1
|
Subramanyam P, Palaniswamy SS. 18 F-FDG metabolic abnormalities precede perfusion and atrophic changes in diagnosis of early frontotemporal dementia: study from a tertiary care university hospital. Nucl Med Commun 2025; 46:248-259. [PMID: 39686671 DOI: 10.1097/mnm.0000000000001942] [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: 12/18/2024]
Abstract
OBJECTIVE Diagnosis of early onset dementia is critical for initiating management. Although structural MRI is an established procedure for dementia evaluation, early cases may be missed. Neurodegenerative diseases lead to reductions in glucose consumption and grey matter volume loss. Our primary aim was to establish whether metabolic changes precede perfusion abnormalities in early cases of dementia especially, frontotemporal dementia (FTD). Secondly to study if cerebral atrophy using Pasquier visual rating scales can be used reliably to correlate with hypometabolism in this group of patients. MATERIALS AND METHODS A total of 56 patients (M:F = 39:17) with memory loss as per the DSM-5 diagnostic criteria were clinically and neurologically examined and referred for 18 F-Fluorodeoxyglucose (FDG) PET brain imaging. A few patients who had a prior (recent, <1 week) MR brain underwent brain 18 F-FDG PET-CT, and all others were considered for simultaneous 18 F-FDG PETMR imaging of brain. T2-weighted images were used to report Pasquier rating scales in all our patients as per recommendation. RESULTS Cognitive assessments were analysed along with neuroimaging findings. Highest diagnostic performance was obtained with 18 F-FDG PET for identifying early FTD in our series of patients. Sensitivity, specificity and accuracy of FDG and arterial spin labeling (ASL) using simultaneous PETMR were found to be 96.34%/90.1%/89 : 53.57%/62.12%/78, respectively. Cerebral atrophy rated using Pasquier visual scales showed the lowest diagnostic performance. Our study showed that the earliest phase of cognitive decline was found to be associated with specific patterns of hypometabolism, even in the absence of atrophy, which are currently considered diagnostic biomarkers. CONCLUSION Metabolic derangements indeed precede perfusion changes and cerebral atrophy in the setting of early dementia. Simultaneous FDG PETMR is being recommended as the investigation of choice for the evaluation of early FTD. This pilot study shows that FDG PET outperforms cognitive assessments and anatomical imaging modalities in early dementia evaluation. Although ASL can detect perfusion deficits in dementia, compared with FDG PET, its sensitivity and specificity for discerning early onset dementia from controls remain inferior to FDG PET. Pasquier scales, although easy to implement, cannot replace FDG PET metabolic findings, which start very early in the neuronal disease process.
Collapse
Affiliation(s)
- Padma Subramanyam
- Department of Nuclear Medicine and Molecular Imaging, Amrita Institute of Medical Sciences, Amrita Vishwavidyapeetham, Cochin, India
| | | |
Collapse
|
2
|
Zorzi G, Gazzola G, Rossato F, Bussè C, Camporese G, Cecchin D, Cagnin A. Clinical validity and reproducibility of a visual rating scale for cingulate island sign in a real-world memory clinic: An FDG-PET/MRI study. Eur J Neurol 2025; 32:e70015. [PMID: 39723490 DOI: 10.1111/ene.70015] [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: 07/16/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024]
Abstract
PURPOSE Brain [18F]FDG-PET is a supportive biomarker for cognitive impairment in Lewy bodies disease (LBD) showing reduced occipital metabolism and presence of the cingulate island sign (CIS), a relative preservation of posterior cingulate cortex (PCC) metabolism compared with precuneus and cuneus. We assess validation, clinical utility, and reproducibility of a qualitative visual CIS scale in the differential diagnosis with Alzheimer's disease (AD) in a memory clinic setting. METHODS Sixty-seven patients were studied: 36 LBD, of whom 30 with dementia (DLB) and 6 with mild cognitive impairment (MCI-LB), and 31 AD (20 typical and 11 atypical presentations). They underwent FDG-PET/MRI scans and were followed for at least 24 months. The visual CIS rating scale was scored by a nuclear medicine physician and a neurologist independently. Qualitative CIS scores were validated with ROI-based semiquantitative FDG analysis. RESULTS Mean CIS scores were 1.84 ± 1.69 for LBD and 0.9 ± 1.24 for AD (p = 0.001). With a cutoff CIS score ≥2, sensitivity was 0.56, and specificity 0.81 (accuracy 0.67). Positive CIS in patients with AD was due to atypical presentations. Negative CIS in LBD was due to (i) normal FDG-PET in MCI-LB or (ii) marked hypometabolism of both the PCC and cuneus. Visual CIS scores correlated with FDG-uptake (r = 0.45; p < 0.001) and held a high inter-specialists concordance. DISCUSSION The visual CIS scale can be successfully scored by different specialists. Lower sensitivity is expected in cases of MCI-LB or dementia due to mixed LBD/AD changes. Specificity may be influenced by the inclusion of atypical AD cases.
Collapse
Affiliation(s)
- Giovanni Zorzi
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
- Neurology, Department of Neuroscience, University of Padova, Padova, Italy
- Clinical Center for the Aging Brain, University Hospital of Padova, Padova, Italy
| | - Gianmarco Gazzola
- Neurology, Department of Neuroscience, University of Padova, Padova, Italy
| | - Francesco Rossato
- Neurology, Department of Neuroscience, University of Padova, Padova, Italy
| | - Cinzia Bussè
- Neurology, Department of Neuroscience, University of Padova, Padova, Italy
| | - Giulia Camporese
- Centre for Cognitive Disorders and Dementia (CDCD AULSS6), Padova, Italy
| | - Diego Cecchin
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
- Nuclear Medicine, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Annachiara Cagnin
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
- Neurology, Department of Neuroscience, University of Padova, Padova, Italy
| |
Collapse
|
3
|
Loewenstein DSL, van Grinsven M, de Pont C, Dautzenberg PLJ, van Strien AM, Henssen D. Assessing the metabolism of the olfactory circuit by use of 18F-FDG PET-CT imaging in patients suspected of suffering from Alzheimer's disease or frontotemporal dementia. Alzheimers Res Ther 2024; 16:241. [PMID: 39472983 PMCID: PMC11520854 DOI: 10.1186/s13195-024-01604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024]
Abstract
PURPOSE The loss of olfactory function is known to occur in patients suffering from (behavioral variant) frontotemporal dementia ((bv)FTD) and Alzheimer's disease (AD), although different pathophysiological mechanisms underpin this clinical symptom in both disorders. This study assessed whether brain metabolism of the olfactory circuit as assessed by positron emission tomography (PET) imaging with 2-[fluorine-18]fluoro-2-deoxy-d-glucose ([18F]-FDG) can distinguish these entities in different subsets of patients. METHODS Patients presenting with cognitive decline were included from a prospectively kept database: (1) bvFTD patients, (2) AD patients and (3) patients with logopenic primary progressive aphasia (PPA). Metabolic rates were calculated for different regions of the olfactory circuit for each subgroup and compared with a cohort of subjects with normal brain metabolism. Additionally, in patients with a logopenic PPA pattern on PET-imaging, statistical parametric mapping (SPM) analysis was performed. RESULTS The metabolism of subdivisions of the olfactory circuit as assessed by [18F]-FDG PET brain imaging to bvFTD and AD from control subjects resulted in sensitivity/specificity rates of 95/87.5% and 80/83.3%, respectively. A sensitivity/specificity rate of 100/87.5% was achieved when used to differentiate AD from bvFTD. In patients with the PPA pattern on imaging, the underlying cause (either FTD or AD) could be determined with a sensitivity/specificity rate of 88/82%. SPM analysis concurred that different regions of the olfactory circuit were affected in patients suffering from AD PPA or bvFTD PPA. CONCLUSION Metabolic dysfunction in the olfactory circuit is different in various neurodegenerative disorders. Further investigation of the correlations between the cerebral metabolism and the mechanisms which drive olfactory dysfunction is needed.
Collapse
Affiliation(s)
- Daniël S L Loewenstein
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen, 6525 EZ, The Netherlands.
| | - Max van Grinsven
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen, 6525 EZ, The Netherlands
| | - Cécile de Pont
- Department of Medical Imaging, Jeroen Bosch Hospital, 's Hertogenbosch, The Netherlands
| | - Paul L J Dautzenberg
- Department of Geriatrics, Jeroen Bosch Hospital, 's Hertogenbosch, The Netherlands
| | - Astrid M van Strien
- Department of Geriatrics, Jeroen Bosch Hospital, 's Hertogenbosch, The Netherlands
| | - Dylan Henssen
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, Nijmegen, 6525 EZ, The Netherlands
- Department of Medical Imaging, Jeroen Bosch Hospital, 's Hertogenbosch, The Netherlands
| |
Collapse
|
4
|
Nadaf A, Jiba U, Chaudhary A, Hasan N, Adil M, Mohammed YH, Kesharwani P, jain GK, Ahmad FJ. Gamma scintigraphy in sensing drug delivery systems. NUCLEAR ENGINEERING AND TECHNOLOGY 2024; 56:4423-4436. [DOI: 10.1016/j.net.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Xu F, Dai Z, Zhang W, Ye Y, Dai F, Hu P, Cheng H. Exploring research hotspots and emerging trends in neuroimaging of vascular cognitive impairment: a bibliometric and visualized analysis. Front Aging Neurosci 2024; 16:1408336. [PMID: 39040547 PMCID: PMC11260638 DOI: 10.3389/fnagi.2024.1408336] [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: 03/28/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Background Vascular cognitive impairment (VCI) manifests in memory impairment, mental slowness, executive dysfunction, behavioral changes, and visuospatial abnormalities, significantly compromising the quality of daily life for patients and causing inconvenience to caregivers. Neuroimaging serves as a crucial approach to evaluating the extent, location, and type of vascular lesions in patients suspected of VCI. Nevertheless, there is still a lack of comprehensive bibliometric analysis to discern the research status and emerging trends concerning VCI neuroimaging. Objective This study endeavors to explore the collaboration relationships of authors, countries, and institutions, as well as the research hotspots and frontiers of VCI neuroimaging by conducting a bibliometric analysis. Methods We performed a comprehensive retrieval within the Core Collection of Web of Science, spanning from 2000 to 2023. After screening the included literature, CiteSpace and VOSviewer were utilized for a visualized analysis aimed at identifying the most prolific author, institution, and journal, as well as extracting valuable information from the analysis of references. Results A total of 1,024 publications were included in this study, comprising 919 articles and 105 reviews. Through the analysis of keywords and references, the research hotspots involve the relationship between neuroimaging of cerebral small vessel disease (CSVD) and VCI, the diagnosis of VCI, and neuroimaging methods pertinent to VCI. Moreover, potential future research directions encompass CSVD, functional and structural connectivity, neuroimaging biomarkers, and lacunar stroke. Conclusion The research in VCI neuroimaging is constantly developing, and we hope to provide insights and references for future studies by delving into the research hotspots and frontiers within this field.
Collapse
Affiliation(s)
- Fangyuan Xu
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Ziliang Dai
- Department of Rehabilitation Medicine, The Second Hospital of Wuhan Iron and Steel (Group) Corp., Wuhan, China
| | - Wendong Zhang
- Department of Neurology, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Yu Ye
- The Second Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Fan Dai
- The Second Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Peijia Hu
- Department of Endocrinology, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Hongliang Cheng
- Department of Neurology, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
6
|
Strobel J, Yousefzadeh-Nowshahr E, Deininger K, Bohn KP, von Arnim CAF, Otto M, Solbach C, Anderl-Straub S, Polivka D, Fissler P, Glatting G, Riepe MW, Higuchi M, Beer AJ, Ludolph A, Winter G. Exploratory Tau PET/CT with [11C]PBB3 in Patients with Suspected Alzheimer's Disease and Frontotemporal Lobar Degeneration: A Pilot Study on Correlation with PET Imaging and Cerebrospinal Fluid Biomarkers. Biomedicines 2024; 12:1460. [PMID: 39062033 PMCID: PMC11274645 DOI: 10.3390/biomedicines12071460] [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: 05/08/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Accurately diagnosing Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) is challenging due to overlapping symptoms and limitations of current imaging methods. This study investigates the use of [11C]PBB3 PET/CT imaging to visualize tau pathology and improve diagnostic accuracy. Given diagnostic challenges with symptoms and conventional imaging, [11C]PBB3 PET/CT's potential to enhance accuracy was investigated by correlating tau pathology with cerebrospinal fluid (CSF) biomarkers, positron emission tomography (PET), computed tomography (CT), amyloid-beta, and Mini-Mental State Examination (MMSE). We conducted [11C]PBB3 PET/CT imaging on 24 patients with suspected AD or FTLD, alongside [11C]PiB PET/CT (13 patients) and [18F]FDG PET/CT (15 patients). Visual and quantitative assessments of [11C]PBB3 uptake using standardized uptake value ratios (SUV-Rs) and correlation analyses with clinical assessments were performed. The scans revealed distinct tau accumulation patterns; 13 patients had no or faint uptake (PBB3-negative) and 11 had moderate to pronounced uptake (PBB3-positive). Significant inverse correlations were found between [11C]PBB3 SUV-Rs and MMSE scores, but not with CSF-tau or CSF-amyloid-beta levels. Here, we show that [11C]PBB3 PET/CT imaging can reveal distinct tau accumulation patterns and correlate these with cognitive impairment in neurodegenerative diseases. Our study demonstrates the potential of [11C]PBB3-PET imaging for visualizing tau pathology and assessing disease severity, offering a promising tool for enhancing diagnostic accuracy in AD and FTLD. Further research is essential to validate these findings and refine the use of tau-specific PET imaging in clinical practice, ultimately improving patient care and treatment outcomes.
