1
|
Olfati N, Akhoundi FH, Litvan I. Atypical Parkinsonian Disorders. Neurol Clin 2025; 43:249-277. [PMID: 40185521 DOI: 10.1016/j.ncl.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
Atypical parkinsonian disorders (APD) include progressive supranuclear palsy, corticobasal degeneration, and multiple system atrophy. Identifying APD is important because they have different pathogenesis, disease course, and prognosis than Parkinson's disease (PD), and require different treatments. Therefore, when encountering patients with parkinsonism, it is of crucial importance to look for "red flags" or signs, such as impairments in higher cortical function, visuomotor system, cerebellar and other motor abnormalities including dystonia, myoclonus, and apraxia that help differentiate them from PD. Although disease-modifying therapies are not yet available, treatments targeting specific symptoms may improve the quality of life in these patients.
Collapse
Affiliation(s)
- Nahid Olfati
- Department of Neurosciences, Parkinson and Other Movement Disorders Center, University of California San Diego, La Jolla, CA, USA
| | - Fahimeh H Akhoundi
- Department of Neurosciences, Parkinson and Other Movement Disorders Center, University of California San Diego, La Jolla, CA, USA
| | - Irene Litvan
- Department of Neurosciences, Parkinson and Other Movement Disorders Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
2
|
Angehrn FN, Duan P, Zhang JY, Hong M. Binding Sites of a PET Ligand in Tau Fibrils with the Alzheimer's Disease Fold from 19F and 13C Solid-State NMR. Biochemistry 2025; 64:1624-1635. [PMID: 40068133 DOI: 10.1021/acs.biochem.5c00016] [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: 03/21/2025]
Abstract
Aggregation of the tau protein into cross-β amyloid fibrils is a hallmark of Alzheimer's disease (AD) and many other neurodegenerative disorders. Developing small molecules that bind these tau fibrils is important for the diagnosis and treatment of tauopathies. Here, we report the binding sites of a positron emission tomography (PET) ligand, PI-2620, to a recombinant tau construct that adopts the C-shaped AD fold. Using solid-state NMR 13C-19F rotational-echo double-resonance (REDOR) experiments, we measured the proximity of protein residues to the fluorine atom of the ligand. These data indicate that PI-2620 binds at two main locations in the concave interior of the C-shaped structure. Molecular docking simulations constrained by these REDOR data identified five binding poses at these two locations. In addition, 2D 13C-13C correlation NMR spectra indicate that PI-2620 decreased the intensities of residues at the protofilament interfaces, indicating that the ligand disordered the filament packing. Quantitative analysis of the 19F NMR spectra indicates that PI-2620 binds these AD-fold tau fibrils with a stoichiometry of ∼20 mol %, in which 10 mol % are immobilized and the rest are mobile. These results provide experimental constraints to the interaction of this second-generation PET tracer with tau fibrils adopting the AD fold and should be useful for the development of future imaging agents with improved stoichiometry and specificity for AD tau.
Collapse
Affiliation(s)
- Frida N Angehrn
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, Massachusetts 02139, United States
| | - Pu Duan
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, Massachusetts 02139, United States
| | - Jia Yi Zhang
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, Massachusetts 02139, United States
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, 170 Albany Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Lindberg A, Tong J, Zheng C, Mueller A, Kroth H, Stephens A, Mathis CA, Vasdev N. Radiosynthesis, In Vitro Characterization, and In Vivo PET Neuroimaging of [ 18F]F-4 for Tau Protein: A First-in-Human PET Study. ACS Chem Neurosci 2025; 16:1182-1189. [PMID: 40063537 PMCID: PMC11926865 DOI: 10.1021/acschemneuro.4c00866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
[18F]PI-2620 is a promising radiopharmaceutical for positron emission tomography (PET) imaging of both Alzheimer's disease (AD) and non-Alzheimer's disease (non-AD) tauopathies in humans. An array of fluorinated derivatives of the carbazole scaffold of PI-2620 were synthesized and evaluated. In vitro binding assays with [3H]PI-2620 in human tissues with AD, progressive supranuclear palsy, and corticobasal degeneration, combined with in silico predictions of blood-brain barrier permeability, led to the selection and radiosynthesis of [18F]F-4 as a promising radiotracer. In vivo PET imaging with [18F]F-4 in healthy rats showed brain uptake and kinetics suitable for neuroimaging, similar to those of [18F]PI-2620. A first-in-human PET imaging study in a healthy subject as well as a patient with AD, in comparison with [18F]PI-2620 in the same AD subject, confirmed that [18F]F-4 is an alternative radiopharmaceutical for imaging tau protein.
Collapse
Affiliation(s)
- Anton Lindberg
- Azrieli Centre
for Neuro-Radiochemistry, Brain Health Imaging Centre, CAMH, Toronto M5T 1R8, Canada
| | - Junchao Tong
- Azrieli Centre
for Neuro-Radiochemistry, Brain Health Imaging Centre, CAMH, Toronto M5T 1R8, Canada
| | - Chao Zheng
- Azrieli Centre
for Neuro-Radiochemistry, Brain Health Imaging Centre, CAMH, Toronto M5T 1R8, Canada
- Department
of Psychiatry, University of Toronto, Toronto M5T 1R8, Canada
- Departments
of Pharmacology and Toxicology and Chemistry, University of Toronto, Toronto M5S 1A1, Canada
| | | | | | | | - Chester A. Mathis
- Department
of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Neil Vasdev
- Azrieli Centre
for Neuro-Radiochemistry, Brain Health Imaging Centre, CAMH, Toronto M5T 1R8, Canada
- Department
of Psychiatry, University of Toronto, Toronto M5T 1R8, Canada
| |
Collapse
|
4
|
Kling A, Kusche-Palenga J, Palleis C, Jäck A, Bernhardt AM, Frontzkowski L, Roemer SN, Slemann L, Zaganjori M, Scheifele M, Paeger L, Bischof GN, van Eimeren T, Drzezga A, Sabri O, Rullmann M, Barthel H, Levin J, Herms J, Franzmeier N, Höglinger G, Roeber S, Brendel M, Gnörich J. Exploring the origins of frequent tau-PET signal in vermal and adjacent regions. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07199-x. [PMID: 40100387 DOI: 10.1007/s00259-025-07199-x] [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: 10/30/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Off-target binding remains a significant challenge in tau-PET neuroimaging. While off-targets including monoamine oxidase enzymes and neuromelanin-containing cells have been identified, recent studies indicated a relevant binding of novel tau tracers to melanin-containing structures. To date, little is known about the effect of melanocytes in the meninges on tracer signals in brain PET data. Thus, we aimed to identify the target structure causal for the frequently observed [18F]PI-2620 PET signal in the vermis and adjacent cerebellar regions. METHODS 274 participants underwent dynamic [18F]PI-2620 tau-PET: 3/4R-tauopathies (n = 85), 4R-tauopathies (n = 147), tau-negative disease controls (n = 24), and healthy controls (n = 18). Standardized uptake value ratio (SUVR) and kinetic parameters including the distribution volume ratio (DVR), tracer clearance (k2) and relative perfusion (R1), were compared among the cohorts and sexes using the Automated Anatomical Labelling (AAL) atlas. Age and p-Tau levels in cerebrospinal fluid (CSF) were assessed for their relationship with vermal tau-PET signal. Furthermore, we combined autoradiographic and histochemical experiments on post-mortem brain tissue of deceased patients (n = 9). RESULTS Male participants revealed higher mean vermal [18F]PI-2620 DVR (0.95 ± 0.13; vs. females 0.88 ± 0.10, p < 0.0001). Sex-related differences were most pronounced in the 3/4R-tauopathy cohort (p < 0.0001). Mean SUVRVer/Cbl, k2 and correlation analyses of kinetic parameters did not differ among groups. Histological assessments revealed co-localization of leptomeningeal pigmented cells with strong autoradiography signal spots within the vermal fissures. Tau-related autoradiography signals, age or p-Tau levels did not correlate significantly with tau-PET signals. Iron deposits did not cause relevant autoradiography signals in the vermis. CONCLUSION Leptomeningeal melanocytes are the primary target structure for [18F]PI-2620 PET binding in anterior vermis, whereas iron and tau deposits do not contribute significantly.
Collapse
Affiliation(s)
- Agnes Kling
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Julia Kusche-Palenga
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Alexander Jäck
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Alexander M Bernhardt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Lukas Frontzkowski
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian N Roemer
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Luna Slemann
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Lars Paeger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Gérard N Bischof
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Research Center Jülich, Jülich, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Research Center Jülich, Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Research Center Jülich, Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Michael Rullmann
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Center of Neuropathology and Prion Research, University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, The Sahlgrenska Academy, Mölndal and Gothenburg, Sweden
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sigrun Roeber
- Center of Neuropathology and Prion Research, University of Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
| |
Collapse
|
5
|
Rullmann M, Henssen D, Melasch JT, Scherlach C, Saur D, Schroeter ML, Tiepolt S, Koglin N, Stephens AW, Hesse S, Strauss M, Brendel M, Mishchenko O, Schildan A, Classen J, Hoffmann KT, Sabri O, Barthel H. Multi-parametric [ 18F]PI-2620 tau PET/MRI for the phenotyping of different Alzheimer's disease variants. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07135-z. [PMID: 39937274 DOI: 10.1007/s00259-025-07135-z] [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: 01/10/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE Heterogeneity in clinical phenotypes has led to the description of different phenotypes of Alzheimer's disease (AD). Besides the most frequent amnestic variant of AD (aAD), patients presenting with language deficits are diagnosed with logopenic variant primary progressive aphasia (lvPPA), whereas patients presenting with visual deficits are classified as posterior cortical atrophy (PCA). METHODS This study set out to investigate the value of a multi-parametric [18F]PI-2620 tau PET/MRI protocol to distinguish aAD, lvPPA and PCA to support clinical diagnosis in 32 patients. Phenotype-specific information about tau accumulation, relative perfusion, grey matter density, functional network alterations and white matter microstructural alterations was collected. RESULTS The aAD patients showed significantly higher tau accumulation, relative hypoperfusion and grey matter density loss in the temporal lobes compared to PCA and lvPPA patients. PCA patients, on the other hand, showed significantly higher tau accumulation in the occipital lobe as compared to aAD patients. Relative hypoperfusion in the occipital lobe and loss of functional connectivity of the posterior cingulate cortex to supplementary visual cortical regions helped to distinguish PCA from lvPPA. Tau accumulation in the cerebellum and microstructural changes in the cingulum were found to help differentiate lvPPA from aAD. CONCLUSION This study highlights structural and functional differences between patients with different AD phenotypes. Differences in regional tau PET signals suggest that refinements in the Braak staging system are needed for the non-aAD cases. These patterns of tau accumulation align with the cascading network failure hypothesis, though more research is needed to warrant the here presented results in larger patient cohorts.
Collapse
Affiliation(s)
- Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany.
| | - Dylan Henssen
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Juliana T Melasch
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Cordula Scherlach
- Department of Neuroradiology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Dorothee Saur
- Department of Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Matthias L Schroeter
- Clinic for Cognitive Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | | | | | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Maria Strauss
- Department of Psychiatry, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Olena Mishchenko
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Andreas Schildan
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Joseph Classen
- Department of Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Karl-Titus Hoffmann
- Department of Neuroradiology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Department of Nuclear Medicine, Hospital Dessau, Dessau, Germany
| |
Collapse
|
6
|
Spiegel C, Marotta C, Bertram K, Vivash L, Harding IH. Brainstem and cerebellar radiological findings in progressive supranuclear palsy. Brain Commun 2025; 7:fcaf051. [PMID: 39958262 PMCID: PMC11829206 DOI: 10.1093/braincomms/fcaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/02/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025] Open
Abstract
Progressive supranuclear palsy is a sporadic neurodegenerative 4-repeat tauopathy associated with significant morbidity. Heterogeneity of symptom expression among this group is increasingly recognized, reflecting variable tau spread and neurodegeneration. Clinical manifestations consist of debilitating and rapidly progressive motor, oculomotor, speech, cognitive and affective impairments. Core pathological changes are noted with a predominance in the midbrain and basal ganglia; however, spread to the more caudal brainstem and cerebellar regions is reported at various stages. Accordingly, whilst midbrain atrophy is the best recognized supportive imaging finding, quantitative neuroimaging studies using MRI and PET approaches have revealed a wider profile of brain abnormalities in cohorts of individuals with progressive supranuclear palsy. This expanded neurobiological scope of disease may account for individual heterogeneity and may highlight additional biological markers that are relevant to diagnosing and tracking the illness. Additionally, there is increasing understanding of the diverse cognitive, affective and speech functions of the cerebellum, which may be implicated in progressive supranuclear palsy beyond current recognition. In this review, we undertake a systematic literature search and summary of in vivo structural and functional neuroimaging findings in the brainstem and cerebellum in progressive supranuclear palsy to date. Novel and multimodal imaging techniques have emerged over recent years, which reveal several infratentorial alterations beyond midbrain atrophy in progressive supranuclear palsy. Most saliently, there is evidence for volume loss and microstructural damage in the pons, middle cerebellar peduncles and cerebellar cortex and deep nuclei, reported alongside recognized midbrain and superior cerebellar peduncle changes. Whilst the literature supporting the presence of these features is not unanimous, the evidence base is compelling, including correlations with disease progression, severity or variant differences. A smaller number of studies report on abnormalities in MRI measures of iron deposition, neuromelanin, viscoelasticity and the glymphatic system involving the infratentorial regions. Molecular imaging studies have also shown increased uptake of tau tracer in the midbrain and cerebellar dentate nucleus, although concern remains regarding possible off-target binding. Imaging of other molecular targets has been sparse, but reports of neurotransmitter, inflammatory and synaptic density alterations in cerebellar and brainstem regions are available. Taken together, there is an established evidence base of in vivo imaging alterations in the brainstem and cerebellum which highlights that midbrain atrophy is often accompanied by other infratentorial alterations in people with progressive supranuclear palsy. Further research examining the contribution of these features to clinical morbidity and inter-individual variability in symptom expression is warranted.
Collapse
Affiliation(s)
- Chloe Spiegel
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne 3004, Australia
- Department of Neurology, Alfred Health, Melbourne 3004, Australia
| | - Cassandra Marotta
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne 3004, Australia
| | - Kelly Bertram
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne 3004, Australia
- Department of Neurology, Alfred Health, Melbourne 3004, Australia
| | - Lucy Vivash
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne 3004, Australia
| | - Ian H Harding
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne 3004, Australia
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| |
Collapse
|
7
|
Yamamoto Y, Takahata K, Seki M, Okusa S, Tatebe H, Ueda R, Endo H, Tagai K, Moriguchi S, Kurose S, Ichihashi M, Matsuura S, Kawamura K, Zhang MR, Ueno Y, Takiyama Y, Tokuda T, Higuchi M, Ito D. SLC9A6-Linked Parkinson Syndrome in Female Heterozygotes Is Associated With PET-Detectable Tau Pathology. Neurol Genet 2025; 11:e200235. [PMID: 39810750 PMCID: PMC11731372 DOI: 10.1212/nxg.0000000000200235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025]
Abstract
Background and Objectives A previous postmortem study of men with Christianson syndrome, a disorder caused by loss-of-function mutations in the gene SLC9A6, reported a mechanistic link between pathologic tau accumulation and progressive symptoms such as cerebellar atrophy and cognitive decline. This study aimed to characterize the relationships between neuropathologic manifestations and tau accumulation in heterozygous women with SLC9A6 mutation. Methods We conducted a multimodal neuroimaging and plasma biomarker study on 3 middle-aged heterozygous women with SLC9A6 mutations (proband 1: mid-50s; proband 2: early 50s; proband 3: mid-40s) presenting with progressive extrapyramidal symptoms. Examinations included 11C-PiB PET; 18F-florzolotau PET; structural MRI; and plasma measures of neurofilament light chain (NfL) polypeptide, glial fibrillary acidic protein, phosphorylated (p)Tau181, Aβ40, and Aβ42. Neuroimaging results of all 3 patients were compared with those of 12 healthy age-matched women (49.8 ± 4.7 years) while plasma biomarker levels of probands 1 and 2 were compared with those of 14 age-matched healthy women (54.1 ± 9.0 years). Results Proband 1 was diagnosed with Parkinson disease while probands 2 and 3 were diagnosed with atypical parkinsonism. 11C-PiB PET results were negative in all patients. 18F-florzolotau PET revealed focal tau accumulations in all 3 patients, predominantly in the striatum contralateral to motor symptoms. Moreover, greater extrapyramidal symptom severity was associated with higher standardized uptake value ratios (SUVRs) for 18F-florzolotau in the striatum. Multiple comparisons showed significantly higher 18F-florzolotau SUVR values in both the caudate and putamen of proband 1, who exhibited the most severe extrapyramidal signs, while no significant increases in 18F-florzolotau SUVR values were detected in any brain region of probands 2 and 3. Structural MRI revealed slightly lower regional subcortical and gray matter volumes in all patients but not significant after multiple comparisons. Finally, plasma NfL concentration was significantly higher in probands 1 and 2 compared with healthy controls. Discussion Our 18F-florzolotau PET analysis revealed greater tau accumulation in the striatum of heterozygous women with SLC9A6 mutation associated with worsening extrapyramidal symptom severity. The heterozygosity of loss-of-function SLC9A6 mutations further suggests that tauopathy may be a primary contributor to extrapyramidal signs.
