1
|
Yakushev I, Verger A, Brendel M, Cecchin D, Fernandez PA, Fraioli F, Grimmer T, Tolboom N, Traub-Weidinger T, Guedj E, Van Weehaeghe D. Lecanemab approval in EU: what should we be ready for?- the EANM perspective. Eur J Nucl Med Mol Imaging 2025; 52:1607-1610. [PMID: 39789225 PMCID: PMC11928400 DOI: 10.1007/s00259-025-07066-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Affiliation(s)
- Igor Yakushev
- Department of Nuclear Medicine, School of Medicine, TUM University Hospital, Technical University of Munich, Klinikum rechts der Isar Ismaninger Str. 22, 81675, Munich, Germany.
| | - Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU Nancy, Université de Lorraine, Nancy, France
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany
| | - Diego Cecchin
- Department of Medicine, Unit of Nuclear Medicine, University Hospital of Padova, Padova, Italy
| | - Pablo Aguiar Fernandez
- CIMUS, Universidade Santiago de Compostela & Nuclear Medicine Department, Univ. Hospital IDIS, Santiago de Compostela, Spain
| | - Francesco Fraioli
- Institute of Nuclear Medicine, University College London, London, UK
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, School of Medicine, TUM University Hospital, Technical University of Munich, Munich, Germany
| | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Tatjana Traub-Weidinger
- Department of Diagnostic and Therapeutic Nuclear Medicine, Clinic Donaustadt, Vienna Health Care Group, Vienna, Austria
| | - Eric Guedj
- Département de Médecine Nucléaire, Aix Marseille Univ, APHM, CNRS, Centrale Marseille, Institut Fresnel, Hôpital de La Timone, CERIMED, Marseille, France
| | | |
Collapse
|
2
|
Pokrzyk J, Kulczyńska-Przybik A, Guzik-Makaruk E, Winkel I, Mroczko B. Clinical Importance of Amyloid Beta Implication in the Detection and Treatment of Alzheimer's Disease. Int J Mol Sci 2025; 26:1935. [PMID: 40076562 PMCID: PMC11900921 DOI: 10.3390/ijms26051935] [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: 12/20/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
The role of amyloid beta peptide (Aβ) in memory regulation has been a subject of substantial interest and debate in neuroscience, because of both physiological and clinical issues. Understanding the dual nature of Aβ in memory regulation is crucial for developing effective treatments for Alzheimer's disease (AD). Moreover, accurate detection and quantification methods of Aβ isoforms have been tested for diagnostic purposes and therapeutic interventions. This review provides insight into the current knowledge about the methods of amyloid beta detection in vivo and in vitro by fluid tests and brain imaging methods (PET), which allow for preclinical recognition of the disease. Currently, the priority in the development of new therapies for Alzheimer's disease has been given to potential changes in the progression of the disease. In light of increasing amounts of data, this review was focused on the diagnostic and therapeutic employment of amyloid beta in Alzheimer's disease.
Collapse
Affiliation(s)
- Justyna Pokrzyk
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland; (J.P.); (B.M.)
| | - Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland; (J.P.); (B.M.)
| | | | - Izabela Winkel
- Dementia Disorders Centre, Medical University of Wroclaw, 50-425 Ścinawa, Poland;
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland; (J.P.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland
| |
Collapse
|
3
|
Juengling F, Wuest F, Schirrmacher R, Abele J, Thiel A, Soucy JP, Camicioli R, Garibotto V. PET Imaging in Dementia: Mini-Review and Canadian Perspective for Clinical Use. Can J Neurol Sci 2025; 52:26-38. [PMID: 38433571 DOI: 10.1017/cjn.2024.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
PET imaging is increasingly recognized as an important diagnostic tool to investigate patients with cognitive disturbances of possible neurodegenerative origin. PET with 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG), assessing glucose metabolism, provides a measure of neurodegeneration and allows a precise differential diagnosis among the most common neurodegenerative diseases, such as Alzheimer's disease, frontotemporal dementia or dementia with Lewy bodies. PET tracers specific for the pathological deposits characteristic of different neurodegenerative processes, namely amyloid and tau deposits typical of Alzheimer's Disease, allow the visualization of these aggregates in vivo. [18F]FDG and amyloid PET imaging have reached a high level of clinical validity and are since 2022 investigations that can be offered to patients in standard clinical care in most of Canada.This article will briefly review and summarize the current knowledge on these diagnostic tools, their integration into diagnostic algorithms as well as perspectives for future developments.