Collapse
Affiliation(s)
- Joachim Strobel
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Katharina Deininger
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Karl Peter Bohn
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Markus Otto
- Department of Neurology, Halle University, 06120 Halle, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Dörte Polivka
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Patrick Fissler
- Psychiatric Services Thurgau (Academic Teaching Hospital of the University of Konstanz), 8596 Münsterlingen, Switzerland
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Matthias W. Riepe
- Department of Psychiatry and Psychotherapy II, Ulm University, 89075 Ulm, Germany
| | - Makoto Higuchi
- National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Ambros J. Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
7
|
Hernández-Lorenzo L, García-Gutiérrez F, Solbas-Casajús A, Corrochano S, Matías-Guiu JA, Ayala JL. Genetic-based patient stratification in Alzheimer's disease. Sci Rep 2024; 14:9970. [PMID: 38693203 PMCID: PMC11063050 DOI: 10.1038/s41598-024-60707-1] [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: 12/19/2023] [Accepted: 04/26/2024] [Indexed: 05/03/2024] Open
Abstract
Alzheimer's disease (AD) shows a high pathological and symptomatological heterogeneity. To study this heterogeneity, we have developed a patient stratification technique based on one of the most significant risk factors for the development of AD: genetics. We addressed this challenge by including network biology concepts, mapping genetic variants data into a brain-specific protein-protein interaction (PPI) network, and obtaining individualized PPI scores that we then used as input for a clustering technique. We then phenotyped each obtained cluster regarding genetics, sociodemographics, biomarkers, fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging, and neurocognitive assessments. We found three clusters defined mainly by genetic variants found in MAPT, APP, and APOE, considering known variants associated with AD and other neurodegenerative disease genetic architectures. Profiling of these clusters revealed minimal variation in AD symptoms and pathology, suggesting different biological mechanisms may activate the neurodegeneration and pathobiological patterns behind AD and result in similar clinical and pathological presentations, even a shared disease diagnosis. Lastly, our research highlighted MAPT, APP, and APOE as key genes where these genetic distinctions manifest, suggesting them as potential targets for personalized drug development strategies to address each AD subgroup individually.
Collapse
Affiliation(s)
- Laura Hernández-Lorenzo
- Department of Computer Architecture and Automation, Computer Science Faculty, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Fernando García-Gutiérrez
- Department of Computer Architecture and Automation, Computer Science Faculty, Complutense University of Madrid, 28040, Madrid, Spain
| | - Ana Solbas-Casajús
- Department of Computer Architecture and Automation, Computer Science Faculty, Complutense University of Madrid, 28040, Madrid, Spain
| | - Silvia Corrochano
- Department of Neurology, San Carlos Research Institute (IdSSC), Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Jordi A Matías-Guiu
- Department of Neurology, San Carlos Research Institute (IdSSC), Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Jose L Ayala
- Department of Computer Architecture and Automation, Computer Science Faculty, Complutense University of Madrid, 28040, Madrid, Spain
- Instituto de Tecnología del Conocimiento, Universidad Complutense de Madrid, 28040, Madrid, Spain
| |
Collapse
|
8
|
Kang S, Jeon S, Lee YG, Ye BS. Alteration of medial temporal lobe metabolism related to Alzheimer's disease and dementia with lewy bodies. Alzheimers Res Ther 2024; 16:89. [PMID: 38654300 PMCID: PMC11036684 DOI: 10.1186/s13195-024-01429-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/11/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Association of medial temporal lobe (MTL) metabolism with Alzheimer's disease (AD) and dementia with Lewy bodies (DLB) has not been evaluated considering their mixed disease (MD). METHODS 131 patients with AD, 133 with DLB, 122 with MD, and 28 normal controls (NCs) underwent neuropsychological tests, assessments for parkinsonism, cognitive fluctuation (CF), and visual hallucinations (VH), and 18F-fluorodeoxyglucose PET to quantify MTL metabolism in the amygdala, hippocampus, and entorhinal cortex. The effects of AD and DLB on MTL metabolism were evaluated using general linear models (GLMs). Associations between MTL metabolism, cognition, and clinical features were evaluated using GLMs or logistic regression models separately performed for the AD spectrum (NC + AD + MD), DLB spectrum (NC + DLB + MD), and disease groups (AD + DLB + MD). Covariates included age, sex, and education. RESULTS AD was associated with hippocampal/entorhinal hypometabolism, whereas DLB was associated with relative amygdalar/hippocampal hypermetabolism. Relative MTL hypermetabolism was associated with lower attention/visuospatial/executive scores and severe parkinsonism in both the AD and DLB spectra and disease groups. Left hippocampal/entorhinal hypometabolism was associated with lower verbal memory scores, whereas right hippocampal hypometabolism was associated with lower visual memory scores in both the AD spectrum and disease groups. Relative MTL hypermetabolism was associated with an increased risk of CF and VH in the disease group, and relative amygdalar hypermetabolism was associated with an increased risk of VH in the DLB spectrum. CONCLUSIONS Entorhinal-hippocampal hypometabolism and relative amygdala-hippocampal hypermetabolism could be characteristics of AD- and DLB-related neurodegeneration, respectively.
Collapse
Affiliation(s)
- Sungwoo Kang
- Department of Neurology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seun Jeon
- Metabolism-Dementia Research Institute , Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Gun Lee
- Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Republic of Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
9
|
Dodel R, Berg D, Duning T, Kalbe E, Meyer PT, Ramirez A, Storch A, Aarsland D, Jessen F. [Dementia with Lewy bodies: old and new knowledge - Part 1: clinical aspects and diagnostics]. DER NERVENARZT 2024; 95:353-361. [PMID: 38092983 PMCID: PMC11014876 DOI: 10.1007/s00115-023-01576-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/26/2023] [Indexed: 04/13/2024]
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) is the second most common neurodegenerative dementia after Alzheimer's disease. Patients with DLB often have a poor prognosis, with worse outcomes than patients with Alzheimer's disease in terms of important parameters, such as quality of life, caregiver burden, health-related costs, frequency of hospital and nursing home admissions, shorter time to severe dementia, and lower survival. The DLB is frequently misdiagnosed and often undertreated. Therefore, it is critical to diagnose DLB as early as possible to ensure optimal care and treatment. OBJECTIVE The aim of this review article is to summarize the main recent findings on diagnostic tools, epidemiology and genetics of DLB. RESULTS Precise clinical diagnostic criteria exist for DLB that enable an etiologic assignment. Imaging techniques are used as standard in DLB, especially also to exclude non-neurodegenerative causes. In particular, procedures in nuclear medicine have a high diagnostic value. DISCUSSION The diagnosis is primarily based on clinical symptoms, although the development of in vivo neuroimaging and biomarkers is changing the scope of clinical diagnosis as well as research into this devastating disease.
Collapse
Affiliation(s)
- Richard Dodel
- Lehrstuhl für Geriatrie, Universität Duisburg-Essen, Virchowstraße 171, 45147, Essen, Deutschland.
| | - Daniela Berg
- Neurologische Klinik, Universität Kiel, Kiel, Deutschland
| | - Thomas Duning
- Neurologische Klinik, Universität Münster, Münster, Deutschland
| | - Elke Kalbe
- Medizinische Psychologie, Neuropsychologie und Gender Studies & Centrum für Neuropsychologische Diagnostik und Intervention (CeNDI), Universität Köln, Köln, Deutschland
| | - Philipp T Meyer
- Klinik für Nuklearmedizin, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Deutschland
| | - Alfredo Ramirez
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universität Köln, Köln, Deutschland
| | - Alexander Storch
- Klinik für Neurologie, Universität Rostock, Rostock, Deutschland
| | - Dag Aarsland
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Stavanger, Norwegen
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, Großbritannien
| | - Frank Jessen
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universität Köln, Köln, Deutschland
| |
Collapse
|
10
|
Kim MS, Park DG, Shin IJ, An YS, Yoon JH. The Role of Dual-Phase 18 F-FP-CIT PET to Early Diagnosis of Corticobasal Syndrome. Clin Nucl Med 2024; 49:124-130. [PMID: 38015725 DOI: 10.1097/rlu.0000000000004979] [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: 11/30/2023]
Abstract
BACKGROUND Corticobasal syndrome (CBS) is a neurodegeneration characterized by asymmetric parkinsonism, dystonia, myoclonus, and apraxia. In the early stage, CBS presents with asymmetric parkinsonism and cortical symptoms (apraxia and alien hand), and neuroimaging finding is often vague, making early clinical differentiation from idiopathic Parkinson disease (IPD) challenging. This study was performed to delineate the specific patterns of cortical hypoperfusion, dopamine transporter (DAT) uptake using dual-phase FP-CIT PET in discriminating between CBS and IPD at early stage. PATIENTS AND METHODS The study enrolled clinically diagnosed CBS (n = 11) and IPD (n = 22) patients (age and sex matched). All participants underwent dual-phase 18 F-FP-CIT PET, and regional SUV ratio (SUVR) was obtained by semiquantitative analysis. The early perfusion imaging and DAT imaging were compared between groups. RESULTS The regional SUVRs (early phase) of the frontal lobe, thalamus, cingulate, and caudate were significantly lower in patients with CBS, whereas the SUVR of occipital lobe was lower in the IPD group. The CBS group exhibited more prominent asymmetry than the IPD group, particularly in the perirolandic area, superior frontal gyrus, and anterior parietal lobe in early phase PET. Striatal DAT uptake (delayed phase) revealed that the caudate showed lower SUVR and prominent asymmetry in the CBS group, and the caudate-to-putamen ratio (CP ratio) was significantly lower in CBS patients ( P < 0.001). Among the parameters (early and delayed), the CP ratio in DAT exhibited the most powerful discriminative power from receiver operating characteristic curve comparison (area under curve = 0.983). CONCLUSIONS This study demonstrated that the dual-phase FP-CIT PET is useful in differentiating CBS and IPD in the early stage of the disease, and a lower CP ratio of DAT imaging is highly informative for distinguishing between corticobasal degeneration and IPD.
Collapse
Affiliation(s)
| | - Dong Gueu Park
- From the Department of Neurology, Ajou University School of Medicine, Suwon
| | - In Ja Shin
- From the Department of Neurology, Ajou University School of Medicine, Suwon
| | - Young Sil An
- Department of Nuclear Medicine, Ajou University School of Medicine, Suwon, South Korea
| | - Jung Han Yoon
- From the Department of Neurology, Ajou University School of Medicine, Suwon
| |
Collapse
|
11
|
Liu F, Shi Y, Wu Q, Chen H, Wang Y, Cai L, Zhang N. The value of FDG combined with PiB PET in the diagnosis of patients with cognitive impairment in a memory clinic. CNS Neurosci Ther 2024; 30:e14418. [PMID: 37602885 PMCID: PMC10848040 DOI: 10.1111/cns.14418] [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: 06/05/2023] [Revised: 07/12/2023] [Accepted: 08/07/2023] [Indexed: 08/22/2023] Open
Abstract
AIMS To analyze the value of 18 F-fluorodeoxyglucose (FDG) positron emission tomography (PET) combined with amyloid PET in cognitive impairment diagnosis. METHODS A total of 187 patients with dementia or mild cognitive impairment (MCI) who underwent 11 C-Pittsburgh compound B (PiB) and FDG PET scans in a memory clinic were included in the final analysis. RESULTS Amyloid-positive and amyloid-negative dementia patient groups showed a significant difference in the proportion of individuals presenting temporoparietal cortex (p < 0.001) and posterior cingulate/precuneus cortex (p < 0.001) hypometabolism. The sensitivity and specificity of this hypometabolic pattern for identifying amyloid pathology were 72.61% and 77.97%, respectively, in patients clinically diagnosed with AD and 60.87% and 76.19%, respectively, in patients with MCI. The initial diagnosis was changed in 32.17% of patients with dementia after considering both PiB and FDG results. There was a significant difference in both the proportion of patients showing the hypometabolic pattern and PiB positivity between dementia conversion patients and patients with a stable diagnosis of MCI (p < 0.05). CONCLUSION Temporoparietal and posterior cingulate/precuneus cortex hypometabolism on FDG PET suggested amyloid pathology in patients with cognitive impairment and is helpful in diagnostic decision-making and predicting AD dementia conversion from MCI, particularly when combined with amyloid PET.
Collapse
Affiliation(s)
- Fang Liu
- Department of NeurologyTianjin Neurological Institute, Tianjin Medical University General HospitalTianjinChina
| | - Yudi Shi
- Department of NeurologyTianjin Neurological Institute, Tianjin Medical University General HospitalTianjinChina
- Health Management CenterTianjin Medical University General Hospital Airport SiteTianjinChina
| | - Qiuyan Wu
- Department of NeurologyTianjin Neurological Institute, Tianjin Medical University General HospitalTianjinChina
| | - Huifeng Chen
- Department of NeurologyTianjin Neurological Institute, Tianjin Medical University General HospitalTianjinChina
- Department of NeurologyTianjin Medical University General Hospital Airport SiteTianjinChina
| | - Ying Wang
- PET/CT CenterTianjin Medical University General HospitalTianjinChina
| | - Li Cai
- PET/CT CenterTianjin Medical University General HospitalTianjinChina
| | - Nan Zhang
- Department of NeurologyTianjin Neurological Institute, Tianjin Medical University General HospitalTianjinChina
- Department of NeurologyTianjin Medical University General Hospital Airport SiteTianjinChina
| |
Collapse
|
12
|
Corriveau-Lecavalier N, Barnard LR, Przybelski SA, Gogineni V, Botha H, Graff-Radford J, Ramanan VK, Forsberg LK, Fields JA, Machulda MM, Rademakers R, Gavrilova RH, Lapid MI, Boeve BF, Knopman DS, Lowe VJ, Petersen RC, Jack CR, Kantarci K, Jones DT. Assessing network degeneration and phenotypic heterogeneity in genetic frontotemporal lobar degeneration by decoding FDG-PET. Neuroimage Clin 2023; 41:103559. [PMID: 38147792 PMCID: PMC10944211 DOI: 10.1016/j.nicl.2023.103559] [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: 10/16/2023] [Revised: 11/21/2023] [Accepted: 12/19/2023] [Indexed: 12/28/2023]
Abstract
Genetic mutations causative of frontotemporal lobar degeneration (FTLD) are highly predictive of a specific proteinopathy, but there exists substantial inter-individual variability in their patterns of network degeneration and clinical manifestations. We collected clinical and 18Fluorodeoxyglucose-positron emission tomography (FDG-PET) data from 39 patients with genetic FTLD, including 11 carrying the C9orf72 hexanucleotide expansion, 16 carrying a MAPT mutation and 12 carrying a GRN mutation. We performed a spectral covariance decomposition analysis between FDG-PET images to yield unbiased latent patterns reflective of whole brain patterns of metabolism ("eigenbrains" or EBs). We then conducted linear discriminant analyses (LDAs) to perform EB-based predictions of genetic mutation and predominant clinical phenotype (i.e., behavior/personality, language, asymptomatic). Five EBs were significant and explained 58.52 % of the covariance between FDG-PET images. EBs indicative of hypometabolism in left frontotemporal and temporo-parietal areas distinguished GRN mutation carriers from other genetic mutations and were associated with predominant language phenotypes. EBs indicative of hypometabolism in prefrontal and temporopolar areas with a right hemispheric predominance were mostly associated with predominant behavioral phenotypes and distinguished MAPT mutation carriers from other genetic mutations. The LDAs yielded accuracies of 79.5 % and 76.9 % in predicting genetic status and predominant clinical phenotype, respectively. A small number of EBs explained a high proportion of covariance in patterns of network degeneration across FTLD-related genetic mutations. These EBs contained biological information relevant to the variability in the pathophysiological and clinical aspects of genetic FTLD, and for offering valuable guidance in complex clinical decision-making, such as decisions related to genetic testing.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic Rochester, USA; Department of Psychiatry and Psychology, Mayo Clinic Rochester, USA
| | | | | | | | - Hugo Botha
- Department of Neurology, Mayo Clinic Rochester, USA
| | | | | | | | - Julie A Fields
- Department of Psychiatry and Psychology, Mayo Clinic Rochester, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic Rochester, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic Jacksonville, USA; VIB-UA Center for Molecular Neurology, VIB, University of Antwerp, Belgium
| | | | - Maria I Lapid
- Department of Psychiatry and Psychology, Mayo Clinic Rochester, USA
| | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic Rochester, USA
| | | | | | | | - David T Jones
- Department of Neurology, Mayo Clinic Rochester, USA; Department of Radiology, Mayo Clinic Rochester, USA.