Collapse
Affiliation(s)
- Yasuharu Yamamoto
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
- Department of Neuropsychiatry, Keio University School of Medicine
| | - Keisuke Takahata
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Morinobu Seki
- Department of Neurology, Keio University School of Medicine
| | - Shohei Okusa
- Department of Neurology, Keio University School of Medicine
| | - Harutsugu Tatebe
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Ryo Ueda
- Office of Radiation Technology, Keio University Hospital, Tokyo
| | - Hironobu Endo
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Kenji Tagai
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Sho Moriguchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Shin Kurose
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Masanori Ichihashi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Sayo Matsuura
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Kazunori Kawamura
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Yuji Ueno
- Department of Neurology, Faculty of Medicine, University of Yamanashi
| | - Yoshihisa Takiyama
- Department of Neurology, Faculty of Medicine, University of Yamanashi
- Fuefuki Central Hospital, Isawa-cho, Yamanashi; and
| | - Takahiko Tokuda
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba
| | - Daisuke Ito
- Memory Center, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Bauer T, Brendel M, Zaganjori M, Bernhardt AM, Jäck A, Stöcklein S, Scheifele M, Levin J, van Eimeren T, Drzezga A, Sabri O, Barthel H, Perneczky R, Höglinger G, Franzmeier N, Gnörich J. Pragmatic algorithm for visual assessment of 4-Repeat tauopathies in [ 18F]PI-2620 PET Scans. Neuroimage 2025; 306:121001. [PMID: 39798829 DOI: 10.1016/j.neuroimage.2025.121001] [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: 07/20/2024] [Revised: 11/11/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
AIM Standardized evaluation of [18F]PI-2620 tau-PET scans in 4R-tauopathies represents an unmet need in clinical practice. This study aims to investigate the effectiveness of visual evaluation of [18F]PI-2620 images for diagnosing 4R-tauopathies and to develop a straight-forward reading algorithm to improve objectivity and data reproducibility. METHODS A total of 83 individuals with [18F]PI-2620 PET scans were included. Participants were classified as probable 4R-tauopathies (n = 29), Alzheimer's disease (AD) (n = 20), α-synucleinopathies (n = 15), and healthy controls (n = 19) based on clinical criteria. Visual assessment of tau-PET scans (choice: 4R-tauopathy, AD-tauopathy, no-tauopathy) was conducted using either 20-40-minute or 40-60-minute intervals, with raw (common) and cerebellar grey matter scaled standardized reading settings (intensity-scaled). Two readers evaluated scans independently and blinded, with a third reader providing consensus in case of discrepant primary evaluation. A regional analysis was performed using the cortex, basal ganglia, midbrain, and dentate nucleus. Sensitivity, specificity, and interrater agreement were calculated for all settings and compared against the visual reads of parametric images (0-60-minutes, distribution volume ratios, DVR). RESULTS Patients with 4R-tauopathies in contrast to non-4R-tauopathies were detected at higher sensitivity in the 20-40-minute frame (common: 79%, scaled: 76%) compared to the 40-60-minute frame (common: 55%, scaled: 62%), albeit with reduced specificity in the common setting (20-40-min: 78%, 40-60-min: 95%), which was ameliorated in the intensity-scaled setting (20-40-min: 91%, 40-60-min: 96%). Combined assessment of multiple brain regions did not significantly improve diagnostic sensitivity, compared to assessing the basal ganglia alone (76% each). Evaluation of intensity-scaled parametric images resulted in higher sensitivity compared to intensity-scaled static scans (86% vs. 76%) at similar specificity (89% vs. 91%). CONCLUSION Visual reading of [18F]PI-2620 tau-PET scans demonstrated reliable detection of 4R-tauopathies, particularly when standardized processing methods and early imaging windows were employed. Parametric images should be preferred for visual assessment of 4R-tauopathies.
Collapse
Affiliation(s)
- Theresa Bauer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Alexander M Bernhardt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Jäck
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Sophia Stöcklein
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn/Cologne, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn/Cologne, Germany; Institute of Neuroscience and Medicine (INM-2), Molecular Organization of the Brain, Forschungszentrum Jülich, Germany
| | - Osama Sabri
- University Hospital Leipzig, Department of Nuclear Medicine, Leipzig, Germany
| | - Henryk Barthel
- University Hospital Leipzig, Department of Nuclear Medicine, Leipzig, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany; Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK; Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, University Hospital, LMU Munich, 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, LMU Munich, Munich, Germany; University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
| |
Collapse
|
9
|
Jiao F, Wang Q, Zhong J, Lin H, Lu J, Wang L, Wang M, Liu F, Jiang J, Zuo C. Relationships Between Glymphatic System Activity and Tau Burden, Dopaminergic Impairment, Abnormal Glucose Metabolism in Progressive Supranuclear Palsy. CNS Neurosci Ther 2025; 31:e70284. [PMID: 39963843 PMCID: PMC11833299 DOI: 10.1111/cns.70284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/23/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is a primary tauopathy characterized by dopaminergic impairment and abnormal glucose metabolism. The glymphatic system can promote the elimination of tau protein. The association between glymphatic function and pathological hallmark in neuroimaging remains unknown. METHODS Diffusion tensor imaging (DTI) and positron emission tomography (PET) scanning with 18F-Florzolotau, 18F-FPCIT, and 18F-FDG were performed in PSP patients. DTI analysis along the perivascular space (ALPS) index was computed to assess glymphatic function, while the semi-quantitative value was employed to measure tau burden and dopaminergic impairment. The PSP-related pattern (PSPRP) served as an indicator of abnormal metabolic brain network activity. RESULTS PSP patients exhibited changes in ALPS index and tau deposition. ALPS index, tau deposition, and PSPRP expression showed significant correlations with clinical scores. Additionally, ALPS index was correlated with tau deposition and PSPRP expression. However, neither ALPS index nor the clinical scores were correlated with striatum dysfunction. Finally, tau deposition in subcortical regions and PSPRP expression exhibited mediating effects between ALPS index and clinical scores. CONCLUSION The glymphatic dysfunction is associated with tau deposition and abnormal metabolic brain network activity and is independent of dopaminergic impairment in PSP.
Collapse
Affiliation(s)
- Fangyang Jiao
- Department of Nuclear Medicine & PET Center, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Qingmin Wang
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Jiayi Zhong
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Huamei Lin
- Department of Nuclear Medicine & PET Center, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Jiaying Lu
- Department of Nuclear Medicine & PET Center, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Luyao Wang
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Min Wang
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Fengtao Liu
- Department of Neurology, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| | - Jiehui Jiang
- Institute of Biomedical Engineering, School of Life ScienceShanghai UniversityShanghaiChina
| | - Chuantao Zuo
- Department of Nuclear Medicine & PET Center, National Clinical Research Center for Aging and Medicine & National Center for Neurological Disorders, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
10
|
Li CH, Fan SP, Shih MC, Weng YH, Chen TF, Li H, Cheng MF, Kuo MC, Peng PL, Higuchi M, Hsiao IT, Lin KJ, Lin CH. Subcortical tau burden correlates with regional brain atrophy and plasma markers in four-repeat tauopathy parkinsonism. JOURNAL OF PARKINSON'S DISEASE 2025; 15:214-226. [PMID: 39973504 DOI: 10.1177/1877718x241298192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background18F-florzolotau positron emission tomography (PET) assists in the in vivo diagnosis of progressive supranuclear palsy (PSP).ObjectiveWe aimed to investigate the relationship between 18F-florzolotau uptake and clinical severity, structural volume changes, and plasma markers in four-repeat tauopathies.MethodsA total of 80 participants were recruited: 35 with PSP (11 with PSP-Richardson syndrome and 24 with PSP non-Richardson syndrome), 9 with corticobasal syndrome (CBS), 10 with Alzheimer's disease (AD), 8 with Parkinson's disease, and 18 controls. All participants underwent 18F-florzolotau PET, brain magnetic resonance imaging (MRI), and plasma biomarker investigation (total and phosphorylated tau [pTau181], neurofilament light chain, and glial fibrillary acidic protein [GFAP]).Results18F-Florzolotau uptake was significantly higher in the subcortical regions of the pallidum, subthalamic nucleus (STN), midbrain, red nucleus, and raphe nucleus in PSP patients compared to the other groups (all p < 0.01). Subcortical tau tracer retention assisted in distinguishing PSP and CBS from controls (AUC = 0.836, p < 0.001). Tau tracer retention could differentiate PSP and CBS from AD in cortical (p < 0.001) and subcortical regions (p = 0.028). The motor severity of PSP positively correlated with tau burden in STN (p = 0.044) and substantia nigra (p = 0.035). Tau tracer uptake was associated with cortical volume changes in CBS (p = 0.031), PSP non-Richardson syndrome (p = 0.003), and AD (p = 0.044). Cortical tau retention correlated with plasma levels of GFAP (p = 0.001) and pTau181 (p = 0.036).ConclusionsSubcortical 18F-Florzolotau uptake assist the diagnosis of 4R tauopathy parkinsonism. Additionally, regional tau burden contributes to structural brain volume changes and correlates with plasma levels of GFAP and pTau181.
Collapse
Affiliation(s)
- Cheng-Hsuan Li
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sung-Pin Fan
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Chieh Shih
- School of Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Hsin Weng
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsun Li
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Mei-Fang Cheng
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Che Kuo
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Pei-Ling Peng
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Makoto Higuchi
- Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Ing-Tsung Hsiao
- Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Ju Lin
- Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Biomedical Engineering, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
11
|
Wongso H, Harada R, Furumoto S. Current Progress and Future Directions in Non-Alzheimer's Disease Tau PET Tracers. ACS Chem Neurosci 2025; 16:111-127. [PMID: 39762194 DOI: 10.1021/acschemneuro.4c00319] [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: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) and non-AD tauopathies are dominant public health issues driven by several factors, especially in the aging population. The discovery of first-generation radiotracers, including [18F]FDDNP, [11C]PBB3, [18F]flortaucipir, and the [18F]THK series, for the in vivo detection of tauopathies has marked a significant breakthrough in the fields of neuroscience and radiopharmaceuticals, creating a robust new category of labeled compounds: tau positron emission tomography (PET) tracers. Subsequently, other tau PET tracers with improved binding properties have been developed using various chemical scaffolds to target the three-repeat/four-repeat (3R/4R) tau folds in AD. In 2020, [18F]flortaucipir was approved by the U.S. Food and Drug Administration for PET imaging of tau pathology in adult patients with cognitive deficits undergoing evaluation for AD. Despite remarkable progress in the development of AD tau PET tracers, imaging agents for rare non-AD tauopathies (4R tauopathies [predominantly expressing a 4R tau isoform], involved in progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, and globular glial tauopathy, and 3R tauopathies [predominantly expressing a 3R tau isoform], such as Pick's disease) remain substantially underdeveloped. In this review, we discuss recent progress in tau PET tracer development, with particular emphasis on clinically validated tracers for AD and their potential use for non-AD tauopathies. Additionally, we highlight the critical need for further development of tau PET tracers specifically designed for non-AD tauopathies, an area that remains significantly underexplored despite its importance in advancing the understanding and diagnosis of these disorders.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten 15314, Indonesia
| | - Ryuichi Harada
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Shozo Furumoto
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
12
|
Tang G, Lu JY, Li XY, Yao RX, Yang YJ, Jiao FY, Chen MJ, Liang XN, Ju ZZ, Ge JJ, Zhao YX, Shen B, Wu P, Sun YM, Wu JJ, Yen TC, Zuo C, Wang J, Zhao QH, Zhang HW, Liu FT. 18F-Florzolotau PET Imaging Unveils Tau Pathology in Dementia with Lewy Bodies. Mov Disord 2025; 40:108-120. [PMID: 39555939 DOI: 10.1002/mds.30055] [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: 07/05/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) commonly exhibits a complex neuropathology, sharing characteristics with Alzheimer's disease (AD), including tau aggregates. However, studies using the 18F-AV-1451 tau tracer have shown inconsistent findings regarding both the extent and topographical distribution of tau pathology in DLB. OBJECTIVES Our aim was to elucidate the topographical patterns of tau deposition in DLB and to investigate the in vivo pathological distinction between DLB and AD in virtue of the 18F-Florzolotau positron emission tomography (PET) imaging. METHODS This cross-sectional study enrolled patients with DLB (n = 24), AD (n = 43), and cognitively healthy controls (n = 18). Clinical assessments and 18F-Florzolotau PET imaging were performed. 18F-Florzolotau binding was quantitatively assessed on PET images using standardized uptake value ratios and voxel-wise analysis. RESULTS 18F-Florzolotau PET imaging revealed widespread tau deposition across various cortical regions in DLB, uncovering heterogeneous topographical patterns. Among patients, 54.17% showed patterns similar to AD, whereas 16.67% exhibited distinct patterns. Compared to AD, DLB exhibited a unique in vivo neuropathological profile, characterized by a lower tau protein burden, heterogeneous topographical distributions, and a specific role of the medial temporal lobe in tau pathology. CONCLUSIONS 18F-Florzolotau PET imaging elucidated tau pathology patterns in DLB, providing valuable insights for future in vivo pathological differentiation and potential disease-modifying therapies. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Gan Tang
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia-Ying Lu
- Department of Nuclear Medicine and PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xin-Yi Li
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Rui-Xin Yao
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu-Jie Yang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Fang-Yang Jiao
- Department of Nuclear Medicine and PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming-Jia Chen
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiao-Niu Liang
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurology, Fudan University, Shanghai, China
| | - Zi-Zhao Ju
- Department of Nuclear Medicine and PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing-Jie Ge
- Department of Nuclear Medicine and PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Xin Zhao
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bo Shen
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Wu
- Department of Nuclear Medicine and PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Min Sun
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian-Jun Wu
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Chuantao Zuo
- Department of Nuclear Medicine and PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qian-Hua Zhao
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Neurology, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Hui-Wei Zhang
- Department of Nuclear Medicine and PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Feng-Tao Liu
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Liu FT, Lu JY, Li XY, Ge JJ, Sun YM, Yen TC, Jiao FY, Chen MJ, Zhao J, Yao RX, Tang G, Xu H, Lan XL, Lu J, Cui RX, Brendel M, Shi K, Guan YH, Rominger A, Wang J, Zuo CT. Visual reading for [ 18F]Florzolotau Tau PET scans in progressive supranuclear palsy. Eur J Nucl Med Mol Imaging 2025; 52:586-598. [PMID: 39352424 DOI: 10.1007/s00259-024-06923-3] [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: 03/15/2024] [Accepted: 09/14/2024] [Indexed: 01/15/2025]
Abstract
PURPOSE The identification of tau accumulation within living brains holds significant potential in facilitating accurate diagnosis of progressive supranuclear palsy (PSP). While visual assessment is frequently employed, standardized methods for tau positron emission tomography (PET) specifically in PSP are absent. We aimed to develop a visual reading algorithm dedicated to the evaluation of [18F]Florzolotau PET in PSP. METHODS 148 PSP and 30 healthy volunteers were divided into a development set (for the establishment of the reading rules; n = 89) and a testing set (for the validation of the reading rules; n = 89). For differential diagnosis, 55 α-synucleinopathies were additionally included into the testing set. The visual reading method was established by an experienced assessor (Reader 0) and was then validated by Reader 0 and two additional readers on regional and overall binary manners. A positive binding in both midbrain and globus pallidus/putamen regions was characterized as a PSP-like pattern, whereas any other pattern was classified as non-PSP-like. RESULTS Reader 1 (94.4%) and Reader 2 (93.8%) showed excellent agreement for the overall binary determination against Reader 0. The regional binary determinations of midbrain and globus pallidus/putamen showed excellent agreement among readers (kappa > 0.80). The overall binary evaluation demonstrated reproducibility of 86.1%, 94.4% and 77.8% for three readers. The visual reading algorithm showed high agreement with regional standardized uptake value ratios and clinical diagnoses. CONCLUSION Through the application of the suggested visual reading algorithm, [18F]Florzorotau PET imaging demonstrated a robust performance for the imaging diagnosis of PSP.
Collapse
Affiliation(s)
- Feng-Tao Liu
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia-Ying Lu
- Department of Nuclear Medicine & PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xin-Yi Li
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing-Jie Ge
- Department of Nuclear Medicine & PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Min Sun
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Fang-Yang Jiao
- Department of Nuclear Medicine & PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming-Jia Chen
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rui-Xin Yao
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Gan Tang
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hao Xu
- Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-Li Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Lu
- Department of Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Rui-Xue Cui
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU, Munich, Germany
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
- Department of Informatics, Technische Universität München, Munich, Germany
| | - Yi-Hui Guan
- Department of Nuclear Medicine & PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University of Bern, Bern, Switzerland
| | - Jian Wang
- Department of Neurology, National Research Center for Aging and Medicine, National Center for Neurological Disorders, and State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Chuan-Tao Zuo
- Department of Nuclear Medicine & PET Center, National Center for Neurological Disorders, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
- International Human Phenome Institutes (Shanghai), Shanghai, China.
- Human Phenome Institute, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Keir G, Roytman M, Mashriqi F, Shahsavarani S, Franceschi AM. Atypical Parkinsonian Syndromes: Structural, Functional, and Molecular Imaging Features. AJNR Am J Neuroradiol 2024; 45:1865-1877. [PMID: 39209485 PMCID: PMC11630880 DOI: 10.3174/ajnr.a8313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/16/2024] [Indexed: 09/04/2024]
Abstract
Atypical parkinsonian syndromes, also known as Parkinson-plus syndromes, are a heterogeneous group of movement disorders, including dementia with Lewy bodies (DLB), progressive supranuclear palsy (PSP), multisystem atrophy (MSA), and corticobasal degeneration (CBD). This review highlights the characteristic structural, functional, and molecular imaging features of these complex disorders. DLB typically demonstrates parieto-occipital hypometabolism with involvement of the cuneus on FDG-PET, whereas dopaminergic imaging, such as [123I]-FP-CIT SPECT (DaTscan) or fluorodopa (FDOPA)-PET, can be utilized as an adjunct for diagnosis. PSP typically shows midbrain atrophy on structural imaging, whereas FDG-PET may be useful to depict frontal lobe hypometabolism and tau-PET confirms underlying tauopathy. MSA typically demonstrates putaminal or cerebellar atrophy, whereas FDG-PET highlights characteristic nigrostriatal or olivopontocerebellar hypometabolism, respectively. Finally, CBD typically shows asymmetric atrophy in the superior parietal lobules and corpus callosum, whereas FDG and tau-PET demonstrate asymmetric hemispheric and subcortical involvement contralateral to the side of clinical deficits. Additional advanced neuroimaging modalities and techniques described may assist in the diagnostic work-up or are promising areas of emerging research.
Collapse
Affiliation(s)
- Graham Keir
- From the Neuroradiology Division (G.K., M.R.), Department of Radiology, Weill Cornell Medical College, NY-Presbyterian Hospital, New York, New York
| | - Michelle Roytman
- From the Neuroradiology Division (G.K., M.R.), Department of Radiology, Weill Cornell Medical College, NY-Presbyterian Hospital, New York, New York
| | - Faizullah Mashriqi
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| | - Shaya Shahsavarani
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| | - Ana M Franceschi
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| |
Collapse
|
15
|
Slemann L, Gnörich J, Hummel S, Bartos LM, Klaus C, Kling A, Kusche-Palenga J, Kunte ST, Kunze LH, Englert AL, Li Y, Vogler L, Katzdobler S, Palleis C, Bernhardt A, Jäck A, Zwergal A, Hopfner F, Roemer-Cassiano SN, Biechele G, Stöcklein S, Bischof G, van Eimeren T, Drzezga A, Sabri O, Barthel H, Respondek G, Grimmer T, Levin J, Herms J, Paeger L, Willroider M, Beyer L, Höglinger GU, Roeber S, Franzmeier N, Brendel M. Neuronal and oligodendroglial, but not astroglial, tau translates to in vivo tau PET signals in individuals with primary tauopathies. Acta Neuropathol 2024; 148:70. [PMID: 39580770 PMCID: PMC11586312 DOI: 10.1007/s00401-024-02834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Tau PET has attracted increasing interest as an imaging biomarker for 4-repeat (4R)-tauopathy progressive supranuclear palsy (PSP). However, the translation of in vitro 4R-tau binding to in vivo tau PET signals is still unclear. Therefore, we performed a translational study using a broad spectrum of advanced methodologies to investigate the sources of [18F]PI-2620 tau PET signals in individuals with 4R-tauopathies, including a pilot PET autopsy study in patients. First, we conducted a longitudinal [18F]PI-2620 PET/MRI study in a 4-repeat-tau mouse model (PS19) and detected elevated [18F]PI-2620 PET signals in the presence of high levels of neuronal tau. An innovative approach involving cell sorting after radiotracer injection in vivo revealed higher tracer uptake in single neurons than in the astrocytes of PS19 mice. Regional [18F]PI-2620 tau PET signals during the lifetime correlated with the abundance of fibrillary tau and with autoradiography signal intensity in PSP patients and disease controls who underwent autopsy 2-63 months after tau PET. In autoradiography, tau-positive neurons and oligodendrocytes with a high AT8 density, but not tau-positive astrocytes, were the drivers of [18F]PI-2620 autoradiography signals in individuals with PSP. The high tau abundance in oligodendrocytes at the boundary of gray and white matter facilitated the identification of an optimized frontal lobe target region to detect the tau burden in patients with PSP. In summary, neuronal and oligodendroglial tau constitutes the dominant source of tau PET radiotracer binding in 4-repeat-tauopathies, translating to an in vivo signal.
Collapse
Affiliation(s)
- Luna Slemann
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Selina Hummel
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Carolin Klaus
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Agnes Kling
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Julia Kusche-Palenga
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Sebastian T Kunte
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Lea H Kunze
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Amelie L Englert
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Yunlei Li
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Letizia Vogler
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Sabrina Katzdobler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Alexander Bernhardt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Alexander Jäck
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- German Center for Vertigo and Balance Disorders, DSGZ, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Franziska Hopfner
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sebastian N Roemer-Cassiano
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU Hospital, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Radiology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sophia Stöcklein
- Department of Radiology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Gerard Bischof
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Thilo van Eimeren
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Department of Neurology, University Hospital Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Gesine Respondek
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Timo Grimmer
- Center for Cognitive Disorders, Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Center of Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lars Paeger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Marie Willroider
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sigrun Roeber
- Center of Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Nicolai Franzmeier
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Stroke and Dementia Research, LMU Hospital, LMU Munich, Munich, Germany
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, , University of Gothenburg, The Sahlgrenska Academy, Mölndal, Gothenburg, Sweden
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
16
|
Daté Y, Bun S, Takahata K, Kubota M, Momota Y, Iwabuchi Y, Tezuka T, Tabuchi H, Seki M, Yamamoto Y, Shikimoto R, Mimura Y, Hoshino T, Kurose S, Shimohama S, Suzuki N, Morimoto A, Oosumi A, Hoshino Y, Jinzaki M, Mimura M, Ito D. Can the clinical sign "head-turning sign" and simple questions in "Neucop-Q" predict amyloid β pathology? Alzheimers Res Ther 2024; 16:250. [PMID: 39568089 PMCID: PMC11580537 DOI: 10.1186/s13195-024-01605-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND To establish simple screening tests to suspect Alzheimer's disease (AD) pathology, the clinical sign "head-turning sign" (HTS), which is a patient's behavior of turning their head towards their partner to seek assistance with questions posed by the examiner during the interview, and the simple screening questionnaire for dementia named "Neucop-Q" were validated in participants diagnosed with amyloid and tau positron emission tomography (PET). METHODS We enrolled 155 patients: 47 cognitive normal, 36 with mild cognitive impairment, 64 with dementia, and 8 with psychiatric disorders. All participants underwent Neucop-Q [three questions: Consciousness/self-awareness of cognitive disabilities (C) normal/impaired (nor/imp), Pleasure/pastime (P) nor/imp, and News/knowledge on current topics (N) nor/imp] and amyloid/tau PET. Additionally, we measured plasma amyloid β (Aβ) 42/40 ratio, phosphorylated tau 181 (pTau181), glial fibrillary acidic protein (GFAP), and neurofilament light (NFL) levels and compared with HTS and Neucop-Q results. RESULTS The specificity and positive predictive value (PPV) of HTS positivity (HTSpos) were the highest (amyloid PET: 0.930 and 0.870, tau PET: 0.944 and 0.957, respectively), while Cimp and Nimp had a high negative predictive value (NPV) for amyloid PET (negativity) (0.750 and 0.725). Pimp showed high specificity for predicting non-AD tau positivity among non-AD participants without amyloid PET positivity (0.854). To validate these findings with PET results, we examined the correlation between well-established AD blood biomarkers and results obtained from these screening tests. HTSpos, Cimp, and Nimp were strongly associated with Aβ42/40 ratio (P < 0.0001, P = 0.0022, and P = 0.001), pTau181 (P < 0.0001, P = 0.0095, and P = 0.001), GFAP (P = 0.0372, P = 0.0088, and P = 0.0002), and amyloid PET Centiloid (P < 0.0001, P = 0.0210, and P = 0.0006), whereas Pimp increased neuroinflammation (GFAP; P = 0.0061) and was associated with non-AD tauopathy. The combination of Neucop-Q questions showed that Cimp/Pnor/Nimp subjects have the highest specificity and PPV (0.972 and 0.833) and were strongly associated with Aβ42/40 ratio (P = 0.0006), pTau181 (P = 0.0006), and amyloid PET Centiloid (P < 0.0001). CONCLUSION HTSpos, Cimp, and Nimp have diagnostic utility in suspecting MCI due to AD and AD, and Pimp has diagnostic value in non-AD tauopathy. HTSpos, Cimp, and Nimp were associated with biomarkers of Aβ pathology. HTS and Neucop-Q may serve as powerful first-line screening in memory clinics. TRIAL REGISTRATION UMIN Clinical Trials Registry (UMIN-CTR) under registration numbers 000032027 (Registration date: 2018/03/31) and 000030248 (Registration date: 2018/01/01).
Collapse
Affiliation(s)
- Yugaku Daté
- Department of Strokology and Neurology, Saiseikai Yokohamashi-Tobu Hospital, 3-6-1 Shimosueyoshi, Tsurumi-ku, Yokohama City, 230-8765, Japan
| | - Shogyoku Bun
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Keisuke Takahata
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Functional Brain Imaging, Institute for Quantum Medical Science Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Chiba, 263-8555, Japan
| | - Masahito Kubota
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yuki Momota
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Functional Brain Imaging, Institute for Quantum Medical Science Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Chiba, 263-8555, Japan
| | - Yu Iwabuchi
- Department of Radiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Toshiki Tezuka
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hajime Tabuchi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Morinobu Seki
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yasuharu Yamamoto
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Functional Brain Imaging, Institute for Quantum Medical Science Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Chiba, 263-8555, Japan
| | - Ryo Shikimoto
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yu Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Takayuki Hoshino
- Memory center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160- 8582, Japan
| | - Shin Kurose
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Functional Brain Imaging, Institute for Quantum Medical Science Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Chiba, 263-8555, Japan
| | - Sho Shimohama
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Natsumi Suzuki
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Ayaka Morimoto
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Azusa Oosumi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yuka Hoshino
- Memory center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160- 8582, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Daisuke Ito
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Memory center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160- 8582, Japan.
| |
Collapse
|
17
|
Studart-Neto A, Barbosa BJAP, Coutinho AM, de Souza LC, Schilling LP, da Silva MNM, Castilhos RM, Bertolucci PHF, Borelli WV, Gomes HR, Fernandes GBP, Barbosa MT, Balthazar MLF, Frota NAF, Forlenza OV, Smid J, Brucki SMD, Caramelli P, Nitrini R, Engelhardt E, Resende EDPF. Guidelines for the use and interpretation of Alzheimer's disease biomarkers in clinical practice in Brazil: recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2024; 18:e2024C001. [PMID: 39534442 PMCID: PMC11556292 DOI: 10.1590/1980-5764-dn-2024-c001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, the diagnostic accuracy of Alzheimer's disease has been enhanced by the development of different types of biomarkers that indicate the presence of neuropathological processes. In addition to improving patient selection for clinical trials, biomarkers can assess the effects of new treatments on pathological processes. However, there is concern about the indiscriminate and poorly supported use of biomarkers, especially in asymptomatic individuals or those with subjective cognitive decline. Difficulties interpreting these tests, high costs, and unequal access make this scenario even more challenging in healthcare. This article presents the recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology (Departamento Científico de Neurologia Cognitiva e Envelhecimento da Academia Brasileira de Neurologia) regarding the rational use and interpretation of Alzheimer's disease biomarkers in clinical practice. The clinical diagnosis of cognitive-behavioral syndrome is recommended as the initial step to guide the request for biomarkers.
Collapse
Affiliation(s)
- Adalberto Studart-Neto
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Breno José Alencar Pires Barbosa
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Pernambuco, Hospital das Clínicas, Recife, Centro de Ciências Médicas, Recife PE, Brazil
- Universidade Federal de Pernambuco, Empresa Brasileira de Serviços Hospitalares, Hospital das Clínicas, Departamento de Neurologia, Recife PE, Brazil
| | - Artur Martins Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Radiologia, Centro de Medicina Nuclear, Laboratório de Investigação Médica (LIM 43), São Paulo SP, Brazil
- Hospital Sírio-Libanês, Medicina Nuclear e Serviço de PET-CT, São Paulo SP, Brazil
| | - Leonardo Cruz de Souza
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Lucas Porcello Schilling
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil
| | - Mari Nilva Maia da Silva
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Nina Rodrigues, Serviço de Neuropsiquiatria, São Luís MA, Brazil
| | - Raphael Machado Castilhos
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Centro de Neurologia Cognitiva e Comportamental, Porto Alegre RS, Brazil
| | - Paulo Henrique Ferreira Bertolucci
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| | - Wyllians Vendramini Borelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Ciências Morfológicas, Porto Alegre RS, Brazil
| | - Hélio Rodrigues Gomes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Laboratório de Líquido Cefalorraquidiano, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Laboratório de Investigação Médica (LIM 15), São Paulo SP, Brazil
- Departamento Científico de Líquido Cefalorraquiano, Academia Brasileira de Neurologia, São Paulo SP, Brazil
| | | | - Maira Tonidandel Barbosa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Marcio Luiz Figueredo Balthazar
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil
| | - Norberto Anízio Ferreira Frota
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil
- Universidade de Fortaleza, Fortaleza, CE, Brazil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brazil
| | - Jerusa Smid
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Sonia Maria Dozzi Brucki
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Paulo Caramelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Ricardo Nitrini
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Eliasz Engelhardt
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Psiquiatria, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| |
Collapse
|
18
|
Kaewchur T, Thientunyakit T, Chamroonrat W, Khiewvan B, Kiatkittikul P, Wongsurawat N, Chotipanich C, Likitjaroen Y, Senanarong V, Pasawang P, Sontrapornpol T, Poon-iad N, Amnuaywattakorn S, Tepmongkol S. Thai Guideline for Nuclear Medicine Investigations of Neurocognitive Disorders: Nuclear Medicine Society of Thailand, the Neurological Society of Thailand, and Thai Medical Physicist Society Collaboration. Diagnostics (Basel) 2024; 14:2474. [PMID: 39594140 PMCID: PMC11592784 DOI: 10.3390/diagnostics14222474] [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: 10/10/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Nuclear medicine investigations play a significant role in diagnosing dementia, mainly using imaging techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT). By providing functional and molecular data via brain imaging, nuclear medicine investigations offer valuable insights that complement clinical evaluations and structural imaging in the early detection, diagnosis, and differentiation of various types of dementia, leading to more accurate diagnosis and personalized treatment planning. Therefore, the Nuclear Medicine Society of Thailand, the Neurological Society of Thailand, and the Thai Medical Physicist Society have collaborated to establish these practical nuclear medicine investigation guidelines aiming to (1) identify the role of nuclear medicine studies in patients with neurocognitive disorders; (2) assist referrers in requesting the most appropriate procedure for diagnosis of each type of neurocognitive disorders; and (3) identify scientific evidence that is useful to assisting nuclear medicine professionals in recommending, performing, interpreting, and reporting the results of nuclear medicine investigations in patients with neurocognitive disorders.
Collapse
Affiliation(s)
- Tawika Kaewchur
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.K.)
- PET/CT and Cyclotron Center, Center for Medicine Excellence, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Tanyaluck Thientunyakit
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.T.); (B.K.); (N.P.-i.)
| | - Wichana Chamroonrat
- Division of Nuclear Medicine, Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (W.C.); (S.A.)
| | - Benjapa Khiewvan
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.T.); (B.K.); (N.P.-i.)
| | - Peerapon Kiatkittikul
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok 10210, Thailand; (P.K.); (C.C.)
| | - Nantaporn Wongsurawat
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Chanisa Chotipanich
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok 10210, Thailand; (P.K.); (C.C.)
| | - Yuttachai Likitjaroen
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand;
| | - Vorapun Senanarong
- Department of Internal Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Panya Pasawang
- Division of Nuclear Medicine, Department of Radiology, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand; (P.P.); (T.S.)
| | - Tanawat Sontrapornpol
- Division of Nuclear Medicine, Department of Radiology, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand; (P.P.); (T.S.)
| | - Nucharee Poon-iad
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (T.T.); (B.K.); (N.P.-i.)
| | - Sasithorn Amnuaywattakorn
- Division of Nuclear Medicine, Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (W.C.); (S.A.)
| | - Supatporn Tepmongkol
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Chulalongkorn University Biomedical Imaging Group (CUBIG), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Chula Neuroscience Center, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
- Center of Excellence in Cognitive Impairment and Dementia, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
19
|
Dilcher R, Wall S, Groß M, Katzdobler S, Wagemann O, Palleis C, Weidinger E, Fietzek U, Bernhardt A, Kurz C, Ferschmann C, Scheifele M, Zaganjori M, Gnörich J, Bürger K, Janowitz D, Rauchmann B, Stöcklein S, Bartenstein P, Villemagne V, Seibyl J, Sabri O, Barthel H, Perneczky R, Schöberl F, Zwergal A, Höglinger GU, Levin J, Franzmeier N, Brendel M. Combining cerebrospinal fluid and PI-2620 tau-PET for biomarker-based stratification of Alzheimer's disease and 4R-tauopathies. Alzheimers Dement 2024; 20:6896-6909. [PMID: 39263969 PMCID: PMC11485081 DOI: 10.1002/alz.14185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Recent advances in biomarker research have improved the diagnosis and monitoring of Alzheimer's disease (AD), but in vivo biomarker-based workflows to assess 4R-tauopathy (4RT) patients are currently missing. We suggest a novel biomarker-based algorithm to characterize AD and 4RTs. METHODS We cross-sectionally assessed combinations of cerebrospinal fluid measures (CSF p-tau181 and t-tau) and 18F-PI-2620 tau-positron emission tomography (PET) in patients with AD (n = 64), clinically suspected 4RTs (progressive supranuclear palsy or corticobasal syndrome, n = 82) and healthy controls (n = 19). RESULTS Elevated CSF p-tau181 and cortical 18F-PI-2620 binding was characteristic for AD while normal CSF p-tau181 with elevated subcortical 18F-PI-2620 binding was characteristic for 4RTs. 18F-PI-2620-assessed posterior cortical hypoperfusion could be used as an additional neuronal injury biomarker in AD. DISCUSSION The specific combination of CSF markers and 18F-PI-2620 tau-PET in disease-specific regions facilitates the biomarker-guided stratification of AD and 4RTs. This has implications for biomarker-aided diagnostic workflows and the advancement in clinical trials. HIGHLIGHTS Novel biomarker-based algorithm for differentiating AD and 4R-tauopathies. A combination of CSF p-tau181 and 18F-PI-2620 discriminates AD versus 4R tauopathies. Hypoperfusion serves as an additional neuronal injury biomarker in AD.
Collapse
Affiliation(s)
- Roxane Dilcher
- NeuroscienceMonash UniversityMelbourneAustralia
- Department of Nuclear MedicineUniversity Hospital of MunichLMU MunichMünchenGermany
| | - Stephan Wall
- Department of Nuclear MedicineUniversity Hospital of MunichLMU MunichMünchenGermany
| | - Mattes Groß
- Institute for Stroke and Dementia ResearchUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Sabrina Katzdobler
- (SyNergy), Munich Cluster for Systems NeurologyMünchenGermany
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
- (DZNE), German Center for Neurodegenerative DiseasesMünchenGermany
| | - Olivia Wagemann
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Carla Palleis
- (SyNergy), Munich Cluster for Systems NeurologyMünchenGermany
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
- (DZNE), German Center for Neurodegenerative DiseasesMünchenGermany
| | - Endy Weidinger
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Urban Fietzek
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Alexander Bernhardt
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
- (DZNE), German Center for Neurodegenerative DiseasesMünchenGermany
| | - Carolin Kurz
- Department of Psychiatry and PsychotherapyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Christian Ferschmann
- Department of Nuclear MedicineUniversity Hospital of MunichLMU MunichMünchenGermany
| | - Maximilian Scheifele
- Department of Nuclear MedicineUniversity Hospital of MunichLMU MunichMünchenGermany
| | - Mirlind Zaganjori
- Department of Nuclear MedicineUniversity Hospital of MunichLMU MunichMünchenGermany
- Department of Psychiatry and PsychotherapyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Johannes Gnörich
- Department of Nuclear MedicineUniversity Hospital of MunichLMU MunichMünchenGermany
| | - Katharina Bürger
- Institute for Stroke and Dementia ResearchUniversity Hospital of Munich, LMU MunichMünchenGermany
- (DZNE), German Center for Neurodegenerative DiseasesMünchenGermany
| | - Daniel Janowitz
- Institute for Stroke and Dementia ResearchUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Boris‐Stephan Rauchmann
- Department of Psychiatry and PsychotherapyUniversity Hospital of Munich, LMU MunichMünchenGermany
- Institute for NeuroradiologyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Sophia Stöcklein
- Department of RadiologyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Peter Bartenstein
- Department of Nuclear MedicineUniversity Hospital of MunichLMU MunichMünchenGermany
| | - Victor Villemagne
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Molecular Imaging & TherapyAustin HealthHeidelbergAustralia
| | - John Seibyl
- Institute for Neurodegenerative DisordersNew HavenConnecticutUSA
| | - Osama Sabri
- Department of Nuclear MedicineUniversity Hospital LeipzigLeipzigGermany
| | - Henryk Barthel
- Department of Nuclear MedicineUniversity Hospital LeipzigLeipzigGermany
| | - Robert Perneczky
- (SyNergy), Munich Cluster for Systems NeurologyMünchenGermany
- (DZNE), German Center for Neurodegenerative DiseasesMünchenGermany
- Department of Psychiatry and PsychotherapyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Florian Schöberl
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
| | - Andreas Zwergal
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
- German Center for Vertigo and Balance Disorders (DSGZ)University Hospital of Munich, LMU MunichMünchenGermany
| | - Günter U. Höglinger
- (SyNergy), Munich Cluster for Systems NeurologyMünchenGermany
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
- (DZNE), German Center for Neurodegenerative DiseasesMünchenGermany
| | - Johannes Levin
- (SyNergy), Munich Cluster for Systems NeurologyMünchenGermany
- Department of NeurologyUniversity Hospital of Munich, LMU MunichMünchenGermany
- (DZNE), German Center for Neurodegenerative DiseasesMünchenGermany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia ResearchUniversity Hospital of Munich, LMU MunichMünchenGermany
- (SyNergy), Munich Cluster for Systems NeurologyMünchenGermany
- Department of Psychiatry and NeurochemistryUniversity of GothenburgThe Sahlgrenska AcademyInstitute of Neuroscience and PhysiologyMölndal and GothenburgSweden
| | - Matthias Brendel
- Department of Nuclear MedicineUniversity Hospital of MunichLMU MunichMünchenGermany
- (SyNergy), Munich Cluster for Systems NeurologyMünchenGermany
- (DZNE), German Center for Neurodegenerative DiseasesMünchenGermany
| |
Collapse
|
20
|
Jang Y, Oh S, Hall AJ, Zhang Z, Tropea TF, Chen-Plotkin A, Rosenthal LS, Dawson TM, Na CH, Pantelyat AY. Biomarker discovery in progressive supranuclear palsy from human cerebrospinal fluid. Clin Proteomics 2024; 21:56. [PMID: 39342078 PMCID: PMC11437921 DOI: 10.1186/s12014-024-09507-3] [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: 06/05/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is a neurodegenerative disorder often misdiagnosed as Parkinson's Disease (PD) due to shared symptoms. PSP is characterized by the accumulation of tau protein in specific brain regions, leading to loss of balance, gaze impairment, and dementia. Diagnosing PSP is challenging, and there is a significant demand for reliable biomarkers. Existing biomarkers, including tau protein and neurofilament light chain (NfL) levels in cerebrospinal fluid (CSF), show inconsistencies in distinguishing PSP from other neurodegenerative disorders. Therefore, the development of new biomarkers for PSP is imperative. METHODS We conducted an extensive proteome analysis of CSF samples from 40 PSP patients, 40 PD patients, and 40 healthy controls (HC) using tandem mass tag-based quantification. Mass spectrometry analysis of 120 CSF samples was performed across 13 batches of 11-plex TMT experiments, with data normalization to reduce batch effects. Pathway, interactome, cell-type-specific enrichment, and bootstrap receiver operating characteristic analyses were performed to identify key candidate biomarkers. RESULTS We identified a total of 3,653 unique proteins. Our analysis revealed 190, 152, and 247 differentially expressed proteins in comparisons of PSP vs. HC, PSP vs. PD, and PSP vs. both PD and HC, respectively. Gene set enrichment and interactome analysis of the differentially expressed proteins in PSP CSF showed their involvement in cell adhesion, cholesterol metabolism, and glycan biosynthesis. Cell-type enrichment analysis indicated a predominance of neuronally-derived proteins among the differentially expressed proteins. The potential biomarker classification performance demonstrated that ATP6AP2 (reduced in PSP) had the highest AUC (0.922), followed by NEFM, EFEMP2, LAMP2, CHST12, FAT2, B4GALT1, LCAT, CBLN3, FSTL5, ATP6AP1, and GGH. CONCLUSION Biomarker candidate proteins ATP6AP2, NEFM, and CHI3L1 were identified as key differentiators of PSP from the other groups. This study represents the first large-scale use of mass spectrometry-based proteome analysis to identify cerebrospinal fluid (CSF) biomarkers specific to progressive supranuclear palsy (PSP) that can differentiate it from Parkinson's disease (PD) and healthy controls. Our findings lay a crucial foundation for the development and validation of reliable biomarkers, which will enhance diagnostic accuracy and facilitate early detection of PSP.
Collapse
Affiliation(s)
- Yura Jang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sungtaek Oh
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna J Hall
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhen Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas F Tropea
- Department of Neurology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Liana S Rosenthal
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Chan Hyun Na
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Alexander Y Pantelyat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Keir G, Li Y, Chiang G. Hybrid PET/MRI in Neurodegenerative Disorders. ADVANCES IN CLINICAL RADIOLOGY 2024; 6:121-135. [PMID: 39583180 PMCID: PMC11583654 DOI: 10.1016/j.yacr.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Affiliation(s)
- Graham Keir
- Neuroradiology Fellow, Division of Neuroradiology, Department of Radiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, USA
| | - Yi Li
- Associate Professor, Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61 st Street, New York, NY 10065, USA
| | - Gloria Chiang
- Vice Chair of Clinical and Translational Research, Director of the Brain Health Imaging Institute, Associate Professor, Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, USA
| |
Collapse
|
22
|
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
|
23
|
Malpetti M, Roemer SN, Harris S, Gross M, Gnörich J, Stephens A, Dewenter A, Steward A, Biel D, Dehsarvi A, Wagner F, Müller A, Koglin N, Weidinger E, Palleis C, Katzdobler S, Rupprecht R, Perneczky R, Rauchmann BS, Levin J, Höglinger GU, Brendel M, Franzmeier N. Neuroinflammation Parallels 18F-PI-2620 Positron Emission Tomography Patterns in Primary 4-Repeat Tauopathies. Mov Disord 2024; 39:1480-1492. [PMID: 39022835 DOI: 10.1002/mds.29924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Preclinical, postmortem, and positron emission tomography (PET) imaging studies have pointed to neuroinflammation as a key pathophysiological hallmark in primary 4-repeat (4R) tauopathies and its role in accelerating disease progression. OBJECTIVE We tested whether microglial activation (1) progresses in similar spatial patterns as the primary pathology tau spreads across interconnected brain regions, and (2) whether the degree of microglial activation parallels tau pathology spreading. METHODS We examined in vivo associations between tau aggregation and microglial activation in 31 patients with clinically diagnosed 4R tauopathies, using 18F-PI-2620 PET and 18F-GE180 (translocator protein [TSPO]) PET. We determined tau epicenters, defined as subcortical brain regions with highest tau PET signal, and assessed the connectivity of tau epicenters to cortical regions of interest using a 3-T resting-state functional magnetic resonance imaging template derived from age-matched healthy elderly controls. RESULTS In 4R tauopathy patients, we found that higher regional tau PET covaries with elevated TSPO-PET across brain regions that are functionally connected to each other (β = 0.414, P < 0.001). Microglial activation follows similar distribution patterns as tau and distributes primarily across brain regions strongly connected to patient-specific tau epicenters (β = -0.594, P < 0.001). In these regions, microglial activation spatially parallels tau distribution detectable with 18F-PI-2620 PET. CONCLUSIONS Our findings indicate that the spatial expansion of microglial activation parallels tau distribution across brain regions that are functionally connected to each other, suggesting that tau and inflammation are closely interrelated in patients with 4R tauopathies. The combination of in vivo tau and inflammatory biomarkers could therefore support the development of immunomodulatory strategies for disease-modifying treatments in these conditions. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Maura Malpetti
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Sebastian N Roemer
- Department of Neurology, LMU Hospital, LMU Hospital, LMU Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany
| | - Mattes Gross
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany
| | | | - Anna Dewenter
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Fabian Wagner
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | | | | | - Endy Weidinger
- Department of Neurology, LMU Hospital, LMU Hospital, LMU Munich, Munich, Germany
| | - Carla Palleis
- Department of Neurology, LMU Hospital, LMU Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, LMU Hospital, LMU Hospital, LMU Munich, Munich, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University Regensburg, Regensburg, Germany
| | - Robert Perneczky
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Munich, Germany
- Aging Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, LMU Hospital, LMU Munich, Munich, Germany
- Department of Neuroradiology, LMU Hospital, LMU Munich, Munich, Germany
| | - Johannes Levin
- Department of Neurology, LMU Hospital, LMU Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, LMU Hospital, LMU Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, The Sahlgrenska Academy, Gothenburg, Sweden
| |
Collapse
|
24
|
Ishiguro T, Kasuga K. Alzheimer's Disease-Related Cerebrospinal Fluid Biomarkers in Progressive Supranuclear Palsy. Brain Sci 2024; 14:859. [PMID: 39335355 PMCID: PMC11430815 DOI: 10.3390/brainsci14090859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Progressive Supranuclear Palsy (PSP) is the most common four-repeat tauopathy. PSP cases are typically characterized by vertical gaze palsy and postural instability; however, various phenotypes have been reported, making antemortem diagnosis based on clinical symptoms challenging. The development of biomarkers reflecting brain pathology and the ability to diagnose patients based on these biomarkers are essential for developing future intervention strategies, including disease-modifying therapies. However, despite many dedicated efforts, no highly specific fluid biomarker for PSP has yet been established. Conversely, several cerebrospinal fluid (CSF) biomarkers of Alzheimer's Disease (AD) have been established, and an AT(N) classification system has been proposed. Typically, among patients with AD, CSF amyloid β42 (Aβ42), but not Aβ40, is decreased, resulting in a reduction in the Aβ42/Aβ40 ratio, while tau phosphorylated at threonine 181 (p-tau181) and total tau (t-tau) are increased. Interestingly, the core CSF AD biomarkers show unique patterns in patients with PSP. Furthermore, reports have indicated that the CSF levels of both Aβ42 and Aβ40 are decreased independently of Aβ accumulation in PSP. Therefore, the Aβ42/Aβ40 ratio could potentially be used to differentiate PSP from AD. Additionally, studies have reported that CSF p-tau and t-tau are reduced in PSP, and that the neurofilament light chain is remarkably increased compared to healthy controls and patients with AD, even though PSP is a neurodegenerative disease associated with tau accumulation. These PSP-specific changes in AD-related core biomarkers may reflect the pathology of PSP and contribute to its diagnosis. As such, elucidating the mechanisms underlying the observed decreases in Aβ and tau levels could facilitate a better understanding of the pathogenesis of PSP.
Collapse
Affiliation(s)
- Takanobu Ishiguro
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8585, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8585, Japan
| |
Collapse
|
25
|
Gnörich J, Koehler M, Wind-Mark K, Klaus C, Zatcepin A, Palumbo G, Lalia M, Monasor LS, Beyer L, Eckenweber F, Scheifele M, Gildehaus FJ, von Ungern-Sternberg B, Barthel H, Sabri O, Bartenstein P, Herms J, Tahirovic S, Franzmeier N, Ziegler S, Brendel M. Towards multicenter β-amyloid PET imaging in mouse models: A triple scanner head-to-head comparison. Neuroimage 2024; 297:120748. [PMID: 39069223 DOI: 10.1016/j.neuroimage.2024.120748] [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: 03/29/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
AIM β-amyloid (Aβ) small animal PET facilitates quantification of fibrillar amyloidosis in Alzheimer's disease (AD) mouse models. Thus, the methodology is receiving growing interest as a monitoring tool in preclinical drug trials. In this regard, harmonization of data from different scanners at multiple sites would allow the establishment large collaborative cohorts and may facilitate efficacy comparison of different treatments. Therefore, we objected to determine the level of agreement of Aβ-PET quantification by a head-to-head comparison of three different state-of-the-art small animal PET scanners, which could help pave the way for future multicenter studies. METHODS Within a timeframe of 5 ± 2 weeks, transgenic APPPS1 (n = 9) and wild-type (WT) (n = 8) mice (age range: 13-16 months) were examined three times by Aβ-PET ([18F]florbetaben) using a Siemens Inveon DPET, a MedisonanoScan PET/MR, and a MedisonanoScan PET/CT with harmonized reconstruction protocols. Cortex-to-white-matter 30-60 min p.i. standardized uptake value ratios (SUVRCTX/WM) were calculated to compare binding differences, effect sizes (Cohen's d) and z-score values of APPPS1 relative to WT mice. Correlation coefficients (Pearson's r) were calculated for the agreement of individual SUVR between different scanners. Voxel-wise analysis was used to determine the agreement of spatial pathology patterns. For validation of PET imaging against the histological gold standard, individual SUVR values were subject to a correlation analysis with area occupancy of methoxy‑X04 staining. RESULTS All three small animal PET scanners yielded comparable group differences between APPPS1 and WT mice (∆PET=20.4 % ± 2.9 %, ∆PET/MR=18.4 % ± 4.5 %, ∆PET/CT=18.1 % ± 3.3 %). Voxel-wise analysis confirmed a high degree of congruency of the spatial pattern (Dice coefficient (DC)PETvs.PET/MR=83.0 %, DCPETvs.PET/CT=69.3 %, DCPET/MRvs.PET/CT=81.9 %). Differences in the group level variance of the three scanners resulted in divergent z-scores (zPET=11.5 ± 1.6; zPET/MR=5.3 ± 1.3; zPET/CT=3.4 ± 0.6) and effect sizes (dPET=8.5, dPET/MR=4.5, dPET/CT=4.1). However, correlations at the individual mouse level were still strong between scanners (rPETvs.PET/MR=0.96, rPETvs.PET/CT=0.91, rPET/MRvs.PET/CT=0.87; all p ≤ 0.0001). Methoxy-X04 staining exhibited a significant correlation across all three PET machines combined (r = 0.76, p < 0.0001) but also at individual level (PET: r = 0.81, p = 0.026; PET/MR: r = 0.89, p = 0.0074; PET/CT: r = 0.93, p = 0.0028). CONCLUSIONS Our comparison of standardized small animal Aβ-PET acquired by three different scanners substantiates the possibility of moving towards a multicentric approach in preclinical AD research. The alignment of image acquisition and analysis methods achieved good overall comparability between data sets. Nevertheless, differences in variance of sensitivity and specificity of different scanners may limit data interpretation at the individual mouse level and deserves methodological optimization.
Collapse
Affiliation(s)
- Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
| | - Mara Koehler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Carolin Klaus
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Giovanna Palumbo
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Manvir Lalia
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Laura Sebastian Monasor
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Graduate School of Systemic Neuroscience, LMU Munich, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, 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, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Center of Neuropathology and Prion Research, University of Munich, Munich Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Nicolai Franzmeier
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Center of Neuropathology and Prion Research, University of Munich, Munich Germany.
| |
Collapse
|
26
|
Quattrone A, Franzmeier N, Huppertz HJ, Klietz M, Roemer SN, Boxer AL, Levin J, Höglinger GU. Magnetic Resonance Imaging Measures to Track Atrophy Progression in Progressive Supranuclear Palsy in Clinical Trials. Mov Disord 2024; 39:1329-1342. [PMID: 38825840 DOI: 10.1002/mds.29866] [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: 01/16/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Several magnetic resonance imaging (MRI) measures have been suggested as progression biomarkers in progressive supranuclear palsy (PSP), and some PSP staging systems have been recently proposed. OBJECTIVE Comparing structural MRI measures and staging systems in tracking atrophy progression in PSP and estimating the sample size to use them as endpoints in clinical trials. METHODS Progressive supranuclear palsy-Richardson's syndrome (PSP-RS) patients with one-year-follow-up longitudinal brain MRI were selected from the placebo arms of international trials (NCT03068468, NCT01110720, NCT01049399) and the DescribePSP cohort. The discovery cohort included patients from the NCT03068468 trial; the validation cohort included patients from other sources. Multisite age-matched healthy controls (HC) were included for comparison. Several MRI measures were compared: automated atlas-based volumetry (44 regions), automated planimetric measures of brainstem regions, and four previously described staging systems, applied to volumetric data. RESULTS Of 508 participants, 226 PSP patients including discovery (n = 121) and validation (n = 105) cohorts, and 251 HC were included. In PSP patients, the annualized percentage change of brainstem and midbrain volume, and a combined index including midbrain, frontal lobe, and third ventricle volume change, were the progression biomarkers with the highest effect size in both cohorts (discovery: >1.6; validation cohort: >1.3). These measures required the lowest sample sizes (n < 100) to detect 30% atrophy progression, compared with other volumetric/planimetric measures and staging systems. CONCLUSIONS This evidence may inform the selection of imaging endpoints to assess the treatment efficacy in reducing brain atrophy rate in PSP clinical trials, with automated atlas-based volumetry requiring smaller sample size than staging systems and planimetry to observe significant treatment effects. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Andrea Quattrone
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
- Institute of Neurology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
- Neuroscience Research Centre, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| | | | - Martin Klietz
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Sebastian N Roemer
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU, Munich, Germany
| | - Adam L Boxer
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Johannes Levin
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| |
Collapse
|
27
|
Roemer SN, Brendel M, Gnörich J, Malpetti M, Zaganjori M, Quattrone A, Gross M, Steward A, Dewenter A, Wagner F, Dehsarvi A, Ferschmann C, Wall S, Palleis C, Rauchmann BS, Katzdobler S, Jäck A, Stockbauer A, Fietzek UM, Bernhardt AM, Weidinger E, Zwergal A, Stöcklein S, Perneczky R, Barthel H, Sabri O, Levin J, Höglinger GU, Franzmeier N. Subcortical tau is linked to hypoperfusion in connected cortical regions in 4-repeat tauopathies. Brain 2024; 147:2428-2439. [PMID: 38842726 DOI: 10.1093/brain/awae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/07/2024] [Accepted: 04/28/2024] [Indexed: 06/07/2024] Open
Abstract
Four-repeat (4R) tauopathies are neurodegenerative diseases characterized by cerebral accumulation of 4R tau pathology. The most prominent 4R tauopathies are progressive supranuclear palsy (PSP) and corticobasal degeneration characterized by subcortical tau accumulation and cortical neuronal dysfunction, as shown by PET-assessed hypoperfusion and glucose hypometabolism. Yet, there is a spatial mismatch between subcortical tau deposition patterns and cortical neuronal dysfunction, and it is unclear how these two pathological brain changes are interrelated. Here, we hypothesized that subcortical tau pathology induces remote neuronal dysfunction in functionally connected cortical regions to test a pathophysiological model that mechanistically links subcortical tau accumulation to cortical neuronal dysfunction in 4R tauopathies. We included 51 Aβ-negative patients with clinically diagnosed PSP variants (n = 26) or corticobasal syndrome (n = 25) who underwent structural MRI and 18F-PI-2620 tau-PET. 18F-PI-2620 tau-PET was recorded using a dynamic one-stop-shop acquisition protocol to determine an early 0.5-2.5 min post tracer-injection perfusion window for assessing cortical neuronal dysfunction, as well as a 20-40 min post tracer-injection window to determine 4R-tau load. Perfusion-PET (i.e. early window) was assessed in 200 cortical regions, and tau-PET was assessed in 32 subcortical regions of established functional brain atlases. We determined tau epicentres as subcortical regions with the highest 18F-PI-2620 tau-PET signal and assessed the connectivity of tau epicentres to cortical regions of interest using a resting-state functional MRI-based functional connectivity template derived from 69 healthy elderly controls from the ADNI cohort. Using linear regression, we assessed whether: (i) higher subcortical tau-PET was associated with reduced cortical perfusion; and (ii) cortical perfusion reductions were observed preferentially in regions closely connected to subcortical tau epicentres. As hypothesized, higher subcortical tau-PET was associated with overall lower cortical perfusion, which remained consistent when controlling for cortical tau-PET. Using group-average and subject-level PET data, we found that the seed-based connectivity pattern of subcortical tau epicentres aligned with cortical perfusion patterns, where cortical regions that were more closely connected to the tau epicentre showed lower perfusion. Together, subcortical tau-accumulation is associated with remote perfusion reductions indicative of neuronal dysfunction in functionally connected cortical regions in 4R-tauopathies. This suggests that subcortical tau pathology may induce cortical dysfunction, which may contribute to clinical disease manifestation and clinical heterogeneity.
Collapse
Affiliation(s)
- Sebastian N Roemer
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Matthias Brendel
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Andrea Quattrone
- Institute of Neurology, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mattes Gross
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Anna Dewenter
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Fabian Wagner
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Christian Ferschmann
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Stephan Wall
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Carla Palleis
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Boris S Rauchmann
- Department of Neuroradiology, University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Alexander Jäck
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Anna Stockbauer
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Urban M Fietzek
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Alexander M Bernhardt
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Endy Weidinger
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
- German Center for Vertigo and Balance Disorders (DSGZ), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Sophia Stöcklein
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Robert Perneczky
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London SW7 2BX, UK
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, 04103 Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital of Leipzig, 04103 Leipzig, Germany
| | - Johannes Levin
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, University Hospital, LMU Munich, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Department of Psychiatry and Neurochemistry, University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, SE 413 90 Mölndal and Gothenburg, Sweden
| |
Collapse
|
28
|
Wuehr M, Peto D, Fietzek UM, Katzdobler S, Nübling G, Zaganjori M, Brendel M, Levin J, Höglinger GU, Zwergal A. Low-intensity vestibular noise stimulation improves postural symptoms in progressive supranuclear palsy. J Neurol 2024; 271:4577-4586. [PMID: 38722328 PMCID: PMC11233287 DOI: 10.1007/s00415-024-12419-9] [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: 01/14/2024] [Revised: 03/01/2024] [Accepted: 04/29/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Postural imbalance and falls are an early disabling symptom in patients with progressive supranuclear palsy (PSP) of multifactorial origin that may involve abnormal vestibulospinal reflexes. Low-intensity noisy galvanic vestibular stimulation (nGVS) is a non-invasive treatment to normalize deficient vestibular function and attenuate imbalance in Parkinson's disease. The presumed therapeutic mode of nGVS is stochastic resonance (SR), a mechanism by which weak sensory noise stimulation can enhance sensory information processing. OBJECTIVE To examine potential treatment effects of nGVS on postural instability in 16 patients with PSP with a clinically probable and [18F]PI-2620 tau-PET-positive PSP. METHODS Effects of nGVS of varying intensity (0-0.7 mA) on body sway were examined, while patients were standing with eyes closed on a posturographic force plate. We assumed a bell-shaped response curve with maximal sway reductions at intermediate nGVS intensities to be indicative of SR. An established SR-curve model was fitted on individual patient outcomes and three experienced human raters had to judge whether responses to nGVS were consistent with the exhibition of SR. RESULTS We found nGVS-induced reductions of body sway compatible with SR in 9 patients (56%) with optimal improvements of 31 ± 10%. In eight patients (50%), nGVS-induced sway reductions exceeded the minimal clinically important difference (improvement: 34 ± 5%), indicative of strong SR. CONCLUSION nGVS yielded clinically relevant reductions in body sway compatible with the exhibition of SR in vestibular sensorimotor pathways in at least half of the assessed patients. Non-invasive vestibular noise stimulation may be thus a well-tolerated treatment strategy to ameliorate postural symptoms in PSP.
Collapse
Affiliation(s)
- Max Wuehr
- German Center for Vertigo and Balance Disorders (DSGZ), LMU University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany.
| | - Daniela Peto
- German Center for Vertigo and Balance Disorders (DSGZ), LMU University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Urban M Fietzek
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Schön Klinik München Schwabing, Munich, Germany
- Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE) E.V., Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE) E.V., Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Georg Nübling
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE) E.V., Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE) E.V., Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE) E.V., Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE) E.V., Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Andreas Zwergal
- German Center for Vertigo and Balance Disorders (DSGZ), LMU University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
29
|
Lindberg A, Murrell E, Tong J, Mason NS, Sohn D, Sandell J, Ström P, Stehouwer JS, Lopresti BJ, Viklund J, Svensson S, Mathis CA, Vasdev N. Ligand-based design of [ 18F]OXD-2314 for PET imaging in non-Alzheimer's disease tauopathies. Nat Commun 2024; 15:5109. [PMID: 38877019 PMCID: PMC11178805 DOI: 10.1038/s41467-024-49258-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 05/30/2024] [Indexed: 06/16/2024] Open
Abstract
Positron emission tomography (PET) imaging of tau aggregation in Alzheimer's disease (AD) is helping to map and quantify the in vivo progression of AD pathology. To date, no high-affinity tau-PET radiopharmaceutical has been optimized for imaging non-AD tauopathies. Here we show the properties of analogues of a first-in-class 4R-tau lead, [18F]OXD-2115, using ligand-based design. Over 150 analogues of OXD-2115 were synthesized and screened in post-mortem brain tissue for tau affinity against [3H]OXD-2115, and in silico models were used to predict brain uptake. [18F]OXD-2314 was identified as a selective, high-affinity non-AD tau PET radiotracer with favorable brain uptake, dosimetry, and radiometabolite profiles in rats and non-human primate and is being translated for first-in-human PET studies.
Collapse
Affiliation(s)
- Anton Lindberg
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Emily Murrell
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Junchao Tong
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - N Scott Mason
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Sohn
- Oxiant Discovery, SE-15136, Södertälje, Sweden
| | | | - Peter Ström
- Novandi Chemistry AB, SE-15136, Södertälje, Sweden
| | | | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.
- Department of Psychiatry, University of Toronto, Toronto, Canada.
| |
Collapse
|
30
|
Bischof GN, Brendel M, Barthel H, Theis H, Barbe M, Bartenstein P, Claasen J, Danek A, Höglinger G, Levin J, Marek K, Neumaier B, Palleis C, Patt M, Rullmann M, Saur D, Schroeter ML, Seibyl J, Song M, Stephens A, Sabri O, Drzezga A, van Eimeren T. Improved Tau PET SUVR Quantification in 4-Repeat Tau Phenotypes with [ 18F]PI-2620. J Nucl Med 2024; 65:952-955. [PMID: 38575191 PMCID: PMC11149601 DOI: 10.2967/jnumed.123.265930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/20/2024] [Indexed: 04/06/2024] Open
Abstract
We used a new data-driven methodology to identify a set of reference regions that enhanced the quantification of the SUV ratio of the second-generation tau tracer 2-(2-([18F]fluoro)pyridin-4-yl)-9H-pyrrolo[2,3-b:4,5-c']dipyridine ([18F]PI-2620) in a group of patients clinically diagnosed with 4-repeat tauopathy, specifically progressive supranuclear palsy or cortical basal syndrome. The study found that SUV ratios calculated using the identified reference regions (i.e., fusiform gyrus and crus-cerebellum) were significantly associated with symptom severity and disease duration. This establishes, for the first time to our knowledge, the suitability of [18F]PI-2620 for tracking disease progression in this 4-repeat disease population. This is an important step toward increased clinical utility, such as patient stratification and monitoring in disease-modifying treatment trials. Additionally, the applied methodology successfully optimized reference regions for automated detection of brain imaging tracers. This approach may also hold value for other brain imaging tracers.
Collapse
Affiliation(s)
- Gérard N Bischof
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany;
- Molecular Organization of the Brain, Institute for Neuroscience and Medicine, Jülich, Germany
| | - Matthias Brendel
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Hendrik Theis
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Barbe
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Peter Bartenstein
- Munich Cluster for Systems Neurology, Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Joseph Claasen
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Adrian Danek
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ken Marek
- InviCRO, LLC, Boston, Massachusetts
- Molecular Neuroimaging, a division of inviCRO, New Haven, Connecticut
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine, Nuclear Chemistry, Research Center Jülich, Jülich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Michael Rullmann
- Clinic for Cognitive Neurology, University Hospital of Leipzig, and Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Dorothee Saur
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Matthias L Schroeter
- Clinic for Cognitive Neurology, University Hospital of Leipzig, and Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - John Seibyl
- InviCRO, LLC, Boston, Massachusetts
- Molecular Neuroimaging, a division of inviCRO, New Haven, Connecticut
| | - Mengmeng Song
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Osama Sabri
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Molecular Organization of the Brain, Institute for Neuroscience and Medicine, Jülich, Germany
- German Center for Neurodegenerative Diseases, Bonn/Cologne, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
31
|
Ferschmann C, Messerschmidt K, Gnörich J, Barthel H, Marek K, Palleis C, Katzdobler S, Stockbauer A, Fietzek U, Finze A, Biechele G, Rumpf JJ, Saur D, Schroeter ML, Rullmann M, Beyer L, Eckenweber F, Wall S, Schildan A, Patt M, Stephens A, Classen J, Bartenstein P, Seibyl J, Franzmeier N, Levin J, Höglinger GU, Sabri O, Brendel M, Scheifele M. Tau accumulation is associated with dopamine deficiency in vivo in four-repeat tauopathies. Eur J Nucl Med Mol Imaging 2024; 51:1909-1922. [PMID: 38366196 PMCID: PMC11139736 DOI: 10.1007/s00259-024-06637-6] [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: 07/11/2023] [Accepted: 02/04/2024] [Indexed: 02/18/2024]
Abstract
PURPOSE We hypothesized that severe tau burden in brain regions involved in direct or indirect pathways of the basal ganglia correlate with more severe striatal dopamine deficiency in four-repeat (4R) tauopathies. Therefore, we correlated [18F]PI-2620 tau-positron-emission-tomography (PET) imaging with [123I]-Ioflupane single-photon-emission-computed tomography (SPECT) for dopamine transporter (DaT) availability. METHODS Thirty-eight patients with clinically diagnosed 4R-tauopathies (21 male; 69.0 ± 8.5 years) and 15 patients with clinically diagnosed α-synucleinopathies (8 male; 66.1 ± 10.3 years) who underwent [18F]PI-2620 tau-PET and DaT-SPECT imaging with a time gap of 3 ± 5 months were evaluated. Regional Tau-PET signals and DaT availability as well as their principal components were correlated in patients with 4R-tauopathies and α-synucleinopathies. Both biomarkers and the residuals of their association were correlated with clinical severity scores in 4R-tauopathies. RESULTS In patients with 4R-tauopathies, [18F]PI-2620 binding in basal ganglia and midbrain regions was negatively associated with striatal DaT availability (i.e. globus pallidus internus and putamen (β = - 0.464, p = 0.006, Durbin-Watson statistics = 1.824) in a multiple regression model. Contrarily, [18F]PI-2620 binding in the dentate nucleus showed no significant regression factor with DaT availability in the striatum (β = 0.078, p = 0.662, Durbin-Watson statistics = 1.686). Patients with α-synucleinopathies did not indicate any regional associations between [18F]PI-2620-binding and DaT availability. Higher DaT-SPECT binding relative to tau burden was associated with better clinical performance (β = - 0.522, p = 0.011, Durbin-Watson statistics = 2.663) in patients with 4R-tauopathies. CONCLUSION Tau burden in brain regions involved in dopaminergic pathways is associated with aggravated dopaminergic dysfunction in patients with clinically diagnosed primary tauopathies. The ability to sustain dopamine transmission despite tau accumulation may preserve motor function.
Collapse
Affiliation(s)
- Christian Ferschmann
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Johannes Gnörich
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Ken Marek
- InviCRO, LLC, Boston, MA, USA
- Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | - Carla Palleis
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Anna Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Urban Fietzek
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Anika Finze
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jost-Julian Rumpf
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Dorothee Saur
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Matthias L Schroeter
- Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Michael Rullmann
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Stephan Wall
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Andreas Schildan
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | | | - Joseph Classen
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - John Seibyl
- InviCRO, LLC, Boston, MA, USA
- Molecular Neuroimaging, A Division of inviCRO, New Haven, CT, USA
| | - Nicolai Franzmeier
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Johannes Levin
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
32
|
Matarazzo M, Pérez-Soriano A, Vafai N, Shahinfard E, Cheng KJC, McKenzie J, Neilson N, Miao Q, Schaffer P, Shinotoh H, Kordower JH, Sossi V, Stoessl AJ. Misfolded protein deposits in Parkinson's disease and Parkinson's disease-related cognitive impairment, a [ 11C]PBB3 study. NPJ Parkinsons Dis 2024; 10:96. [PMID: 38702305 PMCID: PMC11068893 DOI: 10.1038/s41531-024-00708-z] [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: 11/24/2023] [Accepted: 04/11/2024] [Indexed: 05/06/2024] Open
Abstract
Parkinson's disease (PD) is associated with aggregation of misfolded α-synuclein and other proteins, including tau. We designed a cross-sectional study to quantify the brain binding of [11C]PBB3 (a ligand known to bind to misfolded tau and possibly α-synuclein) as a proxy of misfolded protein aggregation in Parkinson's disease (PD) subjects with and without cognitive impairment and healthy controls (HC). In this cross-sectional study, nineteen cognitively normal PD subjects (CN-PD), thirteen cognitively impaired PD subjects (CI-PD) and ten HC underwent [11C]PBB3 PET. A subset of the PD subjects also underwent PET imaging with [11C](+)DTBZ to assess dopaminergic denervation and [11C]PBR28 to assess neuroinflammation. Compared to HC, PD subjects showed higher [11C]PBB3 binding in the posterior putamen but not the substantia nigra. There was no relationship across subjects between [11C]PBB3 and [11C]PBR28 binding in nigrostriatal regions. [11C]PBB3 binding was increased in the anterior cingulate in CI-PD compared to CN-PD and HC, and there was an inverse correlation between cognitive scores and [11C]PBB3 binding in this region across all PD subjects. Our results support a primary role of abnormal protein deposition localized to the posterior putamen in PD. This suggests that striatal axonal terminals are preferentially involved in the pathophysiology of PD. Furthermore, our findings suggest that anterior cingulate pathology might represent a significant in vivo marker of cognitive impairment in PD, in agreement with previous neuropathological studies.
Collapse
Affiliation(s)
- Michele Matarazzo
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Alexandra Pérez-Soriano
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada
| | - Nasim Vafai
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Elham Shahinfard
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Kevin Ju-Chieh Cheng
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Jessamyn McKenzie
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada
| | - Nicole Neilson
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada
| | | | | | - Hitoshi Shinotoh
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, Chiba, Japan
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - A Jon Stoessl
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, BC, Canada.
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
33
|
Zhang Y, Xin M, Wang C, Hu Z, Wang Y, Shao H, Liu J, Zhang C. Neuronal intranuclear inclusion disease with cerebellar white matter tau uptake and incidental meningioma. Eur J Nucl Med Mol Imaging 2024; 51:1794-1795. [PMID: 38273004 DOI: 10.1007/s00259-024-06615-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 01/27/2024]
Affiliation(s)
- Yan Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Mei Xin
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Cheng Wang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zhoumi Hu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yue Wang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hongda Shao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Chenpeng Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| |
Collapse
|
34
|
Bischof GN, Jaeger E, Giehl K, Jessen F, Onur OA, O'Bryant S, Kara E, Weiss PH, Drzezga A. Cortical Tau Aggregation Patterns associated with Apraxia in Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.09.24305535. [PMID: 38645131 PMCID: PMC11030486 DOI: 10.1101/2024.04.09.24305535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Objectives Apraxia is a core feature of Alzheimer's disease, but the pathomechanism of this characteristic symptom is not well understood. Here, we systematically investigated apraxia profiles in a well-defined group of patients with Alzheimer's disease (AD; N=32) who additionally underwent PET imaging with the second-generation tau PET tracer [18F]PI-2620. We hypothesized that specific patterns of tau pathology might be related to apraxic deficits. Methods Patients (N=32) with a biomarker-confirmed diagnosis of Alzheimer's disease were recruited in addition to a sample cognitively unimpaired controls (CU 1 ; N=41). Both groups underwent in-depth neuropsychological assessment of apraxia (Dementia Apraxia Screening Test; DATE and the Cologne Apraxia Screening; KAS). In addition, static PET imaging with [18F]PI-2620 was performed to assess tau pathology in the AD patients. To specifically investigate the association of apraxia with regional tau-pathology, we compared the PET-data from this group with an independent sample of amyloid-negative cognitively intact participants (CU 2; N=54) by generation of z-score-deviation maps as well as voxel- based multiple regression analyses. Results We identified significant clusters of tau-aggregation in praxis-related regions (e.g., supramarginal gyrus, angular gyrus, temporal, parietal and occipital regions) that were associated with apraxia. These regions were similar between the two apraxia assessments. No correlations between tau-tracer uptake in primary motor cortical or subcortical brain regions and apraxia were observed. Conclusions These results suggest that tau deposition in specific cortical brain regions may induce local neuronal dysfunction leading to a dose-dependent functional decline in praxis performance.
Collapse
|
35
|
Ferrone SR, Sanmartin MX, Ohara J, Jimenez JC, Feizullayeva C, Lodato Z, Shahsavarani S, Lacher G, Demissie S, Vialet JM, White TG, Wang JJ, Katz JM, Sanelli PC. Acute ischemic stroke outcomes in patients with COVID-19: a systematic review and meta-analysis. J Neurointerv Surg 2024; 16:333-341. [PMID: 37460215 DOI: 10.1136/jnis-2023-020489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/17/2023] [Indexed: 03/16/2024]
Abstract
BACKGROUND Although patients with COVID-19 have a higher risk of acute ischemic stroke (AIS), the impact on stroke outcomes remains uncertain. AIMS To determine the clinical outcomes of patients with AIS and COVID-19 (AIS-COVID+). METHODS We performed a systematic review and meta-analysis following the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. Our protocol was registered with the International Prospective Register of Systematic Reviews (CRD42020211977). Systematic searches were last performed on June 3, 2021 in EMBASE, PubMed, Web-of-Science, Scopus, and CINAHL Databases. INCLUSION CRITERIA (1) studies reporting outcomes on AIS-COVID+; (2) original articles published in 2020 or later; (3) study participants aged ≥18 years. EXCLUSION CRITERIA (1) case reports with <5 patients, abstracts, review articles; (2) studies analyzing novel interventions. Risk of bias was assessed using the Mixed Methods Appraisal Tool. Random-effects models estimated the pooled OR and 95% confidence intervals (95% CI) for mortality, modified Rankin Scale (mRS) score, length of stay (LOS), and discharge disposition. RESULTS Of the 43 selected studies, 46.5% (20/43) reported patients with AIS without COVID-19 (AIS-COVID-) for comparison. Random-effects model included 7294 AIS-COVID+ and 158 401 AIS-COVID-. Compared with AIS-COVID-, AIS-COVID+ patients had higher in-hospital mortality (OR=3.87 (95% CI 2.75 to 5.45), P<0.001), less mRS scores 0-2 (OR=0.53 (95% CI 0.46 to 0.62), P<0.001), longer LOS (mean difference=4.21 days (95% CI 1.96 to 6.47), P<0.001), and less home discharge (OR=0.31 (95% CI 0.21 to 0.47), P<0.001). CONCLUSIONS Patients with AIS-COVID had worse outcomes, with almost fourfold increased mortality, half the odds of mRS scores 0-2, and one-third the odds of home discharge. These findings confirm the significant impact of COVID-19 on early stroke outcomes.
Collapse
Affiliation(s)
- Sophia R Ferrone
- Institute for Health System Science, Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Maria X Sanmartin
- Institute for Health System Science, Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| | - Joseph Ohara
- Department of Radiology, Northwell Health, Manhasset, NY, USA
| | - Jean C Jimenez
- Department of Radiology, Northwell Health, Manhasset, NY, USA
| | | | - Zachary Lodato
- Institute for Health System Science, Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Shaya Shahsavarani
- Institute for Health System Science, Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Gregory Lacher
- Institute for Health System Science, Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Seleshi Demissie
- Department of Biostatistics, Northwell Health Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - Tim G White
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| | - Jason J Wang
- Institute for Health System Science, Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| | - Jeffrey M Katz
- Department of Neurology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Pina C Sanelli
- Institute for Health System Science, Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Department of Radiology, Northwell Health, Manhasset, NY, USA
| |
Collapse
|
36
|
Varlow C, Mathis CA, Vasdev N. In vitro evaluation of [ 3H]PI-2620 and structural derivatives in non-Alzheimer's tauopathies. Nucl Med Biol 2024; 130-131:108891. [PMID: 38458074 DOI: 10.1016/j.nucmedbio.2024.108891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/28/2024] [Accepted: 02/16/2024] [Indexed: 03/10/2024]
Abstract
Alzheimer's disease (AD) and non-AD tauopathies such as chronic traumatic encephalopathy (CTE), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD) are characterized by the abnormal aggregation of three-repeat (3R) and/or four-repeat (4R) tau isoforms. Several tau-PET tracers have been applied for human imaging of AD and non-AD tauopathies including [18F]PI-2620. Our objective is to evaluate [3H]PI-2620 and two promising structural derivatives, [3H]PI-2014 and [3H]F-4, using in vitro saturation assays and competitive binding assays against new chemical entities based on this scaffold in human AD tissues for comparison with PSP, CBD and CTE tissues. Thin section autoradiography was employed to assess specific binding and distribution of [3H]PI-2620 and [3H]F-4 in fresh-frozen human post-mortem AD, PSP, CBD and CTE tissues. Immunohistochemistry was performed for phospho-tau (AT8) and 4R-tau (RD4). Homogenate filtration binding assays were performed for saturation analysis and competitive binding studies against [3H]PI-2620. All compounds bound with high affinity in AD tissue. In PSP tissue [3H]PI-2620 demonstrated the highest affinity (5.3 nM) and in CBD tissue [3H]F-4 bound with the highest affinity (9.4 nM). Over 40 fluorinated derivatives based on PI-2620 and F-4 were screened in AD and PSP tissue. Notably, compound 2 was the most potent derivative in PSP tissue (Ki = 7.3 nM). By autoradiography, [3H]PI-2620 and [3H]F-4 demonstrated positive signals similar in intensity in AD, PSP and CTE tissues that were displaced by homologous blockade. Binding of both radiotracers aligned with immunostaining for 4R-tau. This work demonstrates that [3H]PI-2620 and [3H]F-4 show promise for imaging 4R-tau aggregates in non-AD tauopathies. PI-2620 continues to serve as a structural scaffold for PET radiotracers with higher affinity for non-AD tau over AD tau.
Collapse
Affiliation(s)
- Cassis Varlow
- Institute of Medical Science, University of Toronto, ON M5S 1A8, Canada; Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada.
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, PA 15213, USA
| | - Neil Vasdev
- Institute of Medical Science, University of Toronto, ON M5S 1A8, Canada; Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada.
| |
Collapse
|
37
|
Parmera JB, de Godoi Carneiro C, de Almeida IJ, de Oliveira MCB, Barbosa PM, Studart‐Neto A, Ono CR, Nitrini R, Buchpiguel CA, Barbosa ER, Brucki SMD, Coutinho AM. Probable 4-Repeat Tauopathy Criteria Predict Brain Amyloid Negativity, Distinct Clinical Features, and FDG-PET/MRI Neurodegeneneration Patterns in Corticobasal Syndrome. Mov Disord Clin Pract 2024; 11:238-247. [PMID: 38155526 PMCID: PMC10928325 DOI: 10.1002/mdc3.13959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/29/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Corticobasal syndrome (CBS) is associated with diverse underlying pathologies, including the four-repeat (4R)-tauopathies. The Movement Disorders Society (MDS) criteria for progressive supranuclear palsy (PSP) proposed the novel category "probable 4R-tauopathy" to address the phenotypic overlap between PSP and corticobasal degeneration (CBD). OBJECTIVES To investigate the clinical ability of the MDS-PSP criteria for probable 4R-tauopathy in predicting a negative amyloid-PET in CBS. Additionally, this study aims to explore CBS patients classified as 4R-tauopathy concerning their clinical features and neuroimaging degeneration patterns. METHODS Thirty-two patients with probable CBS were prospectively evaluated and split into those who fulfilled or did not fulfill the 4R-tauopathy criteria (CBS-4RT+ vs. CBS-4RT-). All patients underwent positron emission tomographies (PET) with [18 F]fluorodeoxyglucose and [11 C]Pittsburgh Compound-B (PIB) on a hybrid PET-MRI scanner to perform multimodal quantitative comparisons with a control group. RESULTS Eleven patients were clinically classified as CBS-4RT+, and only one had a positive PIB-PET. The CBS-4RT+ classification had 92% specificity, 52% sensitivity, and 69% accuracy in predicting a negative PIB-PET. The CBS-4RT+ group presented with dysarthria and perseveration more often than the CBS-4RT- group. Moreover, the CBS-4RT+ group showed a prominent frontal hypometabolism extending to the supplementary motor area and striatum, and brain atrophy at the anterior cingulate and bilateral striata. CONCLUSIONS The 4R-tauopathy criteria were highly specific in predicting a negative amyloid-PET in CBS. Patients classified as 4R-tauopathy presented distinct clinical aspects, as well as brain metabolism and atrophy patterns previously associated with tauopathies.
Collapse
Affiliation(s)
- Jacy Bezerra Parmera
- Department of Neurology, Hospital das ClínicasFaculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Camila de Godoi Carneiro
- Laboratory of Nuclear Medicine (LIM 43), Nuclear Medicine Division and Institute of RadiologyHospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Isabel Junqueira de Almeida
- Department of Physical Therapy, Speech, and Occupational Therapy, Hospital das ClínicasFaculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | | | - Pedro Melo Barbosa
- Department of Neurology, Hospital das ClínicasFaculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Adalberto Studart‐Neto
- Department of Neurology, Hospital das ClínicasFaculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Carla Rachel Ono
- Laboratory of Nuclear Medicine (LIM 43), Nuclear Medicine Division and Institute of RadiologyHospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Ricardo Nitrini
- Department of Neurology, Hospital das ClínicasFaculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Carlos Alberto Buchpiguel
- Laboratory of Nuclear Medicine (LIM 43), Nuclear Medicine Division and Institute of RadiologyHospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Egberto Reis Barbosa
- Department of Neurology, Hospital das ClínicasFaculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Sonia Maria Dozzi Brucki
- Department of Neurology, Hospital das ClínicasFaculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| | - Artur Martins Coutinho
- Laboratory of Nuclear Medicine (LIM 43), Nuclear Medicine Division and Institute of RadiologyHospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HC‐FMUSP)São PauloBrazil
| |
Collapse
|
38
|
Kubota M, Endo H, Takahata K, Tagai K, Suzuki H, Onaya M, Sano Y, Yamamoto Y, Kurose S, Matsuoka K, Seki C, Shinotoh H, Kawamura K, Zhang MR, Takado Y, Shimada H, Higuchi M. In vivo PET classification of tau pathologies in patients with frontotemporal dementia. Brain Commun 2024; 6:fcae075. [PMID: 38510212 PMCID: PMC10953627 DOI: 10.1093/braincomms/fcae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/23/2023] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
Frontotemporal dementia refers to a group of neurodegenerative disorders with diverse clinical and neuropathological features. In vivo neuropathological assessments of frontotemporal dementia at an individual level have hitherto not been successful. In this study, we aim to classify patients with frontotemporal dementia based on topologies of tau protein aggregates captured by PET with 18F-florzolotau (aka 18F-APN-1607 and 18F-PM-PBB3), which allows high-contrast imaging of diverse tau fibrils in Alzheimer's disease as well as in non-Alzheimer's disease tauopathies. Twenty-six patients with frontotemporal dementia, 15 with behavioural variant frontotemporal dementia and 11 with other frontotemporal dementia phenotypes, and 20 age- and sex-matched healthy controls were included in this study. They underwent PET imaging of amyloid and tau depositions with 11C-PiB and 18F-florzolotau, respectively. By combining visual and quantitative analyses of PET images, the patients with behavioural variant frontotemporal dementia were classified into the following subgroups: (i) predominant tau accumulations in frontotemporal and frontolimbic cortices resembling three-repeat tauopathies (n = 3), (ii) predominant tau accumulations in posterior cortical and subcortical structures indicative of four-repeat tauopathies (n = 4); (iii) amyloid and tau accumulations consistent with Alzheimer's disease (n = 4); and (iv) no overt amyloid and tau pathologies (n = 4). Despite these distinctions, clinical symptoms and localizations of brain atrophy did not significantly differ among the identified behavioural variant frontotemporal dementia subgroups. The patients with other frontotemporal dementia phenotypes were also classified into similar subgroups. The results suggest that PET with 18F-florzolotau potentially allows the classification of each individual with frontotemporal dementia on a neuropathological basis, which might not be possible by symptomatic and volumetric assessments.
Collapse
Affiliation(s)
- Manabu Kubota
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Sakyo-ku Kyoto 606-8507, Japan
| | - Hironobu Endo
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Keisuke Takahata
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenji Tagai
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Psychiatry, Jikei University Graduate School of Medicine, Tokyo 105-8461, Japan
| | - Hisaomi Suzuki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Psychiatry, National Hospital OrganizationShimofusa Psychiatric Center, Chiba 266-0007, Japan
| | - Mitsumoto Onaya
- Department of Psychiatry, National Hospital OrganizationShimofusa Psychiatric Center, Chiba 266-0007, Japan
| | - Yasunori Sano
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yasuharu Yamamoto
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shin Kurose
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kiwamu Matsuoka
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Psychiatry, Nara Medical University, Nara 634-8521, Japan
| | - Chie Seki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Hitoshi Shinotoh
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Kazunori Kawamura
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Functional Neurology and Neurosurgery, Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
39
|
Oh M, Oh SJ, Lee SJ, Oh JS, Seo SY, Ryu S, Roh JH, Lee JH, Kim JS. One-Year Longitudinal Changes in Tau Accumulation on [ 18F]PI-2620 PET in the Alzheimer Spectrum. J Nucl Med 2024; 65:453-461. [PMID: 38302152 DOI: 10.2967/jnumed.123.265893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 02/03/2024] Open
Abstract
We investigated the longitudinal changes in cortical tau accumulation and their association with cognitive decline in patients in the Alzheimer disease (AD) continuum using 2-(2-([18F]fluoro)pyridin-4-yl)-9H-pyrrolo[2,3-b:4,5c']dipyridine ([18F]PI-2620) PET. Methods: We prospectively enrolled 52 participants (age, 69.7 ± 8.4 y; 18 men and 34 women): 7 with normal cognition, 28 with mild cognitive impairment, and 17 with AD. They all completed the [18F]PI-2620 and [18F]florbetaben PET, MRI, and neuropsychologic tests at baseline and, excepting the [18F]florbetaben PET, at the 1-y follow-up. Amyloid-β (Aβ) PET images were visually scored as positive (+) or negative (-). Patients on the AD continuum, including Aβ+ mild cognitive impairment and AD, were classified into early-onset (EO+) (<65 y old) or late-onset (LO+) (≥65 y old) groups. [18F]PI-2620 PET SUV ratios (SUVRs) were determined by calculating the cerebral-to-inferior cerebellar ratio. Cortical volumes were calculated using 3-dimensional T1-weighted MRI. The correlation between tau accumulation progression and cognitive decline was also investigated. Results: The global [18F]PI-2620 PET SUVRs were 1.04 ± 0.07 in 15 Aβ- patients, 1.18 ± 0.21 in 20 LO+ patients (age, 76.7 ± 3.8 y), and 1.54 ± 0.38 in 17 EO+ patients (age, 63.4 ± 5.4 y; P < 0.001) at baseline. The global SUVR increased over 1 y by 0.05 ± 0.07 (3.90%) and 0.13 ± 0.22 (8.41%) in the LO+ and EO+ groups, respectively, whereas in the Aβ- groups, it remained unchanged. The EO+ group showed higher global and regional tau deposition than did the Aβ- and LO+ groups (P < 0.05 for each) and rapid accumulation in Braak stage V (0.15 ± 0.25; 9.10% ± 12.27%; P = 0.016 and 0.008), Braak stage VI (0.08 ± 0.12; 7.16% ± 10.06%; P < 0.006 and 0.005), and global SUVR (P = 0.013) compared with the Aβ- group. In the EO+ group, the changes in SUVR in Braak stages II-VI were strongly correlated with the baseline and changes in verbal memory (P < 0.03). The LO+ group showed higher tau accumulation in Braak stage I-IV areas than did the Aβ- group (P < 0.001 for each). In the LO+ group, the change in SUVR in Braak stages III and IV moderately correlated with the change in attention (P < 0.05), and the change in SUVR in Braak stages V and VI moderately correlated with the change in visuospatial function (P < 0.005). Conclusion: These findings suggest that [18F]PI-2620 PET can be a biomarker to provide regional and chronologic information about tau pathology in the AD continuum.
Collapse
Affiliation(s)
- Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Ju Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jungsu S Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Yeon Seo
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Department of Electrical and Electronic Engineering, Yonsei University, Seoul, Korea
| | - Soorack Ryu
- Biostatistical Consulting and Research Laboratory, Medical Research Collaborating Center, Hanyang University, Seoul, Korea
| | - Jee Hoon Roh
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Departments of Biomedical Sciences and Physiology, Korea University College of Medicine, Seoul, Korea; and
- Department of Neurology, Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea;
| |
Collapse
|
40
|
Ghaith AK, El-Hajj VG, Atallah E, Rios Zermeno J, Ravindran K, Gharios M, Hoang H, Bydon M, Ohlsson M, Elmi-Terander A, Tawk RG, Jabbour P. Impact of the pandemic and concomitant COVID-19 on the management and outcomes of middle cerebral artery strokes: a nationwide registry-based study. BMJ Open 2024; 14:e080738. [PMID: 38417967 PMCID: PMC10900352 DOI: 10.1136/bmjopen-2023-080738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/19/2024] [Indexed: 03/01/2024] Open
Abstract
OBJECTIVES To investigate the impact of the COVID-19 pandemic as well as concomitant COVID-19 itself on stroke care, focusing on middle cerebral artery (MCA) territory infarctions. DESIGN Registry-based study. SETTING We used the National Inpatient Sample (NIS) database, which covers a wide range of hospitals within the USA. PARTICIPANTS The NIS was queried for patients with MCA strokes between 2016 and 2020. In total, 35 231 patients were included. OUTCOME MEASURES Outcome measures were postprocedural complications, length of stays (LOSs), in-hospital mortality and non-routine discharge. Propensity score matching using all available baseline variables was performed to reduce confounders when comparing patients with and without concomitant COVID-19. RESULTS Mechanical thrombectomy (MT) was performed in 48.4%, intravenous thrombolysis (IVT) in 38.2%, and both MT and IVT (MT+IVT) in 13.4% of patients. A gradual increase in the use of MT and an opposite decrease in the use of IVT (p<0.001) was detected during the study period. Overall, 25.0% of all patients were admitted for MCA strokes during the pandemic period (2020), of these 209 (2.4%) were concomitantly diagnosed with COVID-19. Patients with MCA strokes and concomitant COVID-19 were significantly younger (64.9 vs 70.0; p<0.001), had significantly worse NIH Stroke Severity scores, and worse outcomes in terms of LOS (12.3 vs 8.2; p<0.001), in-hospital mortality (26.3% vs 9.8%; p<0.001) and non-routine discharge (84.2% vs 76.9%; p=0.013), as compared with those without COVID-19. After matching, only in-hospital mortality rates remained significantly higher in patients with COVID-19 (26.7% vs 8.5%; p<0.001). Additionally, patients with COVID-19 had higher rates of thromboembolic (12.3% vs 7.6%; p=0.035) and respiratory (11.3% vs 6.6%; p=0.029) complications. CONCLUSIONS Among patients with MCA stroke, those with concomitant COVID-19 were significantly younger and had higher stroke severity scores. They were more likely to experience thromboembolic and respiratory complications and in-hospital mortality compared with matched controls.
Collapse
Affiliation(s)
| | | | - Elias Atallah
- Neurological Surgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | | | | | - Maria Gharios
- Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | | | - Marcus Ohlsson
- Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | | - Rabih G Tawk
- Mayo Clinic in Florida, Jacksonville, Florida, USA
| | - Pascal Jabbour
- Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Planche V, Mansencal B, Manjon JV, Meissner WG, Tourdias T, Coupé P. Staging of progressive supranuclear palsy-Richardson syndrome using MRI brain charts for the human lifespan. Brain Commun 2024; 6:fcae055. [PMID: 38444913 PMCID: PMC10914441 DOI: 10.1093/braincomms/fcae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/22/2023] [Accepted: 02/19/2024] [Indexed: 03/07/2024] Open
Abstract
Brain charts for the human lifespan have been recently proposed to build dynamic models of brain anatomy in normal aging and various neurological conditions. They offer new possibilities to quantify neuroanatomical changes from preclinical stages to death, where longitudinal MRI data are not available. In this study, we used brain charts to model the progression of brain atrophy in progressive supranuclear palsy-Richardson syndrome. We combined multiple datasets (n = 8170 quality controlled MRI of healthy subjects from 22 cohorts covering the entire lifespan, and n = 62 MRI of progressive supranuclear palsy-Richardson syndrome patients from the Four Repeat Tauopathy Neuroimaging Initiative (4RTNI)) to extrapolate lifetime volumetric models of healthy and progressive supranuclear palsy-Richardson syndrome brain structures. We then mapped in time and space the sequential divergence between healthy and progressive supranuclear palsy-Richardson syndrome charts. We found six major consecutive stages of atrophy progression: (i) ventral diencephalon (including subthalamic nuclei, substantia nigra, and red nuclei), (ii) pallidum, (iii) brainstem, striatum and amygdala, (iv) thalamus, (v) frontal lobe, and (vi) occipital lobe. The three structures with the most severe atrophy over time were the thalamus, followed by the pallidum and the brainstem. These results match the neuropathological staging of tauopathy progression in progressive supranuclear palsy-Richardson syndrome, where the pathology is supposed to start in the pallido-nigro-luysian system and spreads rostrally via the striatum and the amygdala to the cerebral cortex, and caudally to the brainstem. This study supports the use of brain charts for the human lifespan to study the progression of neurodegenerative diseases, especially in the absence of specific biomarkers as in PSP.
Collapse
Affiliation(s)
- Vincent Planche
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, F-33000 Bordeaux, France
- Centre Mémoire Ressources Recherches, Service de Neurologie des Maladies Neurodégénératives, Pôle de Neurosciences Cliniques, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Boris Mansencal
- CNRS, Univ. Bordeaux, Bordeaux INP, Laboratoire Bordelais de Recherche en Informatique (LABRI), UMR5800, F-33400 Talence, France
| | - Jose V Manjon
- Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA), Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Wassilios G Meissner
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, F-33000 Bordeaux, France
- Service de Neurologie des Maladies Neurodégénératives, Réseau NS-Park/FCRIN, CHU Bordeaux, F-33000, Bordeaux, France
- Department of Medicine, Christchurch, and New Zealand Brain Research Institute, Christchurch, 8011, New Zealand
| | - Thomas Tourdias
- Inserm U1215—Neurocentre Magendie, Bordeaux F-33000, France
- Service de Neuroimagerie diagnostique et thérapeutique, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Pierrick Coupé
- CNRS, Univ. Bordeaux, Bordeaux INP, Laboratoire Bordelais de Recherche en Informatique (LABRI), UMR5800, F-33400 Talence, France
| |
Collapse
|
42
|
Phillips JS, Robinson JL, Cousins KAQ, Wolk DA, Lee EB, McMillan CT, Trojanowski JQ, Grossman M, Irwin DJ. Polypathologic Associations with Gray Matter Atrophy in Neurodegenerative Disease. J Neurosci 2024; 44:e0808232023. [PMID: 38050082 PMCID: PMC10860605 DOI: 10.1523/jneurosci.0808-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/01/2023] [Accepted: 10/26/2023] [Indexed: 12/06/2023] Open
Abstract
Mixed pathologies are common in neurodegenerative disease; however, antemortem imaging rarely captures copathologic effects on brain atrophy due to a lack of validated biomarkers for non-Alzheimer's pathologies. We leveraged a dataset comprising antemortem MRI and postmortem histopathology to assess polypathologic associations with atrophy in a clinically heterogeneous sample of 125 human dementia patients (41 female, 84 male) with T1-weighted MRI ≤ 5 years before death and postmortem ordinal ratings of amyloid-[Formula: see text], tau, TDP-43, and [Formula: see text]-synuclein. Regional volumes were related to pathology using linear mixed-effects models; approximately 25% of data were held out for testing. We contrasted a polypathologic model comprising independent factors for each proteinopathy with two alternatives: a model that attributed atrophy entirely to the protein(s) associated with the patient's primary diagnosis and a protein-agnostic model based on the sum of ordinal scores for all pathology types. Model fits were evaluated using log-likelihood and correlations between observed and fitted volume scores. Additionally, we performed exploratory analyses relating atrophy to gliosis, neuronal loss, and angiopathy. The polypathologic model provided superior fits in the training and testing datasets. Tau, TDP-43, and [Formula: see text]-synuclein burden were inversely associated with regional volumes, but amyloid-[Formula: see text] was not. Gliosis and neuronal loss explained residual variance in and mediated the effects of tau, TDP-43, and [Formula: see text]-synuclein on atrophy. Regional brain atrophy reflects not only the primary molecular pathology but also co-occurring proteinopathies; inflammatory immune responses may independently contribute to degeneration. Our findings underscore the importance of antemortem biomarkers for detecting mixed pathology.
Collapse
Affiliation(s)
- Jeffrey S Phillips
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - John L Robinson
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Katheryn A Q Cousins
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David A Wolk
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Edward B Lee
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Corey T McMillan
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - John Q Trojanowski
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Murray Grossman
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David J Irwin
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
43
|
Kimura T, Sato H, Kano M, Tatsumi L, Tomita T. Novel aspects of the phosphorylation and structure of pathological tau: implications for tauopathy biomarkers. FEBS Open Bio 2024; 14:181-193. [PMID: 37391389 PMCID: PMC10839341 DOI: 10.1002/2211-5463.13667] [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: 04/26/2023] [Revised: 06/17/2023] [Accepted: 06/29/2023] [Indexed: 07/02/2023] Open
Abstract
The deposition of highly phosphorylated and aggregated tau is a characteristic of tauopathies, including Alzheimer's disease. It has long been known that different isoforms of tau are aggregated in different cell types and brain regions in each tauopathy. Recent advances in analytical techniques revealed the details of the biochemical and structural biological differences of tau specific to each tauopathy. In this review, we explain recent advances in the analysis of post-translational modifications of tau, particularly phosphorylation, brought about by the development of mass-spectrometry and Phos-tag technology. We then discuss the structure of tau filaments in each tauopathy revealed by the advent of cryo-EM. Finally, we describe the progress in biofluid and imaging biomarkers for tauopathy. This review summarizes current efforts to elucidate the characteristics of pathological tau and the landscape of the use of tau as a biomarker to diagnose and determine the pathological stage of tauopathy.
Collapse
Affiliation(s)
- Taeko Kimura
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Haruaki Sato
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Maria Kano
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Lisa Tatsumi
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical SciencesThe University of TokyoJapan
| |
Collapse
|
44
|
Aguero C, Dhaynaut M, Amaral AC, Moon SH, Neelamegam R, Scapellato M, Carazo-Casas C, Kumar S, El Fakhri G, Johnson K, Frosch MP, Normandin MD, Gómez-Isla T. Head-to-head comparison of [ 18F]-Flortaucipir, [ 18F]-MK-6240 and [ 18F]-PI-2620 postmortem binding across the spectrum of neurodegenerative diseases. Acta Neuropathol 2024; 147:25. [PMID: 38280071 PMCID: PMC10822013 DOI: 10.1007/s00401-023-02672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/29/2024]
Abstract
We and others have shown that [18F]-Flortaucipir, the most validated tau PET tracer thus far, binds with strong affinity to tau aggregates in Alzheimer's (AD) but has relatively low affinity for tau aggregates in non-AD tauopathies and exhibits off-target binding to neuromelanin- and melanin-containing cells, and to hemorrhages. Several second-generation tau tracers have been subsequently developed. [18F]-MK-6240 and [18F]-PI-2620 are the two that have garnered most attention. Our recent data indicated that the binding pattern of [18F]-MK-6240 closely parallels that of [18F]-Flortaucipir. The present study aimed at the direct comparison of the autoradiographic binding properties and off-target profile of [18F]-Flortaucipir, [18F]-MK-6240 and [18F]-PI-2620 in human tissue specimens, and their potential binding to monoamine oxidases (MAO). Phosphor-screen and high resolution autoradiographic patterns of the three tracers were studied in the same postmortem tissue material from AD and non-AD tauopathies, cerebral amyloid angiopathy, synucleopathies, transactive response DNA-binding protein 43 (TDP-43)-frontotemporal lobe degeneration and controls. Our results show that the three tracers show nearly identical autoradiographic binding profiles. They all strongly bind to neurofibrillary tangles in AD but do not seem to bind to a significant extent to tau aggregates in non-AD tauopathies pointing to their limited utility for the in vivo detection of non-AD tau lesions. None of them binds to lesions containing β-amyloid, α-synuclein or TDP-43 but they all show strong off-target binding to neuromelanin and melanin-containing cells, as well as weaker binding to areas of hemorrhage. The autoradiographic binding signals of the three tracers are only weakly displaced by competing concentrations of selective MAO-B inhibitor deprenyl but not by MAO-A inhibitor clorgyline suggesting that MAO enzymes do not appear to be a significant binding target of any of them. These findings provide relevant insights for the correct interpretation of the in vivo behavior of these three tau PET tracers.
Collapse
Affiliation(s)
- Cinthya Aguero
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ana C Amaral
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - S-H Moon
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Margaret Scapellato
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Carlos Carazo-Casas
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Sunny Kumar
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew P Frosch
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gómez-Isla
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA.
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA.
| |
Collapse
|
45
|
Palleis C, Franzmeier N, Weidinger E, Bernhardt AM, Katzdobler S, Wall S, Ferschmann C, Harris S, Schmitt J, Schuster S, Gnörich J, Finze A, Biechele G, Lindner S, Albert NL, Bartenstein P, Sabri O, Barthel H, Rupprecht R, Nuscher B, Stephens AW, Rauchmann BS, Perneczky R, Haass C, Brendel M, Levin J, Höglinger GU. Association of Neurofilament Light Chain, [ 18F]PI-2620 Tau-PET, TSPO-PET, and Clinical Progression in Patients With β-Amyloid-Negative CBS. Neurology 2024; 102:e207901. [PMID: 38165362 PMCID: PMC10834119 DOI: 10.1212/wnl.0000000000207901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Corticobasal syndrome (CBS) with underlying 4-repeat tauopathy is a progressive neurodegenerative disease characterized by declining cognitive and motor functions. Biomarkers for assessing pathologic brain changes in CBS including tau-PET, 18 kDa translocator protein (TSPO)-PET, structural MRI, neurofilament light chain (NfL), or glial fibrillary acidic protein (GFAP) have recently been evaluated for differential diagnosis and disease staging, yet their association with disease trajectories remains unclear. Therefore, we performed a head-to-head comparison of neuroimaging (tau-PET, TSPO-PET, structural MRI) and plasma biomarkers (NfL, GFAP) as prognostic tools for longitudinal clinical trajectories in β-amyloid (Aβ)-negative CBS. METHODS We included patients with clinically diagnosed Aβ-negative CBS with clinical follow-up data who underwent baseline structural MRI and plasma-NfL analysis for assessing neurodegeneration, [18F]PI-2620-PET for assessing tau pathology, [18F]GE-180-PET for assessing microglia activation, and plasma-GFAP analysis for assessing astrocytosis. To quantify tau and microglia load, we assessed summary scores of whole-brain, cortical, and subcortical PET signal. For structural MRI analysis, we quantified subcortical and cortical gray matter volume. Plasma NfL and GFAP values were assessed using Simoa-based immunoassays. Symptom progression was determined using a battery of cognitive and motor tests (i.e., Progressive Supranuclear Palsy Rating Scale [PSPRS]). Using linear mixed models, we tested whether the assessed biomarkers at baseline were associated with faster symptom progression over time (i.e., time × biomarker interaction). RESULTS Overall, 21 patients with Aβ-negative CBS with ∼2-year clinical follow-up data were included. Patients with CBS with more widespread global tau-PET signal showed faster clinical progression (PSPRS: B/SE = 0.001/0.0005, p = 0.025), driven by cortical rather than subcortical tau-PET. By contrast, patients with higher global [18F]GE-180-PET readouts showed slower clinical progression (PSPRS: B/SE = -0.056/0.023, p = 0.019). No association was found between gray matter volume and clinical progression. Concerning fluid biomarkers, only higher plasma-NfL (PSPRS: B/SE = 0.176/0.046, p < 0.001) but not GFAP was associated with faster clinical deterioration. In a subsequent sensitivity analysis, we found that tau-PET, TSPO-PET, and plasma-NfL showed significant interaction effects with time on clinical trajectories when tested in the same model. DISCUSSION [18F]PI-2620 tau-PET, [18F]GE-180 TSPO-PET, and plasma-NfL show prognostic potential for clinical progression in patients with Aβ-negative CBS with probable 4-repeat tauopathy, which can be useful for clinical decision-making and stratifying patients in clinical trials.
Collapse
Affiliation(s)
- Carla Palleis
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Nicolai Franzmeier
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Endy Weidinger
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Alexander M Bernhardt
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Sabrina Katzdobler
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Stephan Wall
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Christian Ferschmann
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Stefanie Harris
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Julia Schmitt
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Sebastian Schuster
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Johannes Gnörich
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Anika Finze
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Gloria Biechele
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Simon Lindner
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Nathalie L Albert
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Peter Bartenstein
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Osama Sabri
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Henryk Barthel
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Rainer Rupprecht
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Brigitte Nuscher
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Andrew W Stephens
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Boris-Stephan Rauchmann
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Robert Perneczky
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Christian Haass
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Matthias Brendel
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Johannes Levin
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Günter U Höglinger
- From the Departments of Neurology (C.P., E.W., A.M.B., S.K., J.L., G.U.H.), Nuclear Medicine (S.W., C.F., S.H., J.S., S.S., J.G., A.F., G.B., S.L., N.L.A., P.B., M.B.), and Psychiatry and Psychotherapy (B.-S.R., R.P.) and the Institutes for Stroke and Dementia Research (N.F.) and Neuroradiology (B.-S.R.), University Hospital, LMU Munich, Germany; Munich Cluster for Systems Neurology (C.P., N.F., S.K., P.B., R.P., C.H., M.B., J.L.), SyNergy, Germany; German Center for Neurodegenerative Diseases (C.P., E.W., A.M.B., S.K., B.N., B.-S.R., R.P., C.H., M.B., J.L., G.U.H.), DZNE-Munich, Germany; Department of Nuclear Medicine (O.S., H.B.), Leipzig University Medical Centre; Department of Psychiatry and Psychotherapy (R.R.), University of Regensburg, Germany; Life Molecular Imaging GmbH (A.W.S.), Berlin, Germany; Sheffield Institute for Translational Neuroscience (SITraN) (B.-S.R., R.P.), University of Sheffield, United Kingdom; Ageing Epidemiology Research Unit (AGE) (R.P.), School of Public Health, Imperial College London, United Kingdom; and Chair of Metabolic Biochemistry (C.H.), Biomedical Center (BMC), Ludwig-Maximilians-Universität LMU, Munich, Germany
| |
Collapse
|
46
|
Theis H, Barbe MT, Drzezga A, Fink GR, Neumaier B, Bischof GN, van Eimeren T. Progressive Supranuclear Palsy: Subcortical Tau Depositions Are Associated with Cortical Perfusion in Frontal and Limbic Regions. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1271-1276. [PMID: 38995804 PMCID: PMC11380218 DOI: 10.3233/jpd-240210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 07/14/2024]
Abstract
In progressive supranuclear palsy (PSP), subcortical tau and cortical perfusion can be assessed using the tracer [18F]PI-2620. We investigated if subcortical tau (globus pallidus internus, dentate nucleus) and frontal/limbic perfusion correlate in a cohort of 32 PSP patients. Tau in subcortical regions showed significant negative correlation with perfusion in limbic cortex. Perfusion in frontal regions was negatively associated with tau in both subcortical regions, but the significance threshold was only passed for the dentate nucleus. A reason could be a diaschisis-like phenomenon; that is, subcortical tau could lead to reduced connectivity to frontal regions and, thereby, to decreased perfusion.
Collapse
Affiliation(s)
- Hendrik Theis
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Michael T. Barbe
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Gottingen, Germany
- Institute for Neuroscience and Medicine (INM-2), Research Center Jülich, Jülich, Germany
| | - Gereon R. Fink
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
- Institute for Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Cologne, Germany
- Institute of Neuroscience & Medicine (INM-5), Nuclear Chemistry, Research Center Jülich, Jülich, Germany
| | - Gérard N. Bischof
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
- Institute for Neuroscience and Medicine (INM-2), Research Center Jülich, Jülich, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
47
|
Rueb M, Rauen K, Koerte IK, Gersing A, Zetterberg H, Simrén J, Brendel M, Adorjan K. Traumatic Encephalopathy Syndrome and Tauopathy in a 19-Year-Old With Child Abuse. Neurotrauma Rep 2023; 4:857-862. [PMID: 38156074 PMCID: PMC10754342 DOI: 10.1089/neur.2023.0078] [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] [Indexed: 12/30/2023] Open
Abstract
The majority of traumatic encephalopathy syndrome (TES) cases have been reported in former contact sport athletes. This is the first case with TES in a 19-year-old male patient with progressive cognitive decline after daily domestic physical violence through repeated hits to the head for 15 years. The patient presented with a moderate depressive episode and progressive cognitive decline. Tau positron emission tomography (PET) with 220 MBq of [18F]PI-2620 revealed increased focal signal at the frontal and parietal white/gray matter border. Brain magnetic resonance imaging (MRI) showed a cavum septum pellucidum, reduced left-sided hippocampal volume, and a left midbrain lesion. Cerebrospinal fluid results showed elevated total and p-tau. Neurocognitive testing at admission showed memory deficits clearly below average, and hampered dysfunctions according to the slow processing speed with a low mistake rate, indicating the acquired, thus secondary, attentional deficits. We diagnosed the patient with a TES suggestive of chronic traumatic encephalopathy and classified him as having subtle/mild functional limitation with a most likely transition to mild dementia within the TES criteria. This report underlines child abuse as a relevant criterion in diagnosing TES in cases with repetitive hits to the head. In addition to clinical markers, we show the relevance of fluid tau biomarkers and tau-PET to support the diagnosis of TES according to the recently published diagnosis criteria for TES.
Collapse
Affiliation(s)
- Mike Rueb
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Munich, Germany
- Pettenkofer School of Public Health, Munich, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology, LMU University Hospital, LMU Munich, Munich, Germany
- Center for International Health (CIH LMU), LMU University Hospital, LMU Munich, Munich, Germany
| | - Katrin Rauen
- Department of Geriatric Psychiatry, Psychiatric Hospital Zurich, University of Zurich, Zurich, Switzerland
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Munich, Germany
| | - Inga Katharina Koerte
- Psychiatric Neuroimaging Laboratory, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Child and Adolescent Psychiatry, Psychosomatic, and Psychotherapy, LMU University Hospital, Munich, Germany
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandra Gersing
- Department of Neuroradiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, United Kingdom
- UK Dementia Research Institute, University College London, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Kristina Adorjan
- Department of Psychiatry and Psychotherapy, LMU University Hospital, Munich, Germany
- Center for International Health (CIH LMU), LMU University Hospital, LMU Munich, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics, LMU University Hospital, Munich, Germany
| |
Collapse
|
48
|
Strobel J, Müller HP, Ludolph AC, Beer AJ, Sollmann N, Kassubek J. New Perspectives in Radiological and Radiopharmaceutical Hybrid Imaging in Progressive Supranuclear Palsy: A Systematic Review. Cells 2023; 12:2776. [PMID: 38132096 PMCID: PMC10742083 DOI: 10.3390/cells12242776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disease characterized by four-repeat tau deposition in various cell types and anatomical regions, and can manifest as several clinical phenotypes, including the most common phenotype, Richardson's syndrome. The limited availability of biomarkers for PSP relates to the overlap of clinical features with other neurodegenerative disorders, but identification of a growing number of biomarkers from imaging is underway. One way to increase the reliability of imaging biomarkers is to combine different modalities for multimodal imaging. This review aimed to provide an overview of the current state of PSP hybrid imaging by combinations of positron emission tomography (PET) and magnetic resonance imaging (MRI). Specifically, combined PET and MRI studies in PSP highlight the potential of [18F]AV-1451 to detect tau, but also the challenge in differentiating PSP from other neurodegenerative diseases. Studies over the last years showed a reduced synaptic density in [11C]UCB-J PET, linked [11C]PK11195 and [18F]AV-1451 markers to disease progression, and suggested the potential role of [18F]RO948 PET for identifying tau pathology in subcortical regions. The integration of quantitative global and regional gray matter analysis by MRI may further guide the assessment of reduced cortical thickness or volume alterations, and diffusion MRI could provide insight into microstructural changes and structural connectivity in PSP. Challenges in radiopharmaceutical biomarkers and hybrid imaging require further research targeting markers for comprehensive PSP diagnosis.
Collapse
Affiliation(s)
- Joachim Strobel
- Department of Nuclear Medicine, University Hospital Ulm, 89081 Ulm, Germany;
| | - Hans-Peter Müller
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
| | - Albert C. Ludolph
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
- German Center for Neurodegenerative Diseases (DZNE), Ulm University, 89081 Ulm, Germany
| | - Ambros J. Beer
- Department of Nuclear Medicine, University Hospital Ulm, 89081 Ulm, Germany;
| | - Nico Sollmann
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, 89081 Ulm, Germany;
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Jan Kassubek
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
- German Center for Neurodegenerative Diseases (DZNE), Ulm University, 89081 Ulm, Germany
| |
Collapse
|
49
|
Blazhenets G, Soleimani-Meigooni DN, Thomas W, Mundada N, Brendel M, Vento S, VandeVrede L, Heuer HW, Ljubenkov P, Rojas JC, Chen MK, Amuiri AN, Miller Z, Gorno-Tempini ML, Miller BL, Rosen HJ, Litvan I, Grossman M, Boeve B, Pantelyat A, Tartaglia MC, Irwin DJ, Dickerson BC, Baker SL, Boxer AL, Rabinovici GD, La Joie R. [ 18F]PI-2620 Binding Patterns in Patients with Suspected Alzheimer Disease and Frontotemporal Lobar Degeneration. J Nucl Med 2023; 64:1980-1989. [PMID: 37918868 PMCID: PMC10690126 DOI: 10.2967/jnumed.123.265856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/27/2023] [Indexed: 11/04/2023] Open
Abstract
Tau PET has enabled the visualization of paired helical filaments of 3 or 4 C-terminal repeat tau in Alzheimer disease (AD), but its ability to detect aggregated tau in frontotemporal lobar degeneration (FTLD) spectrum disorders is uncertain. We investigated 2-(2-([18F]fluoro)pyridin-4-yl)-9H-pyrrolo[2,3-b:4,5c']dipyridine ([18F]PI-2620), a newer tracer with ex vivo evidence for binding to FTLD tau, in a convenience sample of patients with suspected FTLD and AD using a static acquisition protocol and parametric SUV ratio (SUVr) images. Methods: We analyzed [18F]PI-2620 PET data from 65 patients with clinical diagnoses associated with AD or FTLD neuropathology; most (60/65) also had amyloid-β (Aβ) PET. Scans were acquired 30-60 min after injection; SUVr maps (reference, inferior cerebellar cortex) were created for the full acquisition and for 10-min truncated sliding windows (30-40, 35-45,…50-60 min). Age- and sex-adjusted z score maps were computed for each patient, relative to 23 Aβ-negative cognitively healthy controls (HC). Mean SUVr in the globus pallidus, substantia nigra, subthalamic nuclei, dentate nuclei, white matter, and temporal gray matter was extracted for the full and truncated windows. Results: Patients with suspected AD neuropathology (Aβ-positive patients with mild cognitive impairment or AD dementia) showed high-intensity temporoparietal cortex-predominant [18F]PI-2620 binding. At the group level, patients with clinical diagnoses associated with FTLD (progressive supranuclear palsy with Richardson syndrome [PSP Richardson syndrome], corticobasal syndrome, and nonfluent-variant primary progressive aphasia) exhibited higher globus pallidus SUVr than did HCs; pallidal retention was highest in the PSP Richardson syndrome group, in whom SUVr was correlated with symptom severity (ρ = 0.53, P = 0.05). At the individual level, only half of PSP Richardson syndrome, corticobasal syndrome, and nonfluent-variant primary progressive aphasia patients had a pallidal SUVr above that of HCs. Temporal SUVr discriminated AD patients from HCs with high accuracy (area under the receiver operating characteristic curve, 0.94 [95% CI, 0.83-1.00]) for all time windows, whereas discrimination between patients with PSP Richardson syndrome and HCs using pallidal SUVr was fair regardless of time window (area under the receiver operating characteristic curve, 0.77 [95% CI, 0.61-0.92] at 30-40 min vs. 0.81 [95% CI, 0.66-0.96] at 50-60 min; P = 0.67). Conclusion: [18F]PI-2620 SUVr shows an intense and consistent signal in AD but lower-intensity, heterogeneous, and rapidly decreasing binding in patients with suspected FTLD. Further work is needed to delineate the substrate of [18F]PI-2620 binding and the usefulness of [18F]PI2620 SUVr quantification outside the AD continuum.
Collapse
Affiliation(s)
- Ganna Blazhenets
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - David N Soleimani-Meigooni
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Wesley Thomas
- Lawrence Berkeley National Laboratory, Berkeley, California
| | - Nidhi Mundada
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Stephanie Vento
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Lawren VandeVrede
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Hilary W Heuer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Peter Ljubenkov
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California
| | - Miranda K Chen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Alinda N Amuiri
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Zachary Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Maria L Gorno-Tempini
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Howie J Rosen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Irene Litvan
- University of California, San Diego, San Diego, California
| | - Murray Grossman
- Penn FTD Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | - David J Irwin
- Penn FTD Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, California;
| |
Collapse
|
50
|
Theis H, Bischof GN, Brüggemann N, Dargvainiene J, Drzezga A, Grüter T, Lewerenz J, Leypoldt F, Neumaier B, Wandinger KP, Ayzenberg I, van Eimeren T. In Vivo Measurement of Tau Depositions in Anti-IgLON5 Disease Using [18F]PI-2620 PET. Neurology 2023; 101:e2325-e2330. [PMID: 37879939 PMCID: PMC10727210 DOI: 10.1212/wnl.0000000000207870] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/22/2023] [Indexed: 10/27/2023] Open
Abstract
OBJECTIVES Anti-IgLON5 disease is a recently discovered neurologic disorder combining autoimmunity and neurodegeneration. Core manifestations include sleep disorders, bulbar symptoms, gait abnormalities, and cognitive dysfunction, but other presentations have been reported. Hallmarks are autoantibodies targeting the neuronal surface protein IgLON5, a strong human leukocyte antigen system Class II association, and brainstem and hypothalamus-dominant tau deposits. The purpose of this cohort study was to visualize tau deposition in vivo with the second-generation tau-PET tracer. METHODS A cohort of 4 patients with anti-IgLON5 disease underwent a dynamic PET scan with [18F]PI-2620. One patient received a follow-up scan. Z-deviation maps and a 2-sample t test in comparison with healthy controls (n = 10) were performed. Antibody titers, neurofilament light chain, and disease duration were correlated with brainstem binding potentials. RESULTS Patients demonstrated increased [18F]PI2620 tau binding potentials in the pons, dorsal medulla, and cerebellum. The longitudinal scan after 28 months showed an increase of tracer uptake in the medulla despite immunotherapy. Higher antibody titers and neurofilament light chain correlated with higher tracer retention. DISCUSSION The results indicate that tau depositions in anti-IgLON5 disease can be visualized with [18F]PI-2620 and might correlate with the extent of disease. For validation, a larger longitudinal study is necessary.
Collapse
Affiliation(s)
- Hendrik Theis
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Gérard N Bischof
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Norbert Brüggemann
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Justina Dargvainiene
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Alexander Drzezga
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Thomas Grüter
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Jan Lewerenz
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Frank Leypoldt
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Bernd Neumaier
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Klaus-Peter Wandinger
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Ilya Ayzenberg
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Thilo van Eimeren
- From the Multimodal Neuroimaging Group (H.T., G.N.B., A.D., T.v.E.), Department of Nuclear Medicine, and Department of Neurology (H.T., T.v.E.), Faculty of Medicine and University Hospital Cologne, University of Cologne; Molecular Organization of the Brain (G.N.B., A.D.), Institute for Neuroscience and Medicine (INM-2), Forschungszentrum J̈lich; Department of Neurology (N.B.), Faculty of Medicine and University Hospital Schleswig Holstein (Lübeck), University of Lübeck; Institute of Clinical Chemistry (J.D., F.L., K.-P.W.), University Hospital Schleswig Holstein, Kiel/Lübeck German Center for Neurodegenerative Diseases (DZNE) (A.D.), Bonn-Cologne; Department of Neurology (T.G., I.A.), Faculty of Medicine and St. Josef-Hospital, Ruhr University Bochum; Department of Neurology (J.L.), Faculty of Medicine and University Hospital Ulm, Ulm University; Department of Neurology (F.L.), Faculty of Medicine and University Hospital Schleswig Holstein, Kiel University; Nuclear Chemistry (B.N.), Institute for Neuroscience and Medicine (INM-5), Forschungszentrum Jülich; and Institute of Radiochemistry and Experimental Molecular Imaging (B.N.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany.
| |
Collapse
|