an additional neurodegeneration biomarker in amyloid PET studies in routine clinical practice while being acquired at the same time as amyloid PET images.
Collapse
Affiliation(s)
- Alexander P. Seiffert
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain;
| | - Adolfo Gómez-Grande
- Department of Nuclear Medicine, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.V.-G.); (H.B.)
| | - Alberto Villarejo-Galende
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.V.-G.); (H.B.)
- Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
- Group of Neurodegenerative Diseases, Hospital 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Marta González-Sánchez
- Department of Neurology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
- Group of Neurodegenerative Diseases, Hospital 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Héctor Bueno
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.V.-G.); (H.B.)
- Department of Cardiology and Instituto de Investigación Sanitaria (imas12), Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Enrique J. Gómez
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Patricia Sánchez-González
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
35
|
Early-phase 18F-FP-CIT and 18F-flutemetamol PET were significantly correlated. Sci Rep 2021; 11:12297. [PMID: 34112926 PMCID: PMC8192502 DOI: 10.1038/s41598-021-91891-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/02/2021] [Indexed: 02/02/2023] Open
Abstract
Little is known about whether early-phase PET images of 18F-FP-CIT match those of amyloid PET. Here, we compared early-phase 18F-FP-CIT and 18F-flutemetamol PET images in patients who underwent both within a 1-month interval. The SUVR on early-phase 18F-FP-CIT PET (median, 0.86) was significantly lower than that of 18F-flutemetamol PET (median, 0.91, p < 0.001) for total brain regions including all cerebral lobes and central structures. This significant difference persisted for each brain region except central structures (p = 0.232). The SUVR of total brain regions obtained from early 18F-FP-CIT PET showed a very strong correlation with that of 18F-flutemetamol PET (rho = 0.80, p < 0.001). Among the kinetic parameters, only R1 showed a statistically significant correlation between the two techniques for all brain regions (rho = 0.89, p < 0.001). R1 from 18F-FP-CIT (median, 0.77) was significantly lower in all areas of the brain compared to R1 from 18F-flutemetamol PET (median, 0.81, p < 0.001).18F-FP-CIT demonstrated lower uptake in cortical brain regions than 18F-flutemetamol on early-phase PET. However, both early-phase PETs demonstrated significant correlation of uptake.
Collapse
|
36
|
Schmitt J, Palleis C, Sauerbeck J, Unterrainer M, Harris S, Prix C, Weidinger E, Katzdobler S, Wagemann O, Danek A, Beyer L, Rauchmann BS, Rominger A, Simons M, Bartenstein P, Perneczky R, Haass C, Levin J, Höglinger GU, Brendel M. Dual-Phase β-Amyloid PET Captures Neuronal Injury and Amyloidosis in Corticobasal Syndrome. Front Aging Neurosci 2021; 13:661284. [PMID: 34054506 PMCID: PMC8155727 DOI: 10.3389/fnagi.2021.661284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: In recent years several 18F-labeled amyloid PET (Aβ-PET) tracers have been developed and have obtained clinical approval. There is evidence that Aβ-PET perfusion can provide surrogate information about neuronal injury in neurodegenerative diseases when compared to conventional blood flow and glucose metabolism assessment. However, this paradigm has not yet been tested in neurodegenerative disorders with cortical and subcortical affection. Therefore, we investigated the performance of early acquisition 18F-flutemetamol Aβ-PET in comparison to 18F-fluorodeoxyglucose (FDG)-PET in corticobasal syndrome (CBS). Methods: Subjects with clinically possible or probable CBS were recruited within the prospective Activity of Cerebral Networks, Amyloid and Microglia in Aging and Alzheimer's Disease (ActiGliA) observational study and all CBS cases with an available FDG-PET prior to Aβ-PET were selected. Aβ-PET was acquired 0-10 min p.i. (early-phase) and 90-110 min p.i. (late-phase) whereas FDG-PET was recorded statically from 30 to 50 min p.i. Semiquantitative regional values and asymmetry indices (AI) were compared between early-phase Aβ-PET and FDG-PET. Visual assessments of hypoperfusion and hypometabolism were compared between both methods. Late-phase Aβ-PET was evaluated visually for assessment of Aβ-positivity. Results: Among 20 evaluated patients with CBS, 5 were Aβ-positive. Early-phase Aβ-PET and FDG-PET SUVr correlated highly in cortical (mean R = 0.86, range 0.77-0.92) and subcortical brain regions (mean R = 0.84, range 0.79-0.90). Strong asymmetry was observed in FDG-PET for the motor cortex (mean |AI| = 2.9%), the parietal cortex (mean |AI| = 2.9%), and the thalamus (mean |AI| = 5.5%), correlating well with AI of early-phase Aβ-PET (mean R = 0.87, range 0.62-0.98). Visual assessments of hypoperfusion and hypometabolism were highly congruent. Conclusion: Early-phase Aβ-PET facilitates assessment of neuronal injury in CBS for cortical and subcortical areas. Known asymmetries in CBS are captured by this method, enabling assessment of Aβ-status and neuronal injury with a single radiation exposure at a single visit.