| |
Collapse
|
13
|
Silva-Rodríguez J, Labrador-Espinosa MA, Moscoso A, Schöll M, Mir P, Grothe MJ. Characteristics of amnestic patients with hypometabolism patterns suggestive of Lewy body pathology. Brain 2023; 146:4520-4531. [PMID: 37284793 PMCID: PMC10629761 DOI: 10.1093/brain/awad194] [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: 01/25/2023] [Revised: 04/27/2023] [Accepted: 05/16/2023] [Indexed: 06/08/2023] Open
Abstract
A clinical diagnosis of Alzheimer's disease dementia (ADD) encompasses considerable pathological and clinical heterogeneity. While Alzheimer's disease patients typically show a characteristic temporo-parietal pattern of glucose hypometabolism on 18F-fluorodeoxyglucose (FDG)-PET imaging, previous studies have identified a subset of patients showing a distinct posterior-occipital hypometabolism pattern associated with Lewy body pathology. Here, we aimed to improve the understanding of the clinical relevance of these posterior-occipital FDG-PET patterns in patients with Alzheimer's disease-like amnestic presentations. Our study included 1214 patients with clinical diagnoses of ADD (n = 305) or amnestic mild cognitive impairment (aMCI, n = 909) from the Alzheimer's Disease Neuroimaging Initiative, who had FDG-PET scans available. Individual FDG-PET scans were classified as being suggestive of Alzheimer's (AD-like) or Lewy body (LB-like) pathology by using a logistic regression classifier trained on a separate set of patients with autopsy-confirmed Alzheimer's disease or Lewy body pathology. AD- and LB-like subgroups were compared on amyloid-β and tau-PET, domain-specific cognitive profiles (memory versus executive function performance), as well as the presence of hallucinations and their evolution over follow-up (≈6 years for aMCI, ≈3 years for ADD). Around 12% of the aMCI and ADD patients were classified as LB-like. For both aMCI and ADD patients, the LB-like group showed significantly lower regional tau-PET burden than the AD-like subgroup, but amyloid-β load was only significantly lower in the aMCI LB-like subgroup. LB- and AD-like subgroups did not significantly differ in global cognition (aMCI: d = 0.15, P = 0.16; ADD: d = 0.02, P = 0.90), but LB-like patients exhibited a more dysexecutive cognitive profile relative to the memory deficit (aMCI: d = 0.35, P = 0.01; ADD: d = 0.85 P < 0.001), and had a significantly higher risk of developing hallucinations over follow-up [aMCI: hazard ratio = 1.8, 95% confidence interval = (1.29, 3.04), P = 0.02; ADD: hazard ratio = 2.2, 95% confidence interval = (1.53, 4.06) P = 0.01]. In summary, a sizeable group of clinically diagnosed ADD and aMCI patients exhibit posterior-occipital FDG-PET patterns typically associated with Lewy body pathology, and these also show less abnormal Alzheimer's disease biomarkers as well as specific clinical features typically associated with dementia with Lewy bodies.
Collapse
Affiliation(s)
- Jesús Silva-Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Miguel A Labrador-Espinosa
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Alexis Moscoso
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Michael Schöll
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, 41345 Gothenburg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, WC1ELondon, UK
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, 41345 Gothenburg, Sweden
| | | |
Collapse
|
14
|
Li L, Ji B, Zhao M, Bai L, Chen B. Nonfluent Variant Primary Progressive Aphasia on FDG, 11 C-PIB, and 18 F-APN-1607 PET Imaging. Clin Nucl Med 2023; 48:e539-e541. [PMID: 37756439 DOI: 10.1097/rlu.0000000000004853] [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: 09/29/2023]
Abstract
ABSTRACT A 61-year-old right-handed man presented with decreased cognitive function, short-term memory, fluent speech disorders, and grammatical errors for 1 year. The patient underwent PET imaging with 11 C-PIB, 18 F-FDG, and 18 F-APN-1607. The 11 C-PIB PET showed no amyloid accumulation; the 18 F-FDG PET showed hypometabolism in the bilateral frontal lobe, temporal lobe, and midbrain; and the 18 F-APN-1607 PET showed tau accumulation in the brainstem, basal ganglia, and left inferior frontal gyrus. These findings suggested a diagnosis of nonfluent variant primary progressive aphasia. This case emphasizes the value of combined imaging of glucose metabolism, Aβ, and tau PET in the diagnosis of nonfluent variant primary progressive aphasia.
Collapse
Affiliation(s)
- Lingchao Li
- From the Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | | | | | | | | |
Collapse
|
15
|
Chouliaras L, O'Brien JT. The use of neuroimaging techniques in the early and differential diagnosis of dementia. Mol Psychiatry 2023; 28:4084-4097. [PMID: 37608222 PMCID: PMC10827668 DOI: 10.1038/s41380-023-02215-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
Dementia is a leading cause of disability and death worldwide. At present there is no disease modifying treatment for any of the most common types of dementia such as Alzheimer's disease (AD), Vascular dementia, Lewy Body Dementia (LBD) and Frontotemporal dementia (FTD). Early and accurate diagnosis of dementia subtype is critical to improving clinical care and developing better treatments. Structural and molecular imaging has contributed to a better understanding of the pathophysiology of neurodegenerative dementias and is increasingly being adopted into clinical practice for early and accurate diagnosis. In this review we summarise the contribution imaging has made with particular focus on multimodal magnetic resonance imaging (MRI) and positron emission tomography imaging (PET). Structural MRI is widely used in clinical practice and can help exclude reversible causes of memory problems but has relatively low sensitivity for the early and differential diagnosis of dementia subtypes. 18F-fluorodeoxyglucose PET has high sensitivity and specificity for AD and FTD, while PET with ligands for amyloid and tau can improve the differential diagnosis of AD and non-AD dementias, including recognition at prodromal stages. Dopaminergic imaging can assist with the diagnosis of LBD. The lack of a validated tracer for α-synuclein or TAR DNA-binding protein 43 (TDP-43) imaging remain notable gaps, though work is ongoing. Emerging PET tracers such as 11C-UCB-J for synaptic imaging may be sensitive early markers but overall larger longitudinal multi-centre cross diagnostic imaging studies are needed.
Collapse
Affiliation(s)
- Leonidas Chouliaras
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Specialist Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, St Margaret's Hospital, Epping, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
16
|
Meng M, Liu F, Ma Y, Qin W, Guo L, Peng S, Gordon ML, Wang Y, Zhang N. The identification and cognitive correlation of perfusion patterns measured with arterial spin labeling MRI in Alzheimer's disease. Alzheimers Res Ther 2023; 15:75. [PMID: 37038198 PMCID: PMC10088108 DOI: 10.1186/s13195-023-01222-9] [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: 11/24/2021] [Accepted: 03/28/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Vascular dysfunction, including cerebral hypoperfusion, plays an important role in the pathogenesis and progression of Alzheimer's disease (AD), independent of amyloid and tau pathology. We established an AD-related perfusion pattern (ADRP) measured with arterial spin labeling (ASL) MRI using multivariate spatial covariance analysis. METHODS We obtained multimodal MRI including pseudo-continuous ASL and neurocognitive testing in a total of 55 patients with a diagnosis of mild to moderate AD supported by amyloid PET and 46 normal controls (NCs). An ADRP was established from an identification cohort of 32 patients with AD and 32 NCs using a multivariate analysis method based on scaled subprofile model/principal component analysis, and pattern expression in individual subjects was quantified for both the identification cohort and a validation cohort (23 patients with AD and 14 NCs). Subject expression score of the ADRP was then used to assess diagnostic accuracy and cognitive correlations in AD patients and compared with global and regional cerebral blood flow (CBF) in specific areas identified from voxel-based univariate analysis. RESULTS The ADRP featured negative loading in the bilateral middle and posterior cingulate and precuneus, inferior parietal lobule, and frontal areas, and positive loading in the right cerebellum and bilateral basal areas. Subject expression score of the ADRP was significantly elevated in AD patients compared with NCs (P < 0.001) and showed good diagnostic accuracy for AD with area under receiver-operator curve of 0.87 [95% CI (0.78-0.96)] in the identification cohort and 0.85 in the validation cohort. Moreover, there were negative correlations between subject expression score and global cognitive function and performance in various cognitive domains in patients with AD. The characteristics of the ADRP topography and subject expression scores were supported by analogous findings obtained with regional CBF. CONCLUSIONS We have reported a characteristic perfusion pattern associated with AD using ASL MRI. Subject expression score of this spatial covariance pattern is a promising MRI biomarker for the identification and monitoring of AD.
Collapse
Affiliation(s)
- Meng Meng
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| | - Fang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154, Anshan Road, Tianjin, 300052, China
| | - Yilong Ma
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hofstra University, Hempstead, NY, USA
| | - Wen Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Lining Guo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Shichun Peng
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Marc L Gordon
- The Litwin-Zucker Research Center, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Departments of Neurology and Psychiatry, Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hofstra University, Hempstead, NY, USA
| | - Yue Wang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154, Anshan Road, Tianjin, 300052, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China.
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154, Anshan Road, Tianjin, 300052, China.
| |
Collapse
|
17
|
Grothe MJ, Moscoso A, Silva-Rodríguez J, Lange C, Nho K, Saykin AJ, Nelson PT, Schöll M, Buchert R, Teipel S. Differential diagnosis of amnestic dementia patients based on an FDG-PET signature of autopsy-confirmed LATE-NC. Alzheimers Dement 2023; 19:1234-1244. [PMID: 35971593 PMCID: PMC9929029 DOI: 10.1002/alz.12763] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/13/2022] [Accepted: 07/13/2022] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Limbic age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is common in advanced age and can underlie a clinical presentation mimicking Alzheimer's disease (AD). We studied whether an autopsy-derived fluorodeoxyglucose positron emission tomography (FDG-PET) signature of LATE-NC provides clinical utility for differential diagnosis of amnestic dementia patients. METHODS Ante mortem FDG-PET patterns from autopsy-confirmed LATE-NC (N = 7) and AD (N = 23) patients were used to stratify an independent cohort of clinically diagnosed AD dementia patients (N = 242) based on individual FDG-PET profiles. RESULTS Autopsy-confirmed LATE-NC and AD groups showed markedly distinct temporo-limbic and temporo-parietal FDG-PET patterns, respectively. Clinically diagnosed AD dementia patients showing a LATE-NC-like FDG-PET pattern (N = 25, 10%) were significantly older, showed less abnormal AD biomarker levels, lower APOE ε4, and higher TMEM106B risk allele load. Clinically, they exhibited a more memory-predominant profile and a generally slower disease course. DISCUSSION An autopsy-derived temporo-limbic FDG-PET signature identifies older amnestic patients whose clinical, genetic, and molecular biomarker features are consistent with underlying LATE-NC.
Collapse
Affiliation(s)
- Michel J. Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Alexis Moscoso
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Jesús Silva-Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Catharina Lange
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nuclear Medicine, Berlin, Germany
| | - Kwangsik Nho
- Indiana Alzheimer’s Disease Research Center and Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrew J. Saykin
- Indiana Alzheimer’s Disease Research Center and Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Peter T. Nelson
- Sanders-Brown Center on Aging and Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| | - Michael Schöll
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | | |
Collapse
|
18
|
Retinal Neurodegeneration Measured With Optical Coherence Tomography and Neuroimaging in Alzheimer Disease: A Systematic Review. J Neuroophthalmol 2023; 43:116-125. [PMID: 36255105 DOI: 10.1097/wno.0000000000001673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Optical coherence tomography (OCT) has enabled several retinal alterations to be detected in patients with Alzheimer disease (AD), alterations that could be potential biomarkers. However, the relationship between the retina and other biomarkers of AD has been underresearched. We gathered and analyzed the literature about the relationship between retinal and cerebral alterations detected via neuroimaging in patients with AD, mild cognitive impairment (MCI), and preclinical AD. METHODS This systematic review followed the PRISMA Statement guidelines through the 27 items on its checklist. We searched in PubMed, BVS, Scopus, and the Cochrane Library, using the keywords: Alzheimer's disease, optical coherence tomography, white matter, cortex, atrophy, cortical thickness, neuroimaging, magnetic resonance imaging, and positron emission tomography. We included articles that studied the retina in relation to neuroimaging in patients with AD, MCI, and preclinical AD. We excluded studies without OCT, without neuroimaging, clinical cases, opinion articles, systematic reviews, and animal studies. RESULTS Of a total of 35 articles found, 23 were finally included. Although mixed results were found, most of these corroborate the relationship between retinal and brain disorders. CONCLUSIONS More rigorous research is needed in the field, including homogenized, longitudinal, and prolonged follow-up studies, as well as studies that include all stages of AD. This will enable better understanding of the retina and its implications in AD, leading to the discovery of retinal biomarkers that reflect brain alterations in AD patients in an accessible and noninvasive manner.