Collapse
Affiliation(s)
- Freimut Juengling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Faculty, University of Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ralf Schirrmacher
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Jonathan Abele
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Department of Neurology and Neurosurgery, Lady Davis Institute for Medical Research, McGill University, Montréal, QC, Canada
| | - Jean-Paul Soucy
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Valentina Garibotto
- Diagnostic Department, Nuclear Medicine and Molecular Imaging Division, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
4
|
Jovalekic A, Bullich S, Roé-Vellvé N, Kolinger GD, Howard LR, Elsholz F, Lagos-Quintana M, Blanco-Rodriguez B, Pérez-Martínez E, Gismondi R, Perrotin A, Chapleau M, Keegan R, Mueller A, Stephens AW, Koglin N. Experiences from Clinical Research and Routine Use of Florbetaben Amyloid PET-A Decade of Post-Authorization Insights. Pharmaceuticals (Basel) 2024; 17:1648. [PMID: 39770490 PMCID: PMC11728731 DOI: 10.3390/ph17121648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Florbetaben (FBB) is a radiopharmaceutical approved by the FDA and EMA in 2014 for the positron emission tomography (PET) imaging of brain amyloid deposition in patients with cognitive impairment who are being evaluated for Alzheimer's disease (AD) or other causes of cognitive decline. Initially, the clinical adoption of FBB PET faced significant barriers, including reimbursement challenges and uncertainties regarding its integration into diagnostic clinical practice. This review examines the progress made in overcoming these obstacles and describes the concurrent evolution of the diagnostic landscape. Advances in quantification methods have further strengthened the traditional visual assessment approach. Over the past decade, compelling evidence has emerged, demonstrating that amyloid PET has a strong impact on AD diagnosis, management, and outcomes across diverse clinical scenarios, even in the absence of amyloid-targeted therapies. Amyloid PET imaging has become essential in clinical trials and the application of new AD therapeutics, particularly for confirming eligibility criteria (i.e., the presence of amyloid plaques) and monitoring biological responses to amyloid-lowering therapies. Since its approval, FBB PET has transitioned from a purely diagnostic tool aimed primarily at excluding amyloid pathology to a critical component in AD drug development, and today, it is essential in the diagnostic workup and therapy management of approved AD treatments.
Collapse
Affiliation(s)
| | - Santiago Bullich
- Life Molecular Imaging GmbH, Tegeler Str. 7, 13353 Berlin, Germany
| | - Núria Roé-Vellvé
- Life Molecular Imaging GmbH, Tegeler Str. 7, 13353 Berlin, Germany
| | | | | | - Floriana Elsholz
- Life Molecular Imaging GmbH, Tegeler Str. 7, 13353 Berlin, Germany
| | | | | | | | | | - Audrey Perrotin
- Life Molecular Imaging GmbH, Tegeler Str. 7, 13353 Berlin, Germany
| | - Marianne Chapleau
- Life Molecular Imaging Inc., 75 State Street, Floor 1, Boston, MA 02109, USA
| | - Richard Keegan
- Life Molecular Imaging Inc., 75 State Street, Floor 1, Boston, MA 02109, USA
| | - Andre Mueller
- Life Molecular Imaging GmbH, Tegeler Str. 7, 13353 Berlin, Germany
| | | | - Norman Koglin
- Life Molecular Imaging GmbH, Tegeler Str. 7, 13353 Berlin, Germany
| |
Collapse
|
5
|
Chen K, Wang M, Wu J, Zuo C, Huang Y, Wang W, Zhao M, Zhang Y, Zhang X, Chen S, Liu W, Li M, Ge J, Ma X, Wang J, Zheng L, Guan Y, Dong Q, Cui M, Xie F, Zhao Q, Yu J. Incremental value of amyloid PET in a tertiary memory clinic setting in China. Alzheimers Dement 2024; 20:2516-2525. [PMID: 38329281 PMCID: PMC11032579 DOI: 10.1002/alz.13728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