Collapse
Affiliation(s)
- Julia Schmitt
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Julia Sauerbeck
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Catharina Prix
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Endy Weidinger
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Olivia Wagemann
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Adrian Danek
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Department of Nuclear Medicine, University of Bern, Inselspital, Bern, Switzerland.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, United Kingdom
| | - Christian Haass
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Faculty of Medicine, Chair of Metabolic Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians-Universtität München, Munich, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Medizinische Hochschule Hannover, Hanover, Germany.,Department of Neurology, Technical University Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | |
Collapse
|
37
|
Albano D, Premi E, Peli A, Camoni L, Bertagna F, Turrone R, Borroni B, Calhoun VD, Rodella C, Magoni M, Padovani A, Giubbini R, Paghera B. Correlation between brain glucose metabolism (18F-FDG) and cerebral blood flow with amyloid tracers (18F-Florbetapir) in clinical routine: Preliminary evidences. Rev Esp Med Nucl Imagen Mol 2021; 41:146-152. [DOI: 10.1016/j.remnie.2021.03.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/07/2021] [Indexed: 10/21/2022]
|
38
|
Schwarz AJ. The Use, Standardization, and Interpretation of Brain Imaging Data in Clinical Trials of Neurodegenerative Disorders. Neurotherapeutics 2021; 18:686-708. [PMID: 33846962 PMCID: PMC8423963 DOI: 10.1007/s13311-021-01027-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Imaging biomarkers play a wide-ranging role in clinical trials for neurological disorders. This includes selecting the appropriate trial participants, establishing target engagement and mechanism-related pharmacodynamic effect, monitoring safety, and providing evidence of disease modification. In the early stages of clinical drug development, evidence of target engagement and/or downstream pharmacodynamic effect-especially with a clear relationship to dose-can provide confidence that the therapeutic candidate should be advanced to larger and more expensive trials, and can inform the selection of the dose(s) to be further tested, i.e., to "de-risk" the drug development program. In these later-phase trials, evidence that the therapeutic candidate is altering disease-related biomarkers can provide important evidence that the clinical benefit of the compound (if observed) is grounded in meaningful biological changes. The interpretation of disease-related imaging markers, and comparability across different trials and imaging tools, is greatly improved when standardized outcome measures are defined. This standardization should not impinge on scientific advances in the imaging tools per se but provides a common language in which the results generated by these tools are expressed. PET markers of pathological protein aggregates and structural imaging of brain atrophy are common disease-related elements across many neurological disorders. However, PET tracers for pathologies beyond amyloid β and tau are needed, and the interpretability of structural imaging can be enhanced by some simple considerations to guard against the possible confound of pseudo-atrophy. Learnings from much-studied conditions such as Alzheimer's disease and multiple sclerosis will be beneficial as the field embraces rarer diseases.