Collapse
|
19
|
Review of Technological Challenges in Personalised Medicine and Early Diagnosis of Neurodegenerative Disorders. Int J Mol Sci 2023; 24:ijms24043321. [PMID: 36834733 PMCID: PMC9968142 DOI: 10.3390/ijms24043321] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Neurodegenerative disorders are characterised by progressive neuron loss in specific brain areas. The most common are Alzheimer's disease and Parkinson's disease; in both cases, diagnosis is based on clinical tests with limited capability to discriminate between similar neurodegenerative disorders and detect the early stages of the disease. It is common that by the time a patient is diagnosed with the disease, the level of neurodegeneration is already severe. Thus, it is critical to find new diagnostic methods that allow earlier and more accurate disease detection. This study reviews the methods available for the clinical diagnosis of neurodegenerative diseases and potentially interesting new technologies. Neuroimaging techniques are the most widely used in clinical practice, and new techniques such as magnetic resonance imaging (MRI) and positron emission tomography (PET) have significantly improved the diagnosis quality. Identifying biomarkers in peripheral samples such as blood or cerebrospinal fluid is a major focus of the current research on neurodegenerative diseases. The discovery of good markers could allow preventive screening to identify early or asymptomatic stages of the neurodegenerative process. These methods, in combination with artificial intelligence, could contribute to the generation of predictive models that will help clinicians in the early diagnosis, stratification, and prognostic assessment of patients, leading to improvements in patient treatment and quality of life.
Collapse
|
20
|
Ramesh M, Govindaraju T. Multipronged diagnostic and therapeutic strategies for Alzheimer's disease. Chem Sci 2022; 13:13657-13689. [PMID: 36544728 PMCID: PMC9710308 DOI: 10.1039/d2sc03932j] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and a major contributor to dementia cases worldwide. AD is clinically characterized by learning, memory, and cognitive deficits. The accumulation of extracellular amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) of tau are the pathological hallmarks of AD and are explored as targets for clinical diagnosis and therapy. AD pathology is poorly understood and there are no fully approved diagnosis and treatments. Notwithstanding the gap, decades of research in understanding disease mechanisms have revealed the multifactorial nature of AD. As a result, multipronged and holistic approaches are pertinent to targeting multiple biomarkers and targets for developing effective diagnosis and therapeutics. In this perspective, recent developments in Aβ and tau targeted diagnostic and therapeutic tools are discussed. Novel indirect, combination, and circulating biomarkers as potential diagnostic targets are highlighted. We underline the importance of multiplexing and multimodal detection of multiple biomarkers to generate biomarker fingerprints as a reliable diagnostic strategy. The classical therapeutics targeting Aβ and tau aggregation pathways are described with bottlenecks in the strategy. Drug discovery efforts targeting multifaceted toxicity involving protein aggregation, metal toxicity, oxidative stress, mitochondrial damage, and neuroinflammation are highlighted. Recent efforts focused on multipronged strategies to rationally design multifunctional modulators targeting multiple pathological factors are presented as future drug development strategies to discover potential therapeutics for AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| |
Collapse
|
21
|
Hinge C, Henriksen OM, Lindberg U, Hasselbalch SG, Højgaard L, Law I, Andersen FL, Ladefoged CN. A zero-dose synthetic baseline for the personalized analysis of 2-Deoxy-2-[18F]fluoroglucose: Application in Alzheimer’s disease. Front Neurosci 2022; 16:1053783. [DOI: 10.3389/fnins.2022.1053783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
PurposeBrain 2-Deoxy-2-[18F]fluoroglucose ([18F]FDG-PET) is widely used in the diagnostic workup of Alzheimer’s disease (AD). Current tools for uptake analysis rely on non-personalized templates, which poses a challenge as decreased glucose uptake could reflect neuronal dysfunction, or heterogeneous brain morphology associated with normal aging. Overcoming this, we propose a deep learning method for synthesizing a personalized [18F]FDG-PET baseline from the patient’s own MRI, and showcase its applicability in detecting AD pathology.MethodsWe included [18F]FDG-PET/MRI data from 123 patients of a local cohort and 600 patients from ADNI. A supervised, adversarial model with two connected Generative Adversarial Networks (GANs) was trained on cognitive normal (CN) patients with transfer-learning to generate full synthetic baseline volumes (sbPET) (192 × 192 × 192) which reflect healthy uptake conditioned on brain anatomy. Synthetic accuracy was measured by absolute relative %-difference (Abs%), relative %-difference (RD%), and peak signal-to-noise ratio (PSNR). Lastly, we deployed the sbPET images in a fully personalized method for localizing metabolic abnormalities.ResultsThe model achieved a spatially uniform Abs% of 9.4%, RD% of 0.5%, and a PSNR of 26.3 for CN subjects. The sbPET images conformed to the anatomical information dictated by the MRI and proved robust in presence of atrophy. The personalized abnormality method correctly mapped the pathology of AD subjects while showing little to no anomalies for CN subjects.ConclusionThis work demonstrated the feasibility of synthesizing fully personalized, healthy-appearing [18F]FDG-PET images. Using these, we showcased a promising application in diagnosing AD, and theorized the potential value of sbPET images in other neuroimaging routines.
Collapse
|
22
|
Perovnik M, Vo A, Nguyen N, Jamšek J, Rus T, Tang CC, Trošt M, Eidelberg D. Automated differential diagnosis of dementia syndromes using FDG PET and machine learning. Front Aging Neurosci 2022; 14:1005731. [PMID: 36408106 PMCID: PMC9667048 DOI: 10.3389/fnagi.2022.1005731] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/10/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Metabolic brain imaging with 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography (FDG PET) is a supportive diagnostic and differential diagnostic tool for neurodegenerative dementias. In the clinic, scans are usually visually interpreted. However, computer-aided approaches can improve diagnostic accuracy. We aimed to build two machine learning classifiers, based on two sets of FDG PET-derived features, for differential diagnosis of common dementia syndromes. METHODS We analyzed FDG PET scans from three dementia cohorts [63 dementia due to Alzheimer's disease (AD), 79 dementia with Lewy bodies (DLB) and 23 frontotemporal dementia (FTD)], and 41 normal controls (NCs). Patients' clinical diagnosis at follow-up (25 ± 20 months after scanning) or cerebrospinal fluid biomarkers for Alzheimer's disease was considered a gold standard. FDG PET scans were first visually evaluated. Scans were pre-processed, and two sets of features extracted: (1) the expressions of previously identified metabolic brain patterns, and (2) the mean uptake value in 95 regions of interest (ROIs). Two multi-class support vector machine (SVM) classifiers were tested and their diagnostic performance assessed and compared to visual reading. Class-specific regional feature importance was assessed with Shapley Additive Explanations. RESULTS Pattern- and ROI-based classifier achieved higher overall accuracy than expert readers (78% and 80% respectively, vs. 71%). Both SVM classifiers performed similarly to one another and to expert readers in AD (F1 = 0.74, 0.78, and 0.78) and DLB (F1 = 0.81, 0.81, and 0.78). SVM classifiers outperformed expert readers in FTD (F1 = 0.87, 0.83, and 0.63), but not in NC (F1 = 0.71, 0.75, and 0.92). Visualization of the SVM model showed bilateral temporal cortices and cerebellum to be the most important features for AD; occipital cortices, hippocampi and parahippocampi, amygdala, and middle temporal lobes for DLB; bilateral frontal cortices, middle and anterior cingulum for FTD; and bilateral angular gyri, pons, and vermis for NC. CONCLUSION Multi-class SVM classifiers based on the expression of characteristic metabolic brain patterns or ROI glucose uptake, performed better than experts in the differential diagnosis of common dementias using FDG PET scans. Experts performed better in the recognition of normal scans and a combined approach may yield optimal results in the clinical setting.
Collapse
Affiliation(s)
- Matej Perovnik
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,Center for Neurosciences, The Feinstein Institutes for Medical Research, New York, NY, United States,*Correspondence: Matej Perovnik,
| | - An Vo
- Center for Neurosciences, The Feinstein Institutes for Medical Research, New York, NY, United States
| | - Nha Nguyen
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, United States
| | - Jan Jamšek
- Department of Nuclear Medicine, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Tomaž Rus
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Chris C. Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, New York, NY, United States
| | - Maja Trošt
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia,Department of Nuclear Medicine, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, New York, NY, United States
| |
Collapse
|
23
|
Hedderich DM, Schmitz-Koep B, Schuberth M, Schultz V, Schlaeger SJ, Schinz D, Rubbert C, Caspers J, Zimmer C, Grimmer T, Yakushev I. Impact of normative brain volume reports on the diagnosis of neurodegenerative dementia disorders in neuroradiology: A real-world, clinical practice study. Front Aging Neurosci 2022; 14:971863. [PMID: 36313028 PMCID: PMC9597632 DOI: 10.3389/fnagi.2022.971863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Normative brain volume reports (NBVR) are becoming more available in the work-up of patients with suspected dementia disorders, potentially leveraging the value of structural MRI in clinical settings. The present study aims to investigate the impact of NBVRs on the diagnosis of neurodegenerative dementia disorders in real-world clinical practice. Methods: We retrospectively analyzed data of 112 memory clinic patients, who were consecutively referred for MRI and 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) during a 12-month period. Structural MRI was assessed by two residents with 2 and 3 years of neuroimaging experience. Statements and diagnostic confidence regarding the presence of a neurodegenerative disorder in general (first level) and Alzheimer’s disease (AD) pattern in particular (second level) were recorded without and with NBVR information. FDG-PET served as the reference standard. Results: Overall, despite a trend towards increased accuracy, the impact of NBVRs on diagnostic accuracy was low and non-significant. We found a significant drop of sensitivity (0.75–0.58; p < 0.001) and increase of specificity (0.62–0.85; p < 0.001) for rater 1 at identifying patients with neurodegenerative dementia disorders. Diagnostic confidence increased for rater 2 (p < 0.001). Conclusions: Overall, NBVRs had a limited impact on diagnostic accuracy in real-world clinical practice. Potentially, NBVR might increase diagnostic specificity and confidence of neuroradiology residents. To this end, a well-defined framework for integration of NBVR in the diagnostic process and improved algorithms of NBVR generation are essential.
Collapse
Affiliation(s)
- Dennis M. Hedderich
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- *Correspondence: Dennis M. Hedderich
| | - Benita Schmitz-Koep
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Madeleine Schuberth
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Vivian Schultz
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sarah J. Schlaeger
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - David Schinz
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christian Rubbert
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Dusseldorf, Germany
| | - Julian Caspers
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Dusseldorf, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Sch, Munich, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
24
|
Depressive Pseudodementia with Reversible AD-like Brain Hypometabolism: A Case Report and a Review of the Literature. J Pers Med 2022; 12:jpm12101665. [PMID: 36294804 PMCID: PMC9605211 DOI: 10.3390/jpm12101665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
Recent European guidelines recommend using brain FDG-PET to differentiate between Alzheimer's disease (AD) and depressive pseudodementia (DP), with specific hypometabolism patterns across the former group, and typically normal or frontal hypometabolism in the latter. We report the case of a 74 years-old man with DP (MMSE 16/30), whose FDG-PET visual rating and semiquantitative analysis closely mimicked the typical AD pattern, showing severe hypometabolism in bilateral precuneus, parietal and temporal lobes, and sparing frontal areas, suggesting the diagnosis of moderate AD. Shortly after starting antidepressant polytherapy, he underwent formal NPS testing, which revealed moderate impairment of episodic memory and mild impairment on executive and visuospatial tests, judged consistent with neurodegenerative dementia and concomitant depression. Over the following two years, he improved dramatically: repeated NPS assessment did not show significant deficits, and FDG-PET showed restoration of cerebral metabolism. The confirmation of PET findings via semiquantitative analysis, and their reversion to normality with antidepressant treatment, proved the non-neurodegenerative origin of the initial AD-like FDG-PET abnormalities. We review similar cases and provide a comprehensive analysis of their implications, concluding that reversible FDG-PET widespread hypometabolism might represent a biomarker of pseudodementia. Therefore, we suggest caution when interpreting FDG-PET scans of depressed patients with cognitive impairment.
Collapse
|
25
|
Metabolic Imaging in B-Cell Lymphomas during CAR-T Cell Therapy. Cancers (Basel) 2022; 14:cancers14194700. [PMID: 36230629 PMCID: PMC9562671 DOI: 10.3390/cancers14194700] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Chimeric antigen receptor–engineered T cells are an innovative therapy in hematologic malignancies, especially in patients with refractory/relapsed B-cell lymphomas. Few studies have analyzed the role of [18F]FDG PET/CT in this field; this review aims to illustrate the literature data and the major findings related to [18F]FDG PET/CT use during CAR-T cell therapy in B-cell lymphomas, focusing on the prognostic value of metabolic parameters, as well as on response assessment. Furthermore, this work shows in detail the specific adverse events during CAR-T cell therapy and the role of [18F]FDG PET/CT imaging in their occurrence. Abstract Chimeric antigen receptor–engineered (CAR) T cells are emerging powerful therapies for patients with refractory/relapsed B-cell lymphomas. [18F]FDG PET/CT plays a key role during staging and response assessment in patients with lymphoma; however, the evidence about its utility in CAR-T therapies for lymphomas is limited. This review article aims to provide an overview of the role of PET/CT during CAR-T cell therapy in B-cell lymphomas, focusing on the prognostic value of metabolic parameters, as well as on response assessment. Data from the literature report on the use of [18F]FDG PET/CT at the baseline with two scans performed before treatment started focused on the time of decision (TD) PET/CT and time of transfusion (TT) PET/CT. Metabolic tumor burden is the most studied parameter associated with disease progression and overall survival, making us able to predict the occurrence of adverse effects. Instead, for post-therapy evaluation, 1 month (M1) PET/CT seems the preferable time slot for response assessment and in this setting, the Deauville 5-point scale (DS), volumetric analyses, SUVmax, and its variation between different time points (∆SUVmax) have been evaluated, confirming the usefulness of M1 PET/CT, especially in the case of pseudoprogression. Additionally, an emerging role of PET/CT brain scans is reported for the evaluation of neurotoxicity related to CAR-T therapies. Overall, PET/CT results to be an accurate method in all phases of CAR-T treatment, with particular interest in assessing treatment response. Moreover, PET parameters have been reported to be reliable predictors of outcome and severe toxicity.