INTRODUCTION The objective of this study is to investigate the incremental value of amyloid positron emission tomography (Aβ-PET) in a tertiary memory clinic setting in China. METHODS A total of 1073 patients were offered Aβ-PET using 18F-florbetapir. The neurologists determined a suspected etiology (Alzheimer's disease [AD] or non-AD) with a percentage estimate of their confidence and medication prescription both before and after receiving the Aβ-PET results. RESULTS After disclosure of the Aβ-PET results, etiological diagnoses changed in 19.3% of patients, and diagnostic confidence increased from 69.3% to 85.6%. Amyloid PET results led to a change of treatment plan in 36.5% of patients. Compared to the late-onset group, the early-onset group had a more frequent change in diagnoses and a higher increase in diagnostic confidence. DISCUSSION Aβ-PET has significant impacts on the changes of diagnoses and management in Chinese population. Early-onset cases are more likely to benefit from Aβ-PET than late-onset cases. HIGHLIGHTS Amyloid PET contributes to diagnostic changes and its confidence in Chinese patients. Amyloid PET leads to a change of treatment plans in Chinese patients. Early-onset cases are more likely to benefit from amyloid PET than late-onset cases.
Collapse
Affiliation(s)
- Ke‐Liang Chen
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Ming‐Yu Wang
- School of MedicineQingdao UniversityQingdaoShandongChina
- Departments of NeurologyWeifang People's HospitalWeifangShandongChina
| | - Jie Wu
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Chuan‐Tao Zuo
- Department of Nuclear Medicine & PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Yu‐Yuan Huang
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Wei‐Yi Wang
- Department of Nuclear Medicine & PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Meng Zhao
- Department of Neurologythe First Hospital of Jilin UniversityChangchunJilinChina
| | - Ya‐Ru Zhang
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Xue Zhang
- Department of NeurologyQingdao shi zhongxin yiyuanQingdaoShandongChina
| | - Shu‐Fen Chen
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Wei‐Shi Liu
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Meng‐Meng Li
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Jing‐Jie Ge
- Department of Nuclear Medicine & PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Xiao‐Xi Ma
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Jie Wang
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Li Zheng
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Yi‐Hui Guan
- Department of Nuclear Medicine & PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Qiang Dong
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Mei Cui
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Fang Xie
- Department of Nuclear Medicine & PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Qian‐Hua Zhao
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Jin‐Tai Yu
- Department of Neurology and National Center for Neurological DiseasesHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| |
Collapse
|
6
|
Papaliagkas V, Kalinderi K, Vareltzis P, Moraitou D, Papamitsou T, Chatzidimitriou M. CSF Biomarkers in the Early Diagnosis of Mild Cognitive Impairment and Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24108976. [PMID: 37240322 DOI: 10.3390/ijms24108976] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a rapidly growing disease that affects millions of people worldwide, therefore there is an urgent need for its early diagnosis and treatment. A huge amount of research studies are performed on possible accurate and reliable diagnostic biomarkers of AD. Due to its direct contact with extracellular space of the brain, cerebrospinal fluid (CSF) is the most useful biological fluid reflecting molecular events in the brain. Proteins and molecules that reflect the pathogenesis of the disease, e.g., neurodegeneration, accumulation of Abeta, hyperphosphorylation of tau protein and apoptosis may be used as biomarkers. The aim of the current manuscript is to present the most commonly used CSF biomarkers for AD as well as novel biomarkers. Three CSF biomarkers, namely total tau, phospho-tau and Abeta42, are believed to have the highest diagnostic accuracy for early AD diagnosis and the ability to predict AD development in mild cognitive impairment (MCI) patients. Moreover, other biomarkers such as soluble amyloid precursor protein (APP), apoptotic proteins, secretases and inflammatory and oxidation markers are believed to have increased future prospects.