Collapse
Affiliation(s)
- Adam J Schwarz
- Takeda Pharmaceuticals Ltd., 40 Landsdowne Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
39
|
Heeman F, Yaqub M, Hendriks J, Bader I, Barkhof F, Gispert JD, van Berckel BNM, Lopes Alves I, Lammertsma AA. Parametric imaging of dual-time window [ 18F]flutemetamol and [ 18F]florbetaben studies. Neuroimage 2021; 234:117953. [PMID: 33762215 DOI: 10.1016/j.neuroimage.2021.117953] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/12/2021] [Accepted: 03/05/2021] [Indexed: 11/15/2022] Open
Abstract
Optimal pharmacokinetic models for quantifying amyloid beta (Aβ) burden using both [18F]flutemetamol and [18F]florbetaben scans have previously been identified at a region of interest (ROI) level. The purpose of this study was to determine optimal quantitative methods for parametric analyses of [18F]flutemetamol and [18F]florbetaben scans. Forty-six participants were scanned on a PET/MR scanner using a dual-time window protocol and either [18F]flutemetamol (N=24) or [18F]florbetaben (N=22). The following parametric approaches were used to derive DVR estimates: reference Logan (RLogan), receptor parametric mapping (RPM), two-step simplified reference tissue model (SRTM2) and multilinear reference tissue models (MRTM0, MRTM1, MRTM2), all with cerebellar grey matter as reference tissue. In addition, a standardized uptake value ratio (SUVR) was calculated for the 90-110 min post injection interval. All parametric images were assessed visually. Regional outcome measures were compared with those from a validated ROI method, i.e. DVR derived using RLogan. Visually, RPM, and SRTM2 performed best across tracers and, in addition to SUVR, provided highest AUC values for differentiating between Aβ-positive vs Aβ-negative scans ([18F]flutemetamol: range AUC=0.96-0.97 [18F]florbetaben: range AUC=0.83-0.85). Outcome parameters of most methods were highly correlated with the reference method (R2≥0.87), while lowest correlation were observed for MRTM2 (R2=0.71-0.80). Furthermore, bias was low (≤5%) and independent of underlying amyloid burden for MRTM0 and MRTM1. The optimal parametric method differed per evaluated aspect; however, the best compromise across aspects was found for MRTM0 followed by SRTM2, for both tracers. SRTM2 is the preferred method for parametric imaging because, in addition to its good performance, it has the advantage of providing a measure of relative perfusion (R1), which is useful for measuring disease progression.
Collapse
Affiliation(s)
- Fiona Heeman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands.
| | - Maqsood Yaqub
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Janine Hendriks
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Ilona Bader
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Frederik Barkhof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands; UCL, Institutes of Neurology and Healthcare Engineering, London, United Kingdom
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Centre, Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain; Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Bart N M van Berckel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Isadora Lopes Alves
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Adriaan A Lammertsma
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| |
Collapse
|
40
|
Bergeret S, Queneau M, Rodallec M, Curis E, Dumurgier J, Hugon J, Paquet C, Farid K, Baron JC. [ 18 F]FDG PET may differentiate cerebral amyloid angiopathy from Alzheimer's disease. Eur J Neurol 2021; 28:1511-1519. [PMID: 33460498 DOI: 10.1111/ene.14743] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is a frequent cause of both intracerebral hemorrhage (ICH) and cognitive impairment in the elderly. Diagnosis relies on the Boston criteria, which use magnetic resonance imaging markers including ≥2 exclusively lobar cerebral microbleeds (lCMBs). Although amyloid positron emission tomography (PET) may provide molecular diagnosis, its specificity relative to Alzheimer's disease (AD) is limited due to the prevalence of positive amyloid PET in cognitively normal elderly. Using early-phase 11 C-Pittsburgh compound B as surrogate for tissue perfusion, a significantly lower occipital/posterior cingulate (O/PC) tracer uptake ratio in probable CAA relative to AD was recently reported, consistent with histopathological lesion distribution. We tested whether this finding could be reproduced using [18 F]fluorodeoxyglucose (FDG)-PET, a widely available modality that correlates well with early-phase amyloid PET in both healthy subjects and AD. METHODS From a large memory clinic database, we retrospectively included 14 patients with probable CAA (Boston criteria) and 21 patients with no lCMB fulfilling AD criteria including cerebrospinal fluid biomarkers. In all, [18 F]FDG-PET/computed tomography (CT) was available as part of routine care. No subject had a clinical history of ICH. Regional standardized [18 F]FDG uptake values normalized to the pons (standard uptake value ratio [SUVr]) were obtained, and the O/PC ratio was calculated. RESULTS The SUVr O/PC ratio was significantly lower in CAA versus AD (1.02 ± 0.14 vs. 1.19 ± 0.18, respectively; p = 0.024). CONCLUSIONS Despite the small sample, our findings are consistent with the previous early-phase amyloid PET study. Thus, [18 F]FDG-PET may help differentiate CAA from AD, particularly in cases of amyloid PET positivity. Larger prospective studies, including in CAA-related ICH, are however warranted.
Collapse
Affiliation(s)
- Sébastien Bergeret
- Department of Nuclear Medicine, CHU French West Indies, Fort-de-France, France
| | - Mathieu Queneau
- Department of Nuclear Medicine, Centre Cardiologique du Nord, Saint-Denis, France
| | - Mathieu Rodallec
- Department of Radiology, Centre Cardiologique du Nord, Saint-Denis, France
| | - Emmanuel Curis
- Laboratoire de Biomathématiques, EA 7537 "BioSTM", Faculté de Pharmacie, Université de Paris, Paris, France.,Service de Biostatistiques et d'Information Médicale, Hôpital Saint-Louis, APHP, Paris, France
| | - Julien Dumurgier
- INSERM UMR-S 1144: Therapeutic Optimization in Neuropsychopharmacology, Université de Paris, Paris, France
| | - Jacques Hugon
- INSERM UMR-S 1144: Therapeutic Optimization in Neuropsychopharmacology, Université de Paris, Paris, France.,Cognitive Neurology Center, APHP, Saint-Louis Lariboisière Fernand-Widal Hospital Group, Paris, France
| | - Claire Paquet
- INSERM UMR-S 1144: Therapeutic Optimization in Neuropsychopharmacology, Université de Paris, Paris, France.,Cognitive Neurology Center, APHP, Saint-Louis Lariboisière Fernand-Widal Hospital Group, Paris, France
| | - Karim Farid
- Department of Nuclear Medicine, CHU French West Indies, Fort-de-France, France.,INSERM UMR-S 1144: Therapeutic Optimization in Neuropsychopharmacology, Université de Paris, Paris, France
| | - Jean-Claude Baron
- Department of Neurology, Sainte-Anne Hospital, Université de Paris, Paris, France.,INSERM U1266: Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| |
Collapse
|
41
|
Minoshima S, Mosci K, Cross D, Thientunyakit T. Brain [F-18]FDG PET for Clinical Dementia Workup: Differential Diagnosis of Alzheimer's Disease and Other Types of Dementing Disorders. Semin Nucl Med 2021; 51:230-240. [PMID: 33546814 DOI: 10.1053/j.semnuclmed.2021.01.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PET imaging with [F-18]FDG has been used extensively for research and clinical applications in dementia. In the brain, [F-18]FDG accumulates around synapses and represents local neuronal activity. Patterns of altered [F-18]FDG uptake reflecting local neuronal dysfunction provide differential diagnostic clues for various dementing disorders. Image interpretation can be accomplished by employing statistical brain mapping techniques. Various guidelines have been published to support the appropriate use of [F-18]FDG PET for clinical dementia workup. PET images with [F-18]FDG demonstrate distinct patterns of decreased uptake for Alzheimer's disease (AD), Dementia with Lewy bodies (DLB), and frontotemporal dementia (FTD) as well as its multiple subtypes such as behavioral variant FTD, primary progressive aphasia (PPA), progressive supranuclear palsy, and corticobasal degeneration to aid in the differential diagnoses. Mixed dementia, not only AD + Vascular Dementia, but also AD + other neurodegenerative disorders, should also be considered when interpreting [F-18]FDG PET images. Brain PET imaging with [F-18]FDG remains a valuable component of dementia workup owing to its relatively low cost, differential diagnostic performance, widespread availability, and physicians' experience over more than 40 years since the initial development.
Collapse
Affiliation(s)
- Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT.
| | - Karina Mosci
- Hospital das Forças Armadas (HFA) and Hospital Santa Lucia, Brasilia, Brazil
| | - Donna Cross
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT
| | - Tanyaluck Thientunyakit
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Siriraj Hospital, Bangkok, Thailand
| |
Collapse
|
42
|
Kreuzer A, Sauerbeck J, Scheifele M, Stockbauer A, Schönecker S, Prix C, Wlasich E, Loosli SV, M Kazmierczak P, Unterrainer M, Catak C, Janowitz D, Pogarell O, Palleis C, Perneczky R, Albert NL, Bartenstein P, Danek A, Buerger K, Levin J, Zwergal A, Rominger A, Brendel M, Beyer L. Detection Gap of Right-Asymmetric Neuronal Degeneration by CERAD Test Battery in Alzheimer's Disease. Front Aging Neurosci 2021; 13:611595. [PMID: 33603657 PMCID: PMC7884314 DOI: 10.3389/fnagi.2021.611595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/04/2021] [Indexed: 01/18/2023] Open
Abstract
Objectives: Asymmetric disease characteristics on neuroimaging are common in structural and functional imaging of neurodegenerative diseases, particularly in Alzheimer‘s disease (AD). However, a standardized clinical evaluation of asymmetric neuronal degeneration and its impact on clinical findings has only sporadically been investigated for F-18-fluorodeoxyglucose positron emission tomography (F-18-FDG-PET). This study aimed to evaluate the impact of lateralized neuronal degeneration on the detection of AD by detailed clinical testing. Furthermore, we compared associations between clinical evaluation and lateralized neuronal degeneration between FDG-PET hypometabolism and hippocampal atrophy. Finally, we investigated if specific subtests show associations with lateralized neuronal degeneration. Methods: One-hundred and forty-six patients with a clinical diagnosis of AD (age 71 ± 8) were investigated by FDG-PET and the “Consortium to Establish a Registry for Alzheimer’s disease” (CERAD) test battery. For assessment of neuronal degeneration, FDG-PET hypometabolism in brain regions typically affected in AD were graded by visual (3D-surface projections) and semiquantitative analysis. Asymmetry of the hippocampus (left-right) in magnetic resonance tomography (MRI) was rated visually by the Scheltens scale. Measures of asymmetry were calculated to quantify lateralized neuronal degeneration and asymmetry scores were subsequently correlated with CERAD. Results: Asymmetry with left-dominant neuronal degeneration to FDG-PET was an independent predictor of cognitive impairment (visual: β = −0.288, p < 0.001; semiquantitative: β = −0.451, p < 0.001) when controlled for age, gender, years of education and total burden of neuronal degeneration, whereas hippocampal asymmetry to MRI was not (β = −0.034; p = 0.731). Direct comparison of CERAD-PET associations in cases with right- and left-lateralized neuronal degeneration estimated a detection gap of 2.7 years for right-lateralized cases. Left-hemispheric neuronal degeneration was significantly associated with the total CERAD score and multiple subscores, whereas only MMSE (semiquantitative: β = 0.429, p < 0.001) and constructional praxis (semiquantitative: β = 0.292, p = 0.008) showed significant associations with right-hemispheric neuronal degeneration. Conclusions: Asymmetry of deteriorated cerebral glucose metabolism has a significant impact on the coupling between neuronal degeneration and cognitive function. Right dominant neuronal degeneration shows a delayed detection by global CERAD testing and requires evaluation of specific subdomains of cognitive testing.
Collapse
Affiliation(s)
- Annika Kreuzer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Julia Sauerbeck
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Anna Stockbauer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sonja Schönecker
- Department of Neurology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Catharina Prix
- Department of Neurology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Elisabeth Wlasich
- Department of Neurology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Sandra V Loosli
- Department of Neurology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Philipp M Kazmierczak
- Department of Radiology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Radiology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Cihan Catak
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Oliver Pogarell
- Department of Psychiatry, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital, Ludwig-Maximilians-University, Munich, Germany.,DZNE-German Center for Neurodegenerative Diseases, Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry, University Hospital, Ludwig-Maximilians-University, Munich, Germany.,DZNE-German Center for Neurodegenerative Diseases, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College, London, United Kingdom
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Adrian Danek
- Department of Neurology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University, Munich, Germany.,DZNE-German Center for Neurodegenerative Diseases, Munich, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital, Ludwig-Maximilians-University, Munich, Germany.,DZNE-German Center for Neurodegenerative Diseases, Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Nuclear Medicine Inselspital, University of Bern, Bern, Switzerland
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
43
|
Koh S, Lee SM, Moon SY. Orbitofrontal Hemorrhage and Mild Cognitive Impairment Associated with Othello Syndrome. J Clin Neurol 2021; 17:142-144. [PMID: 33480213 PMCID: PMC7840332 DOI: 10.3988/jcn.2021.17.1.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/10/2020] [Accepted: 10/15/2020] [Indexed: 11/22/2022] Open
Affiliation(s)
- Seungyon Koh
- Department of Neurology, Ajou University School of Medicine, Suwon, Korea.,Department of Medical Sciences, Graduate School of Ajou University, Suwon, Korea
| | - Sun Min Lee
- Department of Neurology, Ajou University School of Medicine, Suwon, Korea.,Department of Medical Sciences, Graduate School of Ajou University, Suwon, Korea
| | - So Young Moon
- Department of Neurology, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
44
|
Abstract
Pathological accumulated misfolded tau underlies various neurodegenerative diseases and associated clinical syndromes. To diagnose those diseases reliable before death or even at early stages, many different tau-specific radiotracers have been developed in the last decade to be used with positron-emission-tomography. In contrast to amyloid-β imaging, different isoforms of tau exist further complicating radiotracer development. First-generation radiotracers like [11C]PBB3, [18F]AV1451 and [18F]THK5351 have been extensively investigated in vitro and in vivo. In Alzheimer's disease (AD), high specific binding could be detected, and evidence of clinical applicability recently led to clinical approval of [18F]flortaucipir ([18F]AV1451) by the FDA. Nevertheless, absent or minor binding to non-AD tau isoforms and high off-target binding to non-tau brain structures limit the diagnostic applicability especially in non-AD tauopathies demanding further tracer development. In vitro assays and autoradiography results of next-generation radiotracers [18F]MK-6240, [18F]RO-948, [18F]PM-PBB3, [18F]GTP-1 and [18F]PI-2620 clearly indicate less off-target binding and high specific binding to tau neurofibrils. First in human studies have been conducted with promising results for all tracers in AD patients, and also some positive experience in non-AD tauopathies. Overall, larger scaled autoradiography and human studies are needed to further evaluate the most promising candidates and support future clinical approval.
Collapse
Affiliation(s)
- Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
45
|
Cognitive reserve hypothesis in frontotemporal dementia: A FDG-PET study. NEUROIMAGE-CLINICAL 2020; 29:102535. [PMID: 33369564 PMCID: PMC7773557 DOI: 10.1016/j.nicl.2020.102535] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/17/2020] [Accepted: 12/12/2020] [Indexed: 11/21/2022]
Abstract
The cognitive reserve hypothesis is also applicable for FTD. The educational level predicts left-temporal hypometabolism in FTD. Residualized cognitive performance is positively correlated with education. Residualized cognitive performance is negatively correlated with hypometabolism.
Background and objective Reserve is defined as the ability to maintain cognitive functions relatively well at a given level of pathology. Early life experiences such as education are associated with lower dementia risk in general. However, whether more years of education guards against the impact of brain alterations also in frontotemporal dementia (FTD) has not been shown in a large patient collective. Therefore, we assessed whether education is associated with relatively high cognitive performance despite the presence of [18F]-fluorodeoxyglucose positron-emission-tomography (FDG-PET) hypometabolism in FTD. Methods Sixty-six FTD subjects (age 67 ± 8 years) and twenty-four cognitively healthy controls (HC) were evaluated. Brain regions with FTD-related glucose hypometabolism in the contrast against HC and brain regions that correlate with the cognitive function were defined by a voxel-based analysis and individual FDG-PET values were extracted from all frontotemporal brain areas. Linear regression analysis served to test if education is associated with residualized cognitive performance and regional FDG-PET hypometabolism after controlling for global cognition. Results Compared to healthy controls, patients with FTD showed glucose hypometabolism in bilateral frontal and temporal brain areas whereas cognition was only associated with deteriorated glucose metabolism in the left temporal lobe. The education level was significantly correlated with the residualized cognitive performance (residuals from regression analysis between hypometabolism and cognitive function as a quantitative index of reserve) and also negatively correlated with left temporal FDG-PET hypometabolism after controlling for cognition. Conclusions In patients with FTD, the education level predicts the existing left temporal FDG-PET hypometabolism at the same cognition level, supporting the cognitive reserve hypothesis in FTD.
Collapse
|
46
|
Bae S, Choi H, Whi W, Paeng JC, Cheon GJ, Kang KW, Lee DS. Spatial Normalization Using Early-Phase [ 18F]FP-CIT PET for Quantification of Striatal Dopamine Transporter Binding. Nucl Med Mol Imaging 2020; 54:305-314. [PMID: 33282002 DOI: 10.1007/s13139-020-00669-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose The precise quantification of dopamine transporter (DAT) density on N-(3-[18F]Fluoropropyl)-2β-carbomethoxy-3β-(4-iodophenyl) nortropane positron emission tomography ([18F]FP-CIT PET) imaging is crucial to measure the degree of striatal DAT loss in patients with parkinsonism. The quantitative analysis requires a spatial normalization process based on a template brain. Since the spatial normalization method based on a delayed-phase PET has limited performance, we suggest an early-phase PET-based method and compared its accuracy, referring to the MRI-based approach as a gold standard. Methods A total of 39 referred patients from the movement disorder clinic who underwent dual-phase [18F]FP-CIT PET and took MRI within 1 year were retrospectively analyzed. The three spatial normalization methods were applied for quantification of [18F]FP-CIT PET-MRI-based anatomical normalization, PET template-based method based on delayed PET, and that based on early PET. The striatal binding ratios (BRs) were compared, and voxelwise paired t tests were implemented between different methods. Results The early image-based normalization showed concordant patterns of putaminal [18F]FP-CIT binding with an MRI-based method. The BRs of the putamen from the MRI-based approach showed higher agreement with early image- than delayed image-based method as presented by Bland-Altman plots and intraclass correlation coefficients (early image-based, 0.980; delayed image-based, 0.895). The voxelwise test exhibited a smaller volume of significantly different counts in putamen between brains processed by early image and MRI compared to that between delayed image and MRI. Conclusion The early-phase [18F]FP-CIT PET can be utilized for spatial normalization of delayed PET image when the MRI image is unavailable and presents better performance than the delayed template-based method in quantitation of putaminal binding ratio.
Collapse
Affiliation(s)
- Sungwoo Bae
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea.,Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wonseok Whi
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Jin Chul Paeng
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea.,Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea.,Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea.,Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.,Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
47
|
Multivariate analysis of dual-point amyloid PET intended to assist the diagnosis of Alzheimer’s disease. Neurocomputing 2020. [DOI: 10.1016/j.neucom.2020.06.081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
48
|
Kerstens VS, Varrone A. Dopamine transporter imaging in neurodegenerative movement disorders: PET vs. SPECT. Clin Transl Imaging 2020. [DOI: 10.1007/s40336-020-00386-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
Purpose
The dopamine transporter (DAT) serves as biomarker for parkinsonian syndromes. DAT can be measured in vivo with single-photon emission computed tomography (SPECT) and positron emission tomography (PET). DAT-SPECT is the current clinical molecular imaging standard. However, PET has advantages over SPECT measurements, and PET radioligands with the necessary properties for clinical applications are on the rise. Therefore, it is time to review the role of DAT imaging with SPECT compared to PET.
Methods
PubMed and Web of Science were searched for relevant literature of the previous 10 years. Four topics for comparison were used: diagnostic accuracy, quantitative accuracy, logistics, and flexibility.
Results
There are a few studies directly comparing DAT-PET and DAT-SPECT. PET and SPECT both perform well in discriminating neurodegenerative from non-neurodegenerative parkinsonism. Clinical DAT-PET imaging seems feasible only recently, thanks to simplified DAT assessments and better availability of PET radioligands and systems. The higher resolution of PET makes more comprehensive assessments of disease progression in the basal ganglia possible. Additionally, it has the possibility of multimodal target assessment.
Conclusion
DAT-SPECT is established for differentiating degenerative from non-degenerative parkinsonism. For further differentiation within neurodegenerative Parkinsonian syndromes, DAT-PET has essential benefits. Nowadays, because of wider availability of PET systems and radioligand production centers, and the possibility to use simplified quantification methods, DAT-PET imaging is feasible for clinical use. Therefore, DAT-PET needs to be considered for a more active role in the clinic to take a step forward to a more comprehensive understanding and assessment of Parkinson’s disease.
Collapse
|
49
|
Yeo SK, Shepelytskyi Y, Grynko V, Albert MS. Molecular Imaging of Fluorinated Probes for Tau Protein and Amyloid-β Detection. Molecules 2020; 25:molecules25153413. [PMID: 32731418 PMCID: PMC7435578 DOI: 10.3390/molecules25153413] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia and results in progressive neurodegeneration. The incidence rate of AD is increasing, creating a major public health issue. AD is characterized by neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein and senile plaques composed of amyloid-β (Aβ). Currently, a definitive diagnosis of AD is accomplished post-mortem. Thus, the use of molecular probes that are able to selectively bind to NFTs or Aβ can be valuable tools for the accurate and early diagnosis of AD. The aim of this review is to summarize and highlight fluorinated molecular probes that can be used for molecular imaging to detect either NFTs or Aβ. Specifically, fluorinated molecular probes used in conjunction with 19F MRI, PET, and fluorescence imaging will be explored.
Collapse
Affiliation(s)
- Sarah K. Yeo
- Biology Department, Lakehead University, Thunder Bay, ON P7B 5E1, Canada;
| | - Yurii Shepelytskyi
- Chemistry and Materials Science Program, Lakehead University, Thunder Bay, ON P7B 5E1, Canada; (Y.S.); (V.G.)
- Thunder Bay Regional Health Research Institute, Thunder Bay, ON P7B 6V4, Canada
| | - Vira Grynko
- Chemistry and Materials Science Program, Lakehead University, Thunder Bay, ON P7B 5E1, Canada; (Y.S.); (V.G.)
- Thunder Bay Regional Health Research Institute, Thunder Bay, ON P7B 6V4, Canada
| | - Mitchell S. Albert
- Thunder Bay Regional Health Research Institute, Thunder Bay, ON P7B 6V4, Canada
- Chemistry Department, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
- Northern Ontario School of Medicine, Thunder Bay, ON P7B 5E1, Canada
- Correspondence: ; Tel.: +1-807-355-9191
| |
Collapse
|
50
|
Um YH, Wang SM, Kim NY, Kang DW, Na HR, Lee CU, Lim HK. Effects of Moderate Intensity Exercise on the Cortical Thickness and Subcortical Volumes of Preclinical Alzheimer's Disease Patients: A Pilot Study. Psychiatry Investig 2020; 17:613-619. [PMID: 32570297 PMCID: PMC7324741 DOI: 10.30773/pi.2020.0214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE We aimed to explore the impact of moderate intensity exercise on the cortical thickness and subcortical volumes of preclinical Alzheimer's disease (AD) patients. METHODS Sixty-three preclinical AD patients with magnetic resonance imaging (MRI) and 18-florbetaben positron emission tomography (PET) data were enrolled in the study. Information on demographic characteristics, cognitive battery scores, self-reported exercise habits were attained. Structural magnetic resonance images were analyzed and processed using Freesurfer v6.0. RESULTS Compared to Exercise group, Non-Exercise group demonstrated reduced cortical thickness in left parstriangularis, rostral middle frontal, entorhinal, superior frontal, lingual, superior parietal, lateral occipital, inferior parietal gyrus, temporal pole, precuneus, insula, fusiform gyrus, right precuneus, superiorparietal, lateral orbitofrontal, rostral middle frontal, medial orbitofrontal, superior frontal, lingual, middle temporal gyrus, insula, supramarginal, parahippocampal, paracentral gyrus. Volumes of right thalamus, caudate, putamen, pallidum, hippocampus, amygdala were also reduced in Non-Exercise group. CONCLUSION Moderate intensity exercise affects cortical and subcortical structures in preclinical AD patients. Thus, physical exercise has a potential to be an effective intervention to prevent future cognitive decline in those at high risk of AD.
Collapse
Affiliation(s)
- Yoo Hyun Um
- Department of Psychiatry, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Republic of Korea
| | - Sheng-Min Wang
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Nak-Young Kim
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Woo Kang
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hae-Ran Na
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chang Uk Lee
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|