Collapse
|
26
|
Paoletti M, Caverzasi E, Mandelli ML, Brown JA, Henry RG, Miller BL, Rosen HJ, DeArmond SJ, Bastianello S, Seeley WW, Geschwind MD. Default Mode Network quantitative diffusion and resting-state functional magnetic resonance imaging correlates in sporadic Creutzfeldt-Jakob disease. Hum Brain Mapp 2022; 43:4158-4173. [PMID: 35662331 PMCID: PMC9374887 DOI: 10.1002/hbm.25945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/14/2022] [Accepted: 05/01/2022] [Indexed: 11/25/2022] Open
Abstract
Grey matter involvement is a well-known feature in sporadic Creutzfeldt-Jakob disease (sCJD), yet precise anatomy-based quantification of reduced diffusivity is still not fully understood. Default Mode Network (DMN) areas have been recently demonstrated as selectively involved in sCJD, and functional connectivity has never been investigated in prion diseases. We analyzed the grey matter involvement using a quantitatively multi-parametric MRI approach. Specifically, grey matter mean diffusivity of 37 subjects with sCJD was compared with that of 30 age-matched healthy controls with a group-wise approach. Differences in mean diffusivity were also examined between the cortical (MM(V)1, MM(V)2C, and VV1) and subcortical (VV2 and MV2K) subgroups of sCJD for those with autopsy data available (n = 27, 73%). We also assessed resting-state functional connectivity of both ventral and dorsal components of DMN in a subset of subject with a rs-fMRI dataset available (n = 17). Decreased diffusivity was predominantly present in posterior cortical regions of the DMN, but also outside of the DMN in temporal areas and in a few limbic and frontal areas, in addition to extensive deep nuclei involvement. Both subcortical and cortical sCJD subgroups showed decreased diffusivity subcortically, whereas only the cortical type expressed significantly decreased diffusivity cortically, mainly in parietal, occipital, and medial-inferior temporal cortices bilaterally. Interestingly, we found abnormally increased connectivity in both dorsal and ventral components of the DMN in sCJD subjects compared with healthy controls. The significance and possible utility of functional imaging as a biomarker for tracking disease progression in prion disease needs to be explored further.
Collapse
Affiliation(s)
- Matteo Paoletti
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of NeuroradiologyIRCCS Mondino FoundationPaviaItaly
| | - Eduardo Caverzasi
- Weill Institute for Neurosciences, Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Brain and Behavioral SciencesUniversity of PaviaPaviaItaly
| | - Maria Luisa Mandelli
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Jesse A. Brown
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Roland G. Henry
- Weill Institute for Neurosciences, Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Graduate Group in BioengineeringUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Bruce L. Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Howard J. Rosen
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Stefano Bastianello
- Department of NeuroradiologyIRCCS Mondino FoundationPaviaItaly
- Department of Brain and Behavioral SciencesUniversity of PaviaPaviaItaly
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Michael D. Geschwind
- Memory and Aging Center, Department of Neurology, Weill Institute for NeuroscienceUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
27
|
Perovnik M, Tomše P, Jamšek J, Emeršič A, Tang C, Eidelberg D, Trošt M. Identification and validation of Alzheimer's disease-related metabolic brain pattern in biomarker confirmed Alzheimer's dementia patients. Sci Rep 2022; 12:11752. [PMID: 35817836 PMCID: PMC9273623 DOI: 10.1038/s41598-022-15667-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 06/28/2022] [Indexed: 12/12/2022] Open
Abstract
Metabolic brain biomarkers have been incorporated in various diagnostic guidelines of neurodegenerative diseases, recently. To improve their diagnostic accuracy a biologically and clinically homogeneous sample is needed for their identification. Alzheimer's disease-related pattern (ADRP) has been identified previously in cohorts of clinically diagnosed patients with dementia due to Alzheimer's disease (AD), meaning that its diagnostic accuracy might have been reduced due to common clinical misdiagnosis. In our study, we aimed to identify ADRP in a cohort of AD patients with CSF confirmed diagnosis, validate it in large out-of-sample cohorts and explore its relationship with patients' clinical status. For identification we analyzed 2-[18F]FDG PET brain scans of 20 AD patients and 20 normal controls (NCs). For validation, 2-[18F]FDG PET scans from 261 individuals with AD, behavioral variant of frontotemporal dementia, mild cognitive impairment and NC were analyzed. We identified an ADRP that is characterized by relatively reduced metabolic activity in temporoparietal cortices, posterior cingulate and precuneus which co-varied with relatively increased metabolic activity in the cerebellum. ADRP expression significantly differentiated AD from NC (AUC = 0.95) and other dementia types (AUC = 0.76-0.85) and its expression correlated with clinical measures of global cognition and neuropsychological indices in all cohorts.
Collapse
Affiliation(s)
- Matej Perovnik
- Department of Neurology, University Medical Center Ljubljana, Zaloska cesta 2, 1000, Ljubljana, Slovenia.
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia.
| | - Petra Tomše
- Department of Nuclear Medicine, University Medical Center Ljubljana, Zaloska cesta 2, 1000, Ljubljana, Slovenia
| | - Jan Jamšek
- Department of Nuclear Medicine, University Medical Center Ljubljana, Zaloska cesta 2, 1000, Ljubljana, Slovenia
| | - Andreja Emeršič
- Laboratory for CSF Diagnostics, Department of Neurology, University Medical Center Ljubljana, Zaloska cesta 2, 1000, Ljubljana, Slovenia
| | - Chris Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Maja Trošt
- Department of Neurology, University Medical Center Ljubljana, Zaloska cesta 2, 1000, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
- Department of Nuclear Medicine, University Medical Center Ljubljana, Zaloska cesta 2, 1000, Ljubljana, Slovenia
| |
Collapse
|
28
|
Perovnik M, Tomše P, Jamšek J, Tang C, Eidelberg D, Trošt M. Metabolic brain pattern in dementia with Lewy bodies: Relationship to Alzheimer's disease topography. Neuroimage Clin 2022; 35:103080. [PMID: 35709556 PMCID: PMC9207351 DOI: 10.1016/j.nicl.2022.103080] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/26/2022] [Accepted: 06/05/2022] [Indexed: 10/28/2022]
Abstract
PURPOSE Dementia with Lewy bodies (DLB) is the second most common neurodegenerative dementia, that shares clinical and metabolic similarities with both Alzheimer's and Parkinson's disease. In this study we aimed to identify a DLB-related pattern (DLBRP), study its relationship with other metabolic brain patterns and explore its diagnostic and prognostic value. METHODS A cohort of 79 participants with DLB, 63 with dementia due to Alzheimer's disease (AD) and 41 normal controls (NCs) and their 2-[18F]FDG PET scans were analysed for identification and validation of DLBRP. Voxel-wise correlation and multiple linear regression were used to study the relation between DLBRP and Alzheimer's disease-related pattern (ADRP), Parkinson's disease-related pattern (PDRP) and PD-related cognitive pattern (PDCP). Diagnostic and prognostic value of DLBRP and of modified DLBRP after accounting for ADRP overlap (DLBRP ⊥ ADRP), were explored. RESULTS The newly identified DLBRP shared topographic similarities with ADRP (R2 = 24%) and PDRP (R2 = 37%), but not with PDCP. We could accurately discriminate between DLB and NC (AUC = 0.99) based on DLBRP expression, and between DLB and AD (AUC = 0.87) based on DLBRP ⊥ ADRP expression. DLBRP expression correlated with cognitive impairment, but the correlation was lost after accounting for ADRP overlap. DLBRP and DLBRP ⊥ ADRP correlated with patients' survival time. CONCLUSION DLBRP has proven to be a specific metabolic brain biomarker of DLB, sharing similarities with ADRP and PDRP, but not PDCP. We observed a similar metabolic mechanism underlying cognitive impairment in DLB and AD. DLB-specific metabolic changes were more detrimental for overall survival.
Collapse
Affiliation(s)
- Matej Perovnik
- Department of Neurology, University Medical Center Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia.
| | - Petra Tomše
- Department of Nuclear Medicine, University Medical Center Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia
| | - Jan Jamšek
- Department of Nuclear Medicine, University Medical Center Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia
| | - Chris Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | - Maja Trošt
- Department of Neurology, University Medical Center Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; Department of Nuclear Medicine, University Medical Center Ljubljana, Zaloška cesta 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
29
|
Halhouli O, Zhang Q, Aldridge GM. Caring for patients with cognitive dysfunction, fluctuations and dementia caused by Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:407-434. [PMID: 35248204 DOI: 10.1016/bs.pbr.2022.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cognitive dysfunction is one of the most prevalent non-motor symptoms in patients with Parkinson's disease (PD). While it tends to worsen in the later stages of disease, it can occur at any time, with 15-20% of patients exhibiting cognitive deficits at diagnosis (Aarsland et al., 2010; Goldman and Sieg, 2020). The characteristic features of cognitive dysfunction include impairment in executive function, visuospatial abilities, and attention, which vary in severity from subtle impairment to overt dementia (Martinez-Horta and Kulisevsky, 2019). To complicate matters, cognitive dysfunction is prone to fluctuate in PD patients, impacting diagnosis and the ability to assess progression and decision-making capacity. The diagnosis of cognitive impairment or dementia has a huge impact on patient independence, quality of life, life expectancy and caregiver burden (Corallo et al., 2017; Lawson et al., 2016; Leroi et al., 2012). It is therefore essential that physicians caring for patients with PD provide education, screening and treatment for this aspect of the disease. In this chapter, we provide a practical guide for the assessment and management of various degrees of cognitive dysfunction in patients with PD by approaching the disease at different stages. We address risk factors for cognitive dysfunction, prevention strategies prior to making the diagnosis, available tools for screening. Lastly, we review aspects of care, management and considerations, including decision-making capacity, that occur after the patient has been diagnosed with cognitive dysfunction or dementia.
Collapse
Affiliation(s)
- Oday Halhouli
- University of Iowa, Department of Neurology, Iowa City, IA, United States
| | - Qiang Zhang
- University of Iowa, Department of Neurology, Iowa City, IA, United States
| | | |
Collapse
|
30
|
Díaz-Álvarez J, Matias-Guiu JA, Cabrera-Martín MN, Pytel V, Segovia-Ríos I, García-Gutiérrez F, Hernández-Lorenzo L, Matias-Guiu J, Carreras JL, Ayala JL. Genetic Algorithms for Optimized Diagnosis of Alzheimer's Disease and Frontotemporal Dementia Using Fluorodeoxyglucose Positron Emission Tomography Imaging. Front Aging Neurosci 2022; 13:708932. [PMID: 35185510 PMCID: PMC8851241 DOI: 10.3389/fnagi.2021.708932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Genetic algorithms have a proven capability to explore a large space of solutions, and deal with very large numbers of input features. We hypothesized that the application of these algorithms to 18F-Fluorodeoxyglucose Positron Emission Tomography (FDG-PET) may help in diagnosis of Alzheimer's disease (AD) and Frontotemporal Dementia (FTD) by selecting the most meaningful features and automating diagnosis. We aimed to develop algorithms for the three main issues in the diagnosis: discrimination between patients with AD or FTD and healthy controls (HC), differential diagnosis between behavioral FTD (bvFTD) and AD, and differential diagnosis between primary progressive aphasia (PPA) variants. Genetic algorithms, customized with K-Nearest Neighbor and BayesNet Naives as the fitness function, were developed and compared with Principal Component Analysis (PCA). K-fold cross validation within the same sample and external validation with ADNI-3 samples were performed. External validation was performed for the algorithms distinguishing AD and HC. Our study supports the use of FDG-PET imaging, which allowed a very high accuracy rate for the diagnosis of AD, FTD, and related disorders. Genetic algorithms identified the most meaningful features with the minimum set of features, which may be relevant for automated assessment of brain FDG-PET images. Overall, our study contributes to the development of an automated, and optimized diagnosis of neurodegenerative disorders using brain metabolism.
Collapse
Affiliation(s)
- Josefa Díaz-Álvarez
- Department of Computer Architecture and Communications, Centro Universitario de Mérida, Universidad de Extremadura, Badajoz, Spain
| | - Jordi A. Matias-Guiu
- Department of Neurology, Hospital Clinico San Carlos, San Carlos Research Health Institute (IdISSC), Universidad Complutense, Madrid, Spain
| | - María Nieves Cabrera-Martín
- Department of Nuclear Medicine, Hospital Clinico San Carlos, San Carlos Research Health Institute (IdISSC), Universidad Complutense, Madrid, Spain
| | - Vanesa Pytel
- Department of Neurology, Hospital Clinico San Carlos, San Carlos Research Health Institute (IdISSC), Universidad Complutense, Madrid, Spain
| | - Ignacio Segovia-Ríos
- Department of Computer Architecture and Communications, Centro Universitario de Mérida, Universidad de Extremadura, Badajoz, Spain
| | - Fernando García-Gutiérrez
- Department of Neurology, Hospital Clinico San Carlos, San Carlos Research Health Institute (IdISSC), Universidad Complutense, Madrid, Spain
- Department of Computer Architecture and Automation, Universidad Complutense, Madrid, Spain
| | - Laura Hernández-Lorenzo
- Department of Neurology, Hospital Clinico San Carlos, San Carlos Research Health Institute (IdISSC), Universidad Complutense, Madrid, Spain
- Department of Computer Architecture and Automation, Universidad Complutense, Madrid, Spain
| | - Jorge Matias-Guiu
- Department of Neurology, Hospital Clinico San Carlos, San Carlos Research Health Institute (IdISSC), Universidad Complutense, Madrid, Spain
| | - José Luis Carreras
- Department of Nuclear Medicine, Hospital Clinico San Carlos, San Carlos Research Health Institute (IdISSC), Universidad Complutense, Madrid, Spain
| | - José L. Ayala
- Department of Computer Architecture and Automation, Universidad Complutense, Madrid, Spain
| | | |
Collapse
|
31
|
Bezafibrate Exerts Neuroprotective Effects in a Rat Model of Sporadic Alzheimer’s Disease. Pharmaceuticals (Basel) 2022; 15:ph15020109. [PMID: 35215222 PMCID: PMC8877080 DOI: 10.3390/ph15020109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Bezafibrate, a pan-peroxisome proliferator-activated receptor (PPAR) agonist, reportedly attenuated tau pathology in a transgenic mouse model of primary tauopathy. Since tau pathology is a neuropathological hallmark of Alzheimer’s disease (AD), bezafibrate may be a potential drug for the treatment of AD. However, no study has investigated its effects in AD models. Thus, we aimed to evaluate whether bezafibrate has neuroprotective effects in a sporadic AD model induced by streptozotocin (STZ) intracerebroventricular (ICV) injection. Rats were administered STZ-ICV (3 mg/kg) followed by bezafibrate (50 mg/kg/day, intraperitoneal) for 4 weeks. Behavior tests and positron emission tomography (PET) were performed to evaluate longitudinal changes in cognitive function, tau pathology, and cerebral glucose metabolism. Immunofluorescence staining was performed to assess neuronal survival and microglial accumulation. STZ-ICV administration induced significant cognitive impairment and substantial neuronal loss, tau pathology, glucose hypometabolism, and microgliosis in the cortex and hippocampus, while bezafibrate effectively attenuated these abnormalities. This study demonstrated that bezafibrate has long-lasting neuroprotective effects in a sporadic AD model. Our data indicate that the neuroprotective effects of bezafibrate might be associated with its ability to ameliorate tau pathology, brain glucose hypometabolism, and neuroinflammation. These findings suggest that bezafibrate is a potential multi-target drug candidate for the treatment of AD.
Collapse
|
32
|
Abstract
Positron emission tomography greatly advanced our understanding on the underlying neural mechanisms of movement disorders. PET with flurodeoxyglucose (FDG) is especially useful as it depicts regional metabolic activity level that can predict patients' symptoms. Multivariate pattern analysis has been used to determine and quantify the co-varying brain networks associated with specific clinical traits of neurodegenerative disease. The result is a biomarker, useful for diagnosis, treatments, and follow up studies. Parkinsonian traits and parkinsonisms are associated with specific spatial pattern of metabolic abnormality useful for differential diagnosis. This approach has also been used for monitoring disease progression and novel treatment responses mostly in Parkinson's disease. In this book chapter, we, illustrate and discuss the significance of the brain networks associated with disease and their modification with neuroplastic changes.
Collapse
|
33
|
Guedj E, Varrone A, Boellaard R, Albert NL, Barthel H, van Berckel B, Brendel M, Cecchin D, Ekmekcioglu O, Garibotto V, Lammertsma AA, Law I, Peñuelas I, Semah F, Traub-Weidinger T, van de Giessen E, Van Weehaeghe D, Morbelli S. EANM procedure guidelines for brain PET imaging using [ 18F]FDG, version 3. Eur J Nucl Med Mol Imaging 2021; 49:632-651. [PMID: 34882261 PMCID: PMC8803744 DOI: 10.1007/s00259-021-05603-w] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022]
Abstract
The present procedural guidelines summarize the current views of the EANM Neuro-Imaging Committee (NIC). The purpose of these guidelines is to assist nuclear medicine practitioners in making recommendations, performing, interpreting, and reporting results of [18F]FDG-PET imaging of the brain. The aim is to help achieve a high-quality standard of [18F]FDG brain imaging and to further increase the diagnostic impact of this technique in neurological, neurosurgical, and psychiatric practice. The present document replaces a former version of the guidelines that have been published in 2009. These new guidelines include an update in the light of advances in PET technology such as the introduction of digital PET and hybrid PET/MR systems, advances in individual PET semiquantitative analysis, and current broadening clinical indications (e.g., for encephalitis and brain lymphoma). Further insight has also become available about hyperglycemia effects in patients who undergo brain [18F]FDG-PET. Accordingly, the patient preparation procedure has been updated. Finally, most typical brain patterns of metabolic changes are summarized for neurodegenerative diseases. The present guidelines are specifically intended to present information related to the European practice. The information provided should be taken in the context of local conditions and regulations.
Collapse
Affiliation(s)
- Eric Guedj
- APHM, CNRS, Centrale Marseille, Institut Fresnel, Timone Hospital, CERIMED, Nuclear Medicine Department, Aix Marseille Univ, Marseille, France. .,Service Central de Biophysique et Médecine Nucléaire, Hôpital de la Timone, 264 rue Saint Pierre, 13005, Marseille, France.
| | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Healthcare Services, Stockholm, Sweden
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands.,Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, Leipzig University, Leipzig, Germany
| | - Bart van Berckel
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Matthias Brendel
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany.,German Centre of Neurodegenerative Diseases (DZNE), Site Munich, Bonn, Germany
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - Ozgul Ekmekcioglu
- Sisli Hamidiye Etfal Education and Research Hospital, Nuclear Medicine Dept., University of Health Sciences, Istanbul, Turkey
| | - Valentina Garibotto
- NIMTLab, Faculty of Medicine, Geneva University, Geneva, Switzerland.,Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands.,Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Iván Peñuelas
- Department of Nuclear Medicine, Clinica Universidad de Navarra, IdiSNA, University of Navarra, Pamplona, Spain
| | - Franck Semah
- Nuclear Medicine Department, University Hospital, Lille, France
| | - Tatjana Traub-Weidinger
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands.,Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, Meibergdreef 9, Amsterdam, The Netherlands
| | | | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Nuclear Medicine Unit, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| |
Collapse
|
34
|
Wang Y, Sadaghiani MS, Tian F, Fitzgerald KC, Solnes L, Newsome SD. Brain and Muscle Metabolic Changes by FDG-PET in Stiff Person Syndrome Spectrum Disorders. Front Neurol 2021; 12:692240. [PMID: 34603180 PMCID: PMC8484315 DOI: 10.3389/fneur.2021.692240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/30/2021] [Indexed: 11/30/2022] Open
Abstract
Objective: To report clinical characteristics and fluorodeoxyglucose positron emission tomography (FDG-PET) findings in the brain and muscles of individuals with stiff person syndrome (SPS) spectrum disorders (SPSSDs). Methods: Retrospective cohort study from 1997 to 2018 at Johns Hopkins Hospital identified 170 individuals with SPS or cerebellar ataxia (CA) associated with anti-glutamic acid decarboxylase (anti-GAD)-65 antibodies. Fifty-one underwent FDG-PET, with 50 involving the body and 30 with dedicated brain acquisition. The clinical and immunological profiles were extracted via medical record review. The brain scans were analyzed quantitatively using the NeuroQ software, with comparison with an averaged normal database. The body scans were reviewed qualitatively by a blinded nuclear medicine radiologist. Results: Mean age of symptom onset was 41.5 years (range 12–75 years). Majority were female (68%) and White (64%). Of the patients, 82% had SPS (majority being classic phenotype), and 18% had CA. Three had a paraneoplastic process. Forty-seven had serum anti-GAD, two with anti-amphiphysin, and one with anti-glycine receptor antibodies. Brain metabolic abnormalities were seen in both SPS and CA, with significant differences between the groups noted in the right superior frontal cortex, right sensorimotor cortex, left inferior parietal cortex, bilateral thalami, vermis, and left cerebellum. Of the patients, 62% demonstrated muscle hypermetabolism, most commonly bilateral, involving the upper extremities or axial muscles. Neither brain nor muscle metabolism was correlated with functional outcomes nor treatments. Conclusions: Metabolic changes as seen by FDG-PET are present in the brain and muscle in many individuals with SPSSD. Future studies are needed to assess whether FDG-PET can help aid in the diagnosis and/or monitoring of individuals with SPSSD.
Collapse
Affiliation(s)
- Yujie Wang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mohammad S Sadaghiani
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Fan Tian
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lilja Solnes
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Scott D Newsome
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
35
|
Chen MK, Mecca AP, Naganawa M, Gallezot JD, Toyonaga T, Mondal J, Finnema SJ, Lin SF, O’Dell RS, McDonald JW, Michalak HR, Vander Wyk B, Nabulsi NB, Huang Y, Arnsten AFT, van Dyck CH, Carson RE. Comparison of [ 11C]UCB-J and [ 18F]FDG PET in Alzheimer's disease: A tracer kinetic modeling study. J Cereb Blood Flow Metab 2021; 41:2395-2409. [PMID: 33757318 PMCID: PMC8393289 DOI: 10.1177/0271678x211004312] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/29/2021] [Accepted: 02/21/2021] [Indexed: 11/16/2022]
Abstract
[11C]UCB-J PET for synaptic vesicle glycoprotein 2 A (SV2A) has been proposed as a suitable marker for synaptic density in Alzheimer's disease (AD). We compared [11C]UCB-J binding for synaptic density and [18F]FDG uptake for metabolism (correlated with neuronal activity) in 14 AD and 11 cognitively normal (CN) participants. We assessed both absolute and relative outcome measures in brain regions of interest, i.e., K1 or R1 for [11C]UCB-J perfusion, VT (volume of distribution) or DVR to cerebellum for [11C]UCB-J binding to SV2A; and Ki or KiR to cerebellum for [18F]FDG metabolism. [11C]UCB-J binding and [18F]FDG metabolism showed a similar magnitude of reduction in the medial temporal lobe of AD -compared to CN participants. However, the magnitude of reduction of [11C]UCB-J binding in neocortical regions was less than that observed with [18F]FDG metabolism. Inter-tracer correlations were also higher in the medial temporal regions between synaptic density and metabolism, with lower correlations in neocortical regions. [11C]UCB-J perfusion showed a similar pattern to [18F]FDG metabolism, with high inter-tracer regional correlations. In summary, we conducted the first in vivo PET imaging of synaptic density and metabolism in the same AD participants and reported a concordant reduction in medial temporal regions but a discordant reduction in neocortical regions.
Collapse
Affiliation(s)
- Ming-Kai Chen
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Adam P Mecca
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Jean-Dominique Gallezot
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Jayanta Mondal
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Sjoerd J Finnema
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Shu-fei Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Ryan S O’Dell
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Julia W McDonald
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Hannah R Michalak
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Brent Vander Wyk
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Amy FT Arnsten
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
36
|
Ferrando R, Damian A. Brain SPECT as a Biomarker of Neurodegeneration in Dementia in the Era of Molecular Imaging: Still a Valid Option? Front Neurol 2021; 12:629442. [PMID: 34040574 PMCID: PMC8141564 DOI: 10.3389/fneur.2021.629442] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/06/2021] [Indexed: 12/21/2022] Open
Abstract
Biomarkers are playing a progressively leading role in both clinical practice and scientific research in dementia. Although amyloid and tau biomarkers have gained ground in the clinical community in recent years, neurodegeneration biomarkers continue to play a key role due to their ability to identify different patterns of brain involvement that sign the transition between asymptomatic and symptomatic stages of the disease with high sensitivity and specificity. Both 18F-FDG positron emission tomography (PET) and perfusion single photon emission computed tomography (SPECT) have proved useful to reveal the functional alterations underlying various neurodegenerative diseases. Although the focus of nuclear neuroimaging has shifted to PET, the lower cost and wider availability of SPECT make it a still valid alternative for the study of patients with dementia. This review discusses the principles of both techniques, compares their diagnostic performance for the diagnosis of neurodegenerative diseases and highlights the role of SPECT to characterize patients from low- and middle-income countries, where special care of additional costs is particularly needed to meet the new recommendations for the diagnosis and characterization of patients with dementia.
Collapse
Affiliation(s)
- Rodolfo Ferrando
- Centro de Medicina Nuclear e Imagenología Molecular, Hospital de Clínicas, Universidad de la República (UdelaR), Montevideo, Uruguay.,Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Andres Damian
- Centro de Medicina Nuclear e Imagenología Molecular, Hospital de Clínicas, Universidad de la República (UdelaR), Montevideo, Uruguay.,Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| |
Collapse
|
37
|
Traini E, Carotenuto A, Fasanaro AM, Amenta F. Volume Analysis of Brain Cognitive Areas in Alzheimer's Disease: Interim 3-Year Results from the ASCOMALVA Trial. J Alzheimers Dis 2021; 76:317-329. [PMID: 32508323 PMCID: PMC7369051 DOI: 10.3233/jad-190623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Cerebral atrophy is a common feature of several neurodegenerative disorders, including Alzheimer’s disease (AD). In AD, brain atrophy is associated with loss of gyri and sulci in the temporal and parietal lobes, and in parts of the frontal cortex and cingulate gyrus. Objective: The ASCOMALVA trial has assessed, in addition to neuropsychological analysis, whether the addition of the cholinergic precursor choline alphoscerate to treatment with donepezil has an effect on brain volume loss in patients affected by AD associated with cerebrovascular injury. Methods: 56 participants to the randomized, placebo-controlled, double-blind ASCOMALVA trial were assigned to donepezil + placebo (D + P) or donepezil + choline alphoscerate (D + CA) treatments and underwent brain magnetic resonance imaging and neuropsychological tests every year for 4 years. An interim analysis of 3-year MRI data was performed by voxel morphometry techniques. Results: The D + P group (n = 27) developed atrophy of the gray and white matter with concomitant increase in ventricular space volume. In the D + CA group (n = 29) the gray matter atrophy was less pronounced compared to the D + P group in frontal and temporal lobes, hippocampus, and amygdala. These morphological data are consistent with the results of the neuropsychological tests. Conclusion: Our findings indicate that the addition of choline alphoscerate to standard treatment with the cholinesterase inhibitor donepezil counters to some extent the loss in volume occurring in some brain areas of AD patients. The observation of parallel less pronounced decrease in cognitive and functional tests in patients with the same treatment suggests that the morphological changes observed may have functional relevance.
Collapse
Affiliation(s)
- Enea Traini
- Clinical Research, Telemedicine and Telepharmacy Centre, University of Camerino, Camerino, Italy
| | - Anna Carotenuto
- Clinical Research, Telemedicine and Telepharmacy Centre, University of Camerino, Camerino, Italy.,Neurology Unit, National Hospital "A. Cardarelli", Naples, Italy
| | | | - Francesco Amenta
- Clinical Research, Telemedicine and Telepharmacy Centre, University of Camerino, Camerino, Italy
| |
Collapse
|
38
|
Frantellizzi V, Pani A, Ricci M, Locuratolo N, Fattapposta F, De Vincentis G. Neuroimaging in Vascular Cognitive Impairment and Dementia: A Systematic Review. J Alzheimers Dis 2021; 73:1279-1294. [PMID: 31929166 DOI: 10.3233/jad-191046] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cerebrovascular diseases are well established causes of cognitive impairment. Different etiologic entities, such as vascular dementia (VaD), vascular cognitive impairment, subcortical (ischemic) VaD, and vascular cognitive disorder, are included in the umbrella definition of vascular cognitive impairment and dementia (VCID). Because of the variability of VCID clinical presentation, there is no agreement on criteria defining the neuropathological threshold of this disorder. In fact, VCID is characterized by cerebral hemodynamic alteration which ranges from decreased cerebral blood flow to small vessels disease and involves a multifactorial process that leads to demyelination and gliosis, including blood-brain barrier disruption, hypoxia, and hypoperfusion, oxidative stress, neuroinflammation and alteration on neurovascular unit coupling, cerebral microbleeds, or superficial siderosis. Numerous criteria for the definition of VaD have been described: the National Institute of Neurological Disorders and Stroke Association Internationale pour Recherche'-et-l'Enseignement en Neurosciences criteria, the State of California Alzheimer's Disease Diagnostic and Treatment Centers criteria, DSM-V criteria, the Diagnostic Criteria for Vascular Cognitive Disorders (a VASCOG Statement), and Vascular Impairment of Cognition Classification Consensus Study. Neuroimaging is fundamental for definition and diagnosis of VCID and should be used to assess the extent, location, and type of vascular lesions. MRI is the most sensible technique, especially if used according to standardized protocols, even if CT plays an important role in several conditions. Functional neuroimaging, in particular functional MRI and PET, may facilitate differential diagnosis among different forms of dementia. This systematic review aims to explore the state of the art and future perspective of non-invasive diagnostics of VCID.
Collapse
Affiliation(s)
| | - Arianna Pani
- Clinical Pharmacology and Toxicology, University of Milan "Statale", Italy
| | - Maria Ricci
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | | | | | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
39
|
Sergienko VB, Ansheles AA. Nuclear medicine and molecular imaging in clinical practice: yesterday, today and tomorrow. TERAPEVT ARKH 2021; 93:357-362. [DOI: 10.26442/00403660.2021.04.200673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 05/31/2021] [Indexed: 11/22/2022]
Abstract
Over the past 40 years, nuclear medicine has grown to be the largest non-invasive diagnostic and therapeutic industry in the world, playing a pivotal role in various fields and disciplines of clinical practice and contributing to improved quality of life and patient prognosis. Over the first 20 years of the XXI century, the number of radionuclide procedures in the world has increased significantly, primarily due to innovations in radiopharmaceuticals, continuous improvement of the technical properties of equipment and the expansion of the boundaries of multimodal imaging. The review examines the historical and current trends in the development of nuclear medicine in the world and in Russia, including those related to radionuclide diagnostics, therapy and theranostics.
Collapse
|
40
|
Borders C, Sajjadi SA. Diagnosis and Management of Cognitive Concerns in the Oldest-Old. Curr Treat Options Neurol 2021; 23:10. [PMID: 33786000 PMCID: PMC7994350 DOI: 10.1007/s11940-021-00665-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 11/30/2022]
Abstract
Purpose of review The fastest-growing group of elderly individuals is the "oldest-old," usually defined as those age 85 years and above. These individuals account for much of the rapid increase in cases of dementing illness throughout the world but remain underrepresented in the body of literature on this topic. The aim of this review is first to outline the unique contributing factors and complications that must be considered by clinicians in evaluating an oldest-old individual with cognitive complaints. Secondly, the evidence for management of these cognitive concerns is reviewed. Recent findings In addition to well-established associations between impaired cognition and physical disability, falls, and frailty, there is now evidence that exercise performed decades earlier confers a cognitive benefit in the oldest-old. Moreover, though aggressive blood pressure control is critical earlier in life for prevention of strokes, renal disease, and other comorbidities, hypertension started after age 80 is in fact associated with a decreased risk of clinical dementia, carrying significant implications for the medical management of oldest-old individuals. The oldest-old are more likely to reside in care facilities, where social isolation might be exacerbated by a consistently lower rate of internet-connected device use. The COVID-19 pandemic has not only highlighted the increased mortality rate among the oldest-old but has also brought the increased social isolation in this group to the forte. Summary Differing from the "younger-old" in a number of respects, the oldest-old is a unique population not just in their vulnerability to cognitive disorders but also in the diagnostic challenges they can pose. The oldest-old are more likely to be afflicted by sensory deficits, physical disability, poor nutrition, frailty, and depression, which must be accounted for in the assessment of cognitive complaints as they may confound or complicate the presentation. Social isolation and institutionalization are also associated with impaired cognition, perhaps as sequelae, precipitants, or both. Ante-mortem diagnostic tools remain particularly limited among the oldest-old, especially given the likelihood of these individuals to have multiple co-occurring types of neuropathology, and the presence of neuropathology in those who remain cognitively intact. In addition to the symptomatic treatments indicated for patients of all ages with dementia, management of cognitive impairment in the oldest-old may be further optimized by use of assistive devices, augmentation of dietary protein, and liberalization of medication regimens for risk factors such as hypertension.
Collapse
Affiliation(s)
- Candace Borders
- Department of Neurology, University of California, Irvine, CA USA
| | | |
Collapse
|
41
|
Gjerum L, Andersen BB, Bruun M, Simonsen AH, Henriksen OM, Law I, Hasselbalch SG, Frederiksen KS. Comparison of the clinical impact of 2-[18F]FDG-PET and cerebrospinal fluid biomarkers in patients suspected of Alzheimer's disease. PLoS One 2021; 16:e0248413. [PMID: 33711065 PMCID: PMC7954298 DOI: 10.1371/journal.pone.0248413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/26/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The two biomarkers 2-[18F]FDG-PET and cerebrospinal fluid biomarkers are both recommended to support the diagnosis of Alzheimer's disease. However, there is a lack of knowledge for the comparison of the two biomarkers in a routine clinical setting. OBJECTIVE The aim was to compare the clinical impact of 2-[18F]FDG-PET and cerebrospinal fluid biomarkers on diagnosis, prognosis, and patient management in patients suspected of Alzheimer's disease. METHODS Eighty-one patients clinically suspected of Alzheimer's disease were retrospectively included from the Copenhagen Memory Clinic. As part of the clinical work-up all patients had a standard diagnostic program examination including MRI and ancillary investigations with 2-[18F]FDG-PET and cerebrospinal fluid biomarkers. An incremental study design was used to evaluate the clinical impact of the biomarkers. First, the diagnostic evaluation was based on the standard diagnostic program, then the diagnostic evaluation was revised after addition of either cerebrospinal fluid biomarkers or 2-[18F]FDG-PET. At each diagnostic evaluation, two blinded dementia specialists made a consensus decision on diagnosis, prediction of disease course, and change in patient management. Confidence in the decision was measured on a visual analogue scale (0-100). After 6 months, the diagnostic evaluation was performed with addition of the other biomarker. A clinical follow-up after 12 months was used as reference for diagnosis and disease course. RESULTS The two biomarkers had a similar clinical value across all diagnosis when added individually to the standard diagnostic program. However, for the correctly diagnosed patient with Alzheimer's disease cerebrospinal fluid biomarkers had a significantly higher impact on diagnostic confidence (mean scores±SD: 88±11 vs. 82±11, p = 0.046) and a significant reduction in the need for ancillary investigations (23 vs. 18 patients, p = 0.049) compared to 2-[18F]FDG-PET. CONCLUSION The two biomarkers had similar clinical impact on diagnosis, but cerebrospinal fluid biomarkers had a more significant value in corroborating the diagnosis of Alzheimer's disease compared to 2-[18F]FDG-PET.
Collapse
Affiliation(s)
- Le Gjerum
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Bo Andersen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Marie Bruun
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anja Hviid Simonsen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Otto Mølby Henriksen
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Steen Frederiksen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Li Q, Hou W, Li L, Su M, Ren Y, Wang W, Zou K, Tian R, Sun X. The use of systematic review evidence to support the development of guidelines for positron emission tomography: a cross-sectional survey. Eur Radiol 2021; 31:6992-7002. [PMID: 33683391 DOI: 10.1007/s00330-021-07756-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/06/2021] [Accepted: 02/04/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To examine to what degree guidelines for PET and PET/CT used systematic review evidence. METHODS The latest version of guidelines for PET, PET/CT or PET/MRI published in English in PubMed until December 2019 was analysed in two categories: (1) for indications, if mainly discussing the appropriate use of PET in diverse conditions; (2) for procedures, if providing step-by-step instructions for imaging. We surveyed the general characteristics and the use of systematic review evidence for developing recommendations across all guidelines, and surveyed the citation of evidence for five recommendation topics in guidelines for procedures. RESULTS Forty-seven guidelines, published between 2004 and 2020, were included. Guidelines for indications were developed mainly on systematic reviews (13 of 19, 68.4%). Among those, 12 (63.2%) reported the level of evidence, 4 (21.1%) reported the strength of recommendations, 3 (15.8%) described external review and 7 (36.8%) involved methodologists. Guidelines for procedures were seldom developed on systematic reviews (1 of 27, 3.7%). Among those, 1 (3.7%) reported the level of evidence, 1 (3.7%) reported the strength of recommendations, 3 (11.1%) described external review and 1 (3.7%) involved methodologists. Systematic review evidence was cited by 2 (7.4%) procedure guidelines per recommendation topic in median. CONCLUSION The use of systematic review evidence for developing recommendations among PET or PET/CT guidelines was suboptimal. While our survey is an icebreaking attempt to explore a key element (i.e. use of systematic review evidence) for developing nuclear medicine guidelines, assessments of other domains of guideline quality may help capture the entire picture. KEY POINTS • The use of systematic review evidence for developing recommendations among guidelines for PET or PET/CT was suboptimal. • Only 13 (68.4%) guidelines for indications and 1 (3.7%) guideline for procedures systematically reviewed the literature during guideline development. • For each recommendation topic we examined, only a median of 2 (7.4%) procedure guidelines cited systematic review evidence.
Collapse
Affiliation(s)
- Qianrui Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Chinese Evidence-based Medicine Centre, Cochrane China Centre and MAGIC China Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenxiu Hou
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ling Li
- Chinese Evidence-based Medicine Centre, Cochrane China Centre and MAGIC China Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Minggang Su
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Ren
- Chinese Evidence-based Medicine Centre, Cochrane China Centre and MAGIC China Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen Wang
- Chinese Evidence-based Medicine Centre, Cochrane China Centre and MAGIC China Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kang Zou
- Chinese Evidence-based Medicine Centre, Cochrane China Centre and MAGIC China Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Xin Sun
- Chinese Evidence-based Medicine Centre, Cochrane China Centre and MAGIC China Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
43
|
Verdelho A, Wardlaw J, Pavlovic A, Pantoni L, Godefroy O, Duering M, Charidimou A, Chabriat H, Biessels GJ. Cognitive impairment in patients with cerebrovascular disease: A white paper from the links between stroke ESO Dementia Committee. Eur Stroke J 2021; 6:5-17. [PMID: 33817330 PMCID: PMC7995319 DOI: 10.1177/23969873211000258] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Many daily-life clinical decisions in patients with cerebrovascular disease and cognitive impairment are complex. Evidence-based information sustaining these decisions is frequently lacking. The aim of this paper is to propose a practical clinical approach to cognitive impairments in patients with known cerebrovascular disease. METHODS The document was produced by the Dementia Committee of the European Stroke Organisation (ESO), based on evidence from the literature where available and on the clinical experience of the Committee members. This paper was endorsed by the ESO. FINDINGS Many patients with stroke or other cerebrovascular disease have cognitive impairment, but this is often not recognized. With improvement in acute stroke care, and with the ageing of populations, it is expected that more stroke survivors and more patients with cerebrovascular disease will need adequate management of cognitive impairment of vascular etiology. This document was conceived for the use of strokologists and for those clinicians involved in cerebrovascular disease, with specific and practical hints concerning diagnostic tools, cognitive impairment management and decision on some therapeutic options.Discussion and conclusions: It is essential to consider a possible cognitive deterioration in every patient who experiences a stroke. Neuropsychological evaluation should be adapted to the clinical status. Brain imaging is the most informative biomarker concerning prognosis. Treatment should always include adequate secondary prevention.
Collapse
Affiliation(s)
- Ana Verdelho
- Department of Neurosciences and Mental Health, CHLN-Hospital de Santa Maria, Instituto de Medicina Molecular – IMM e Instituto de Saúde Ambiental –ISAMB, Faculdade de Medicina, University of Lisbon, Lisbon, Portugal
| | - Joanna Wardlaw
- Centre for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Aleksandra Pavlovic
- Faculty for Special Education and Rehabilitation, University of Belgrade, Belgrade, Serbia
| | - Leonardo Pantoni
- Stroke and Dementia Lab, "Luigi Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Olivier Godefroy
- Department of Neurology, Amiens University Hospital, Laboratory of Functional Neurosciences1,6 (UR UPJV 4559), Jules Verne Picardy University, Amiens, France
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Germany
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Andreas Charidimou
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Hugues Chabriat
- Department of Neurology, FHU NeuroVasc, Hôpital Lariboisiere, University of Paris, Paris, France
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, the Netherlands
| |
Collapse
|
44
|
Levin F, Ferreira D, Lange C, Dyrba M, Westman E, Buchert R, Teipel SJ, Grothe MJ. Data-driven FDG-PET subtypes of Alzheimer's disease-related neurodegeneration. Alzheimers Res Ther 2021; 13:49. [PMID: 33608059 PMCID: PMC7896407 DOI: 10.1186/s13195-021-00785-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/03/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous research has described distinct subtypes of Alzheimer's disease (AD) based on the differences in regional patterns of brain atrophy on MRI. We conducted a data-driven exploration of distinct AD neurodegeneration subtypes using FDG-PET as a sensitive molecular imaging marker of neurodegenerative processes. METHODS Hierarchical clustering of voxel-wise FDG-PET data from 177 amyloid-positive patients with AD dementia enrolled in the Alzheimer's Disease Neuroimaging Initiative (ADNI) was used to identify distinct hypometabolic subtypes of AD, which were then further characterized with respect to clinical and biomarker characteristics. We then classified FDG-PET scans of 217 amyloid-positive patients with mild cognitive impairment ("prodromal AD") according to the identified subtypes and studied their domain-specific cognitive trajectories and progression to dementia over a follow-up interval of up to 72 months. RESULTS Three main hypometabolic subtypes were identified: (i) "typical" (48.6%), showing a classic posterior temporo-parietal hypometabolic pattern; (ii) "limbic-predominant" (44.6%), characterized by old age and a memory-predominant cognitive profile; and (iii) a relatively rare "cortical-predominant" subtype (6.8%) characterized by younger age and more severe executive dysfunction. Subtypes classified in the prodromal AD sample demonstrated similar subtype characteristics as in the AD dementia sample and further showed differential courses of cognitive decline. CONCLUSIONS These findings complement recent research efforts on MRI-based identification of distinct AD atrophy subtypes and may provide a potentially more sensitive molecular imaging tool for early detection and characterization of AD-related neurodegeneration variants at prodromal disease stages.
Collapse
Affiliation(s)
- Fedor Levin
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Rostock, Germany
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Lange
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Martin Dyrba
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Rostock, Germany
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ralph Buchert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Rostock, Germany.
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot, s/n, 41013, Sevilla, Spain.
| |
Collapse
|
45
|
Outcomes of clinical utility in amyloid-PET studies: state of art and future perspectives. Eur J Nucl Med Mol Imaging 2021; 48:2157-2168. [PMID: 33594474 PMCID: PMC8175294 DOI: 10.1007/s00259-020-05187-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/28/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE To review how outcomes of clinical utility are operationalized in current amyloid-PET validation studies, to prepare for formal assessment of clinical utility of amyloid-PET-based diagnosis. METHODS Systematic review of amyloid-PET research studies published up to April 2020 that included outcomes of clinical utility. We extracted and analyzed (a) outcome categories, (b) their definition, and (c) their methods of assessment. RESULTS Thirty-two studies were eligible. (a) Outcome categories were clinician-centered (found in 25/32 studies, 78%), patient-/caregiver-centered (in 9/32 studies, 28%), and health economics-centered (5/32, 16%). (b) Definition: Outcomes were mainly defined by clinical researchers; only the ABIDE study expressly included stakeholders in group discussions. Clinician-centered outcomes mainly consisted of incremental diagnostic value (25/32, 78%) and change in patient management (17/32, 53%); patient-/caregiver-centered outcomes considered distress after amyloid-pet-based diagnosis disclosure (8/32, 25%), including quantified burden of procedure for patients' outcomes (n = 8) (1/8, 12.5%), impact of disclosure of results (6/8, 75%), and psychological implications of biomarker-based diagnosis (75%); and health economics outcomes focused on costs to achieve a high-confidence etiological diagnosis (5/32, 16%) and impact on quality of life (1/32, 3%). (c) Assessment: all outcome categories were operationalized inconsistently across studies, employing 26 different tools without formal rationale for selection. CONCLUSION Current studies validating amyloid-PET already assessed outcomes for clinical utility, although non-clinician-based outcomes were inconsistent. A wider participation of stakeholders may help produce a more thorough and systematic definition and assessment of outcomes of clinical utility and help collect evidence informing decisions on reimbursement of amyloid-PET.
Collapse
|
46
|
Bauckneht M, Chincarini A, Brendel M, Rominger A, Beyer L, Bruffaerts R, Vandenberghe R, Kramberger MG, Trost M, Garibotto V, Nicastro N, Frisoni GB, Lemstra AW, van Berckel BNM, Pilotto A, Padovani A, Ochoa-Figueroa MA, Davidsson A, Camacho V, Peira E, Arnaldi D, Pardini M, Donegani MI, Raffa S, Miceli A, Sambuceti G, Aarsland D, Nobili F, Morbelli S. Associations among education, age, and the dementia with Lewy bodies (DLB) metabolic pattern: A European-DLB consortium project. Alzheimers Dement 2021; 17:1277-1286. [PMID: 33528089 DOI: 10.1002/alz.12294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION We assessed the influence of education as a proxy of cognitive reserve and age on the dementia with Lewy bodies (DLB) metabolic pattern. METHODS Brain 18F-fluorodeoxyglucose positron emission tomography and clinical/demographic information were available in 169 probable DLB patients included in the European DLB-consortium database. Principal component analysis identified brain regions relevant to local data variance. A linear regression model was applied to generate age- and education-sensitive maps corrected for Mini-Mental State Examination score, sex (and either education or age). RESULTS Age negatively covaried with metabolism in bilateral middle and superior frontal cortex, anterior and posterior cingulate, reducing the expression of the DLB-typical cingulate island sign (CIS). Education negatively covaried with metabolism in the left inferior parietal cortex and precuneus (making the CIS more prominent). DISCUSSION These findings point out the importance of tailoring interpretation of DLB biomarkers considering the concomitant effect of individual, non-disease-related variables such as age and cognitive reserve.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Chincarini
- National Institute of Nuclear Physics (INFN), Genoa Section, Genoa, Italy
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Rose Bruffaerts
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Neurology Department, University Hospitals Leuven, Leuven, Belgium.,Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Neurology Department, University Hospitals Leuven, Leuven, Belgium
| | - Milica G Kramberger
- Department of Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Trost
- Department of Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and NIMTLab, Geneva University, Geneva, Switzerland
| | - Nicolas Nicastro
- Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland.,Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Giovanni B Frisoni
- LANVIE (Laboratoire de Neuroimagerie du Vieillissement), Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Parkinson's Disease Rehabilitation Centre, FERB ONLUS-S, Isidoro Hospital, Trescore Balneario, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Miguel A Ochoa-Figueroa
- Department of Clinical Physiology in Linköping, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Department of Diagnostic Radiology, Linköping University Hospital, Linköping, Sweden.,Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Anette Davidsson
- Department of Clinical Physiology in Linköping, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Valle Camacho
- Servicio de Medicina Nuclear, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Enrico Peira
- National Institute of Nuclear Physics (INFN), Genoa Section, Genoa, Italy.,Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
| | - Dario Arnaldi
- Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy.,Clinical Neurology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Pardini
- Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy.,Clinical Neurology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Stefano Raffa
- Department of Health Sciences, University of Genoa, Italy
| | - Alberto Miceli
- Department of Health Sciences, University of Genoa, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Health Sciences, University of Genoa, Italy
| | - Dag Aarsland
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Stavanger, Norway.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King's College, London, UK
| | - Flavio Nobili
- Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy.,Clinical Neurology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Morbelli
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Health Sciences, University of Genoa, Italy
| |
Collapse
|
47
|
Abstract
This article presents an overview of imaging agents for PET that have been applied for research and diagnostic purposes in patients affected by dementia. Classified by the target which the agents visualize, seven groups of tracers can be distinguished, namely radiopharmaceuticals for: (1) Misfolded proteins (ß-amyloid, tau, α-synuclein), (2) Neuroinflammation (overexpression of translocator protein), (3) Elements of the cholinergic system, (4) Elements of monoamine neurotransmitter systems, (5) Synaptic density, (6) Cerebral energy metabolism (glucose transport/ hexokinase), and (7) Various other proteins. This last category contains proteins involved in mechanisms underlying neuroinflammation or cognitive impairment, which may also be potential therapeutic targets. Many receptors belong to this category: AMPA, cannabinoid, colony stimulating factor 1, metabotropic glutamate receptor 1 and 5 (mGluR1, mGluR5), opioid (kappa, mu), purinergic (P2X7, P2Y12), sigma-1, sigma-2, receptor for advanced glycation endproducts, and triggering receptor expressed on myeloid cells-1, besides several enzymes: cyclooxygenase-1 and 2 (COX-1, COX-2), phosphodiesterase-5 and 10 (PDE5, PDE10), and tropomyosin receptor kinase. Significant advances in neuroimaging have been made in the last 15 years. The use of 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) for quantification of regional cerebral glucose metabolism is well-established. Three tracers for ß-amyloid plaques have been approved by the Food and Drug Administration and European Medicines Agency. Several tracers for tau neurofibrillary tangles are already applied in clinical research. Since many novel agents are in the preclinical or experimental stage of development, further advances in nuclear medicine imaging can be expected in the near future. PET studies with established tracers and tracers for novel targets may result in early diagnosis and better classification of neurodegenerative disorders and in accurate monitoring of therapy trials which involve these targets. PET data have prognostic value and may be used to assess the response of the human brain to interventions, or to select the appropriate treatment strategy for an individual patient.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sofia Marcolini
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands
| | - Peter Paul de Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands; University of Antwerp, Born-Bunge Institute, Neurochemistry and Behavior, Campus Drie Eiken, Wilrijk, Belgium
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands; Ghent University, Ghent, Belgium
| |
Collapse
|
48
|
Dev SI, Dickerson BC, Touroutoglou A. Neuroimaging in Frontotemporal Lobar Degeneration: Research and Clinical Utility. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:93-112. [PMID: 33433871 PMCID: PMC8787866 DOI: 10.1007/978-3-030-51140-1_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Frontotemporal lobar dementia (FTLD) is a clinically and pathologically complex disease. Advances in neuroimaging techniques have provided a specialized set of tools to investigate underlying pathophysiology and identify clinical biomarkers that aid in diagnosis, prognostication, monitoring, and identification of appropriate endpoints in clinical trials. In this chapter, we review data discussing the utility of neuroimaging biomarkers in sporadic FTLD, with an emphasis on current and future clinical applications. Among those modalities readily utilized in clinical settings, T1-weighted structural magnetic resonance imaging (MRI) and 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) are best supported in differential diagnosis and as targets for clinical trial endpoints. However, a number of nonclinical neuroimaging modalities, including diffusion tensor imaging and resting-state functional connectivity MRI, show promise as biomarkers to predict progression and as clinical trial endpoints. Other neuroimaging modalities, including amyloid PET, Tau PET, and arterial spin labeling MRI, are also discussed, though more work is required to establish their utility in FTLD in clinical settings.
Collapse
Affiliation(s)
- Sheena I Dev
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, USA
| | - Bradford C Dickerson
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, USA.
| | - Alexandra Touroutoglou
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
49
|
Schaeffer MJ, Chan L, Barber PA. The neuroimaging of neurodegenerative and vascular disease in the secondary prevention of cognitive decline. Neural Regen Res 2021; 16:1490-1499. [PMID: 33433462 PMCID: PMC8323688 DOI: 10.4103/1673-5374.303011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Structural brain changes indicative of dementia occur up to 20 years before the onset of clinical symptoms. Efforts to modify the disease process after the onset of cognitive symptoms have been unsuccessful in recent years. Thus, future trials must begin during the preclinical phases of the disease before symptom onset. Age related cognitive decline is often the result of two coexisting brain pathologies: Alzheimer’s disease (amyloid, tau, and neurodegeneration) and vascular disease. This review article highlights some of the common neuroimaging techniques used to visualize the accumulation of neurodegenerative and vascular pathologies during the preclinical stages of dementia such as structural magnetic resonance imaging, positron emission tomography, and white matter hyperintensities. We also describe some emerging neuroimaging techniques such as arterial spin labeling, diffusion tensor imaging, and quantitative susceptibility mapping. Recent literature suggests that structural imaging may be the most sensitive and cost-effective marker to detect cognitive decline, while molecular positron emission tomography is primarily useful for detecting disease specific pathology later in the disease process. Currently, the presence of vascular disease on magnetic resonance imaging provides a potential target for optimizing vascular risk reduction strategies, and the presence of vascular disease may be useful when combined with molecular and metabolic markers of neurodegeneration for identifying the risk of cognitive impairment.
Collapse
Affiliation(s)
- Morgan J Schaeffer
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Leona Chan
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Philip A Barber
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
50
|
Szturm T, Beheshti I, Mahana B, Hobson DE, Goertzen A, Ko JH. Imaging Cerebral Glucose Metabolism during Dual-Task Walking in Patients with Parkinson's disease. J Neuroimaging 2020; 31:356-362. [PMID: 33289947 DOI: 10.1111/jon.12812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Gait impairment is a hallmark of Parkinson's disease (PD). Natural walking involves more cognitive demand than treadmill walking or in-laboratory walking tests because patients have to actively work on navigation and top-down cognitive control which taxes cognitive reserve in the prefrontal cortex. To mimic the prefrontal engagement occurring with natural walking in a controlled and safe environment, dual-task (DT) treadmill walking has been developed. In this study, we tested the feasibility of imaging DT walking-related changes in brain glucose metabolism in patients with PD. METHODS Fifteen patients with PD were scanned with fluorodeoxyglucose (FDG) positron emission tomography. Five patients performed DT walking, and 10 patients were rested during the FDG uptake period. First, the images were contrasted between the groups. Second, the walking-related brain glucose metabolism was inspected at the individual level. RESULTS Consistently increased glucose metabolism was identified in DT walking versus rest in the primary visual/sensorimotor areas, thalamus, superior colliculus, and cerebellum. In individual level analysis, patients with less progressed disease (n = 3) showed prefrontal activity during DT walking while patients with more progressed disease (n = 2) did not. CONCLUSION This study confirms the feasibility of imaging glucose metabolism during DT walking in patients with PD. We also report that during DT walking, there is a lesser degree of prefrontal engagement in the patients with more progressed disease compared to those with less progressed disease, implying increased degrees of frontal dysfunction with PD progression.
Collapse
Affiliation(s)
- Tony Szturm
- College of Rehabilitation Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Graduate Program in Biomedical Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Iman Beheshti
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada.,Kleysen Institute for Advanced Medicine, Health Science Centre, Winnipeg, Manitoba, Canada
| | - Bhuvan Mahana
- College of Rehabilitation Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Douglas E Hobson
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew Goertzen
- Graduate Program in Biomedical Engineering, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Radiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ji Hyun Ko
- Graduate Program in Biomedical Engineering, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada.,Kleysen Institute for Advanced Medicine, Health Science Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|