Collapse
Affiliation(s)
- Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Alexandrion University Campus, 57400 Sindos, Greece
| | - Kallirhoe Kalinderi
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Patroklos Vareltzis
- Department of Chemical Engineering, School of Engineering, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Despoina Moraitou
- Laboratory of Psychology, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Theodora Papamitsou
- Histology and Embryology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Chatzidimitriou
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Alexandrion University Campus, 57400 Sindos, Greece
| |
Collapse
|
7
|
Caprioglio C, Garibotto V, Jessen F, Frölich L, Allali G, Assal F, Frisoni GB, Altomare D. The Clinical Use of Alzheimer's Disease Biomarkers in Patients with Mild Cognitive Impairment: A European Alzheimer's Disease Consortium Survey. J Alzheimers Dis 2022; 89:535-551. [PMID: 35912743 PMCID: PMC9535580 DOI: 10.3233/jad-220333] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Recent advances occurred in the field of Alzheimer's disease (AD) biomarkers and the introduction of a research framework grounded on a biomarker-based definition of AD might have fostered an increased clinical use of AD biomarkers. For this reason, an up-to-date depiction of the clinical use of AD biomarkers is needed. OBJECTIVE To investigate the clinical use of the main AD biomarkers in patients with mild cognitive impairment (MCI) by examining the beliefs and preferences of professionals (clinicians and biomarker experts) of the European Alzheimer's Disease Consortium (EADC). METHODS 150 professionals filled in an online survey from May to September 2020. The investigated biomarkers were medial temporal lobe atrophy score (MTA) on structural MRI, typical AD (i.e., temporoparietal and posterior cingulate) hypometabolism on FDG-PET, CSF (Aβ42, p-tau, t-tau), amyloid-PET and tau-PET. RESULTS The frequency of responders reporting a frequent-to-constant use of MTA (77%) is higher than that of those reporting a frequent-to-constant use of the other AD biomarkers (i.e. , CSF 45%, p = 0.014; FDG-PET: 32%, p < 0.001; amyloid-PET: 8%, p < 0.001; and tau-PET: 2%, p < 0.001). CSF is considered the most valuable biomarker in terms of additional diagnostic value, followed by amyloid-PET, tau-PET, and typical AD hypometabolism on FDG-PET. CONCLUSION AD biomarkers are widely used across European memory clinics with a clinical research background for the diagnosis of MCI. Overall, we observed that CSF is currently considered as the most useful biomarker, followed by amyloid-PET.
Collapse
Affiliation(s)
- Camilla Caprioglio
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland,Geneva Memory Center, Geneva University Hospitals, Geneva, Switzerland
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland,Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland
| | - Frank Jessen
- Department of Psychiatry, University Hospital and Medical Faculty, University of Cologne, Cologne, Germany,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute for Mental Health, University of Heidelberg, Mannheim, Germany
| | - Gilles Allali
- Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland,Department of Neurology, Division of Cognitive & Motor Aging, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | - Frédéric Assal
- Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Giovanni B. Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland,Geneva Memory Center, Geneva University Hospitals, Geneva, Switzerland
| | - Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland,Geneva Memory Center, Geneva University Hospitals, Geneva, Switzerland,Correspondence to: Daniele Altomare, PhD, Memory Clinic, Geneva University Hospitals, Rue Gabrielle-Perret-Gentil 6, 1205 Geneva, Switzerland. Tel.: +41 22 372 58 00; E-mail:
| | | |
Collapse
|
8
|
Mahaman YAR, Embaye KS, Huang F, Li L, Zhu F, Wang JZ, Liu R, Feng J, Wang X. Biomarkers used in Alzheimer's disease diagnosis, treatment, and prevention. Ageing Res Rev 2022; 74:101544. [PMID: 34933129 DOI: 10.1016/j.arr.2021.101544] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), being the number one in terms of dementia burden, is an insidious age-related neurodegenerative disease and is presently considered a global public health threat. Its main histological hallmarks are the Aβ senile plaques and the P-tau neurofibrillary tangles, while clinically it is marked by a progressive cognitive decline that reflects the underlying synaptic loss and neurodegeneration. Many of the drug therapies targeting the two pathological hallmarks namely Aβ and P-tau have been proven futile. This is probably attributed to the initiation of therapy at a stage where cognitive alterations are already obvious. In other words, the underlying neuropathological changes are at a stage where these drugs lack any therapeutic value in reversing the damage. Therefore, there is an urgent need to start treatment in the very early stage where these changes can be reversed, and hence, early diagnosis is of primordial importance. To this aim, the use of robust and informative biomarkers that could provide accurate diagnosis preferably at an earlier phase of the disease is of the essence. To date, several biomarkers have been established that, to a different extent, allow researchers and clinicians to evaluate, diagnose, and more specially exclude other related pathologies. In this study, we extensively reviewed data on the currently explored biomarkers in terms of AD pathology-specific and non-specific biomarkers and highlighted the recent developments in the diagnostic and theragnostic domains. In the end, we have presented a separate elaboration on aspects of future perspectives and concluding remarks.
Collapse
|
9
|
PET imaging in dementia. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00089-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
10
|
Jeong YM, Lee JG, Cho HJ, Lee WS, Jeong J, Lee JS. Differential Clearance of Aβ Species from the Brain by Brain Lymphatic Endothelial Cells in Zebrafish. Int J Mol Sci 2021; 22:11883. [PMID: 34769316 PMCID: PMC8584359 DOI: 10.3390/ijms222111883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 02/01/2023] Open
Abstract
The failure of amyloid beta (Aβ) clearance is a major cause of Alzheimer's disease, and the brain lymphatic systems play a crucial role in clearing toxic proteins. Recently, brain lymphatic endothelial cells (BLECs), a non-lumenized lymphatic cell in the vertebrate brain, was identified, but Aβ clearance via this novel cell is not fully understood. We established an in vivo zebrafish model using fluorescently labeled Aβ42 to investigate the role of BLECs in Aβ clearance. We discovered the efficient clearance of monomeric Aβ42 (mAβ42) compared to oligomeric Aβ42 (oAβ42), which was illustrated by the selective uptake of mAβ42 by BLECs and peripheral transport. The genetic depletion, pharmacological inhibition via the blocking of the mannose receptor, or the laser ablation of BLECs resulted in the defective clearance of mAβ42. The treatment with an Aβ disaggregating agent facilitated the internalization of oAβ42 into BLECs and improved the peripheral transport. Our findings reveal a new role of BLECs in the differential clearance of mAβ42 from the brain and provide a novel therapeutic strategy based on promoting Aβ clearance.
Collapse
Affiliation(s)
- Yun-Mi Jeong
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Jae-Geun Lee
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Hyun-Ju Cho
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Wang Sik Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jinyoung Jeong
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Department of Biotechnology, KRIBB School, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Jeong-Soo Lee
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
- Department of Functional Genomics, KRIBB School, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| |
Collapse
|
11
|
The approval of a disease-modifying treatment for Alzheimer's disease: impact and consequences for the nuclear medicine community. Eur J Nucl Med Mol Imaging 2021; 48:3033-3036. [PMID: 34272989 DOI: 10.1007/s00259-021-05485-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
12
|
Garibotto V, Boccardi M, Chiti A, Frisoni GB. Molecular imaging and fluid biomarkers of Alzheimer's disease neuropathology: an opportunity for integrated diagnostics. Eur J Nucl Med Mol Imaging 2021; 48:2067-2069. [PMID: 33688995 DOI: 10.1007/s00259-020-05116-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Valentina Garibotto
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland. .,Nuclear Medicine and Molecular Division, University Hospitals of Geneva, Rue Gabrielle-Perret-Gentil 4, 1205, Geneva, Switzerland.
| | - Marina Boccardi
- German Center for Neurodegenerative Diseases (DZNE) Rostock-Greifswald, Rostock, Germany
| | - Arturo Chiti
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Giovanni B Frisoni
- Memory Clinic, University Hospital, Geneva, Switzerland.,LANVIE - Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
| |
Collapse
|
13
|
Alongi P, Chiaravalloti A, Berti V, Vellani C, Trifirò G, Puccini G, Carli G, Chincarini A, Morbelli S, Perani D, Sestini S. Amyloid PET in the diagnostic workup of neurodegenerative disease. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00428-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Sakurai K, Kaneda D, Inui S, Uchida Y, Morimoto S, Nihashi T, Kato T, Ito K, Hashizume Y. Simple Quantitative Indices for the Differentiation of Advanced-Stage Alzheimer's Disease and Other Limbic Tauopathies. J Alzheimers Dis 2021; 81:1093-1102. [PMID: 33843680 DOI: 10.3233/jad-210043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The differentiation of Alzheimer's disease (AD) from age-related limbic tauopathies (LT), including argyrophilic grain disease (AGD) and senile dementia of the neurofibrillary tangle type (SD-NFT), is often challenging because specific clinical diagnostic criteria have not yet been established. Despite the utility of specific biomarkers evaluating amyloid and tau to detect the AD-related pathophysiological changes, the expense and associated invasiveness preclude their use as first-line diagnostic tools for all demented patients. Therefore, less invasive and costly biomarkers would be valuable in routine clinical practice for the differentiation of AD and LT. OBJECTIVE The purpose of this study is to develop a simple reproducible method on magnetic resonance imaging (MRI) that could be adopted in daily clinical practice for the differentiation of AD and other forms of LT. METHODS Our newly proposed three quantitative indices and well-known medial temporal atrophy (MTA) score were evaluated using MRI of pathologically-proven advanced-stage 21 AD, 10 AGD, and 2 SD-NFT patients. RESULTS Contrary to MTA score, hippocampal angle (HPA), inferior horn area (IHA), and ratio between HPA and IHA (i.e., IHPA index) demonstrated higher diagnostic performance and reproducibility, especially to differentiate advanced-stage AD patients with Braak neurofibrillary tangle stage V/VI from LT patients (the area under the receiver-operating-characteristic curve of 0.83, 089, and 0.91; intraclass correlation coefficients of 0.930, 0.998, and 0.995, respectively). CONCLUSION Quantitative indices reflecting hippocampal deformation with ventricular enlargement are useful to differentiate advanced-stage AD from LT. This simple and convenient method could be useful in daily clinical practice.
Collapse
Affiliation(s)
- Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Daita Kaneda
- Choju Medical Institute, Fukushimura Hospital, Toyohashi, Japan
| | - Shohei Inui
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuto Uchida
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Satoru Morimoto
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Takashi Nihashi
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Takashi Kato
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kengo Ito
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | | |
Collapse
|
15
|
Pyun JM, Ryu JS, Lee R, Shim KH, Youn YC, Ryoo N, Han SW, Park YH, Kang S, An SSA, Kim S. Plasma Amyloid-β Oligomerization Tendency Predicts Amyloid PET Positivity. Clin Interv Aging 2021; 16:749-755. [PMID: 33958861 PMCID: PMC8096417 DOI: 10.2147/cia.s312473] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023] Open
Abstract
Purpose Among other emerging amyloid-targeting blood-based biomarkers, Multimer Detection System-Oligomeric Amyloid-β (MDS-OAβ) measures dynamic changes in concentration of oligomeric amyloid-β (OAβ), which is considered the main pathogenic culprit of Alzheimer’s disease (AD), in plasma after spiking with synthetic amyloid-β (Aβ). We aimed to investigate the predictability of MDS-OAβ on amyloid positron emission tomography (PET) positivity. Patients and Methods A total of 96 subjects who visited Seoul National University Bundang Hospital for medical check-up complaining of cognitive decline and had undergone extensive medical assessment were recruited. Amyloid statuses were dichotomized into positive or negative based on visual assessment of amyloid PET. Plasma OAβ concentration was measured by MDS-OAβ. In the previous validation study, 0.78ng/mL was established as the cut-off value and the plasma OAβ concentration higher than or equal to the cut-off value was defined as MDS-OAβ positive. Results MDS-OAβ positivity could discriminate amyloid PET positivity with the AUC value of 0.855 (95% CI 0.776–0.933). Adding MDS-OAβ positivity to prediction models including age, MMSE score, and APOE ε4 status improved performance up to the AUC value of 0.926 (95% CI 0.871–0.980). Conclusion The Aβ oligomerization tendency in plasma could predict amyloid PET positivity with high performance, and, when it is combined with age, MMSE score, and APOE ε4 status, predictability was improved substantially. This suggests the potential of MDS-OAβ as a useful initial stage test in the clinical and research fields of AD.
Collapse
Affiliation(s)
- Jung-Min Pyun
- Department of Neurology, Uijeongbu Eulji Medical Center, Eulji University, Uijeongbu-si, Gyeonggi-do, Republic of Korea
| | - Ji Sun Ryu
- Research and Development, PeopleBio Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Ryan Lee
- Research and Development, PeopleBio Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyu Hawn Shim
- Department of Neurology, Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Nayoung Ryoo
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Sang-Won Han
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Young Ho Park
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Sungmin Kang
- Research and Development, PeopleBio Inc., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seong Soo A An
- Department of Bionanotechnology, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
16
|
Hugon J, Paquet C. The PKR/P38/RIPK1 Signaling Pathway as a Therapeutic Target in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22063136. [PMID: 33808629 PMCID: PMC8003462 DOI: 10.3390/ijms22063136] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/23/2022] Open
Abstract
Neuropathological lesions in Alzheimer’s disease (AD) include amyloid plaques formed by the accumulation of amyloid peptides, neurofibrillary tangles made of hyperphosphorylated tau protein, synaptic and neuronal degenerations, and neuroinflammation. The cause of AD is unknown, but according to the amyloid hypothesis, amyloid oligomers could lead to the activation of kinases such as eukaryotic translation initiation factor 2-alpha kinase 2 (PKR), p38, and receptor-interacting serine/threonine-protein kinase 1 (RIPK1), which all belong to the same stress-activated pathway. Many toxic kinase activations have been described in AD patients and in experimental models. A p38 mitogen-activated protein kinase inhibitor was recently tested in clinical trials but with unsuccessful results. The complex PKR/P38/RIPK1 (PKR/dual specificity mitogen-activated protein kinase kinase 6 (MKK6)/P38/MAP kinase-activated protein kinase 2 (MK2)/RIPK1) is highly activated in AD brains and in the brains of AD transgenic animals. To delineate the implication of this pathway in AD, we carried out a search on PubMed including PKR/MKK6/p38/MK2/RIPK1, Alzheimer, and therapeutics. The involvement of this signaling pathway in the genesis of AD lesions, including Aβ accumulations and tau phosphorylation as well as cognitive decline, is demonstrated by the reports described in this review. A future combination strategy with kinase inhibitors should be envisaged to modulate the consequences for neurons and other brain cells linked to the abnormal activation of this pathway.
Collapse
Affiliation(s)
- Jacques Hugon
- Correspondence: ; Tel.: +33-140-054-313; Fax: +33-140-054-339
| | | |
Collapse
|
17
|
Altomare D, Caprioglio C, Assal F, Allali G, Mendes A, Ribaldi F, Ceyzeriat K, Martins M, Tomczyk S, Stampacchia S, Dodich A, Boccardi M, Chicherio C, Frisoni GB, Garibotto V. Diagnostic value of amyloid-PET and tau-PET: a head-to-head comparison. Eur J Nucl Med Mol Imaging 2021; 48:2200-2211. [PMID: 33638661 PMCID: PMC8175315 DOI: 10.1007/s00259-021-05246-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/07/2021] [Indexed: 11/26/2022]
Abstract
Purpose Assess the individual and combined diagnostic value of amyloid-PET and tau-PET in a memory clinic population. Methods Clinical reports of 136 patients were randomly assigned to two diagnostic pathways: AMY-TAU, amyloid-PET is presented before tau-PET; and TAU-AMY, tau-PET is presented before amyloid-PET. Two neurologists independently assessed all reports with a balanced randomized design, and expressed etiological diagnosis and diagnostic confidence (50–100%) three times: (i) at baseline based on the routine diagnostic workup, (ii) after the first exam (amyloid-PET for the AMY-TAU pathway, and tau-PET for the TAU-AMY pathway), and (iii) after the remaining exam. The main outcomes were changes in diagnosis (from AD to non-AD or vice versa) and in diagnostic confidence. Results Amyloid-PET and tau-PET, when presented as the first exam, resulted in a change of etiological diagnosis in 28% (p = 0.006) and 28% (p < 0.001) of cases, and diagnostic confidence increased by 18% (p < 0.001) and 19% (p < 0.001) respectively, with no differences between exams (p > 0.05). We observed a stronger impact of a negative amyloid-PET versus a negative tau-PET (p = 0.014). When added as the second exam, amyloid-PET and tau-PET resulted in a further change in etiological diagnosis in 6% (p = 0.077) and 9% (p = 0.149) of cases, and diagnostic confidence increased by 4% (p < 0.001) and 5% (p < 0.001) respectively, with no differences between exams (p > 0.05). Conclusion Amyloid-PET and tau-PET significantly impacted diagnosis and diagnostic confidence in a similar way, although a negative amyloid-PET has a stronger impact on diagnosis than a negative tau-PET. Adding either of the two as second exam further improved diagnostic confidence. Trial number PB 2016-01346. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05246-x.
Collapse
Affiliation(s)
- Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland. .,Memory Clinic, Geneva University Hospitals, Geneva, Switzerland.
| | - Camilla Caprioglio
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.,Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Frédéric Assal
- Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gilles Allali
- Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland.,Department of Neurology, Division of Cognitive & Motor Aging, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | - Aline Mendes
- Division of Geriatrics, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.,Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), Saint John of God Clinical Research Centre, Brescia, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Kelly Ceyzeriat
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Marta Martins
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.,Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Szymon Tomczyk
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.,Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Sara Stampacchia
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alessandra Dodich
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Marina Boccardi
- Late Translational Dementia Research Group, German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Rostock, Germany
| | | | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.,Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Valentina Garibotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
18
|
Librizzi D, Cabanel N, Zavorotnyy M, Riehl E, Kircher T, Luster M, Hooshyar Yousefi B. Clinical Relevance of [ 18F]Florbetaben and [ 18F]FDG PET/CT Imaging on the Management of Patients with Dementia. Molecules 2021; 26:molecules26051282. [PMID: 33652938 PMCID: PMC7956266 DOI: 10.3390/molecules26051282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 11/18/2022] Open
Abstract
PET of β-Amyloid plaques (Aβ) using [18F]florbetaben ([18F]FBB) and [18F]fluorodeoxyglucose ([18F]FDG) increasingly aid clinicians in early diagnosis of dementia, including Alzheimer’s disease (AD), frontotemporal disease, dementia with Lewy bodies, and vascular dementia. The aim of this retrospective analysis was to evaluate clinical relevance of [18F]FBB, [18F]FDG PET and complimentary CSF measurements in patients with suspected dementia. In this study, 40 patients with clinically suspected or history of dementia underwent (1) measurement of Aβ peptides, total tau, and p-tau protein levels in the cerebrospinal fluid (CSF) compared with healthy controls (HC); (2) clinical and neuropsychological assessment, which included Consortium to Establish a Registry for Alzheimer’s Disease neuropsychological assessment battery (CERAD-NAB); (3) [18F]FBB and [18F]FDG PET imaging within an average of 3 weeks. The subjects were within 15 days stratified using PET, CSF measurements as HC, mild cognitive impaired (MCI) and dementia including Alzheimer´s disease. The predictive dementia-related cognitive decline values were supporting the measurements. PET images were evaluated visually and quantitatively using standard uptake value ratios (SUVR). Twenty-one (52.5%) subjects were amyloid-positive (Aβ+), with a median neocortical SUVR of 1.80 for AD versus 1.20 relative to the respective 19 (47.5 %) amyloid-negative (Aβ-) subjects. Moreover, the [18F]FDG and [18F]FBB confirmed within a sub-group of 10 patients a good complimentary role by correlation between amyloid pathology and brain glucose metabolism in 8 out of 10 subjects. The results suggest the clinical relevance for [18F]FBB combined with [18F]FDG PET retention and CFS measurements serving the management of our patients with dementia. Therefore, [18F]FBB combined with [18F]FDG PET is a helpful tool for differential diagnosis, and supports the patients’ management as well as treatment.
Collapse
Affiliation(s)
- Damiano Librizzi
- Department of Nuclear Medicine, Philipps-University of Marburg, 35043 Marburg, Germany; (D.L.); (E.R.); (M.L.)
| | - Nicole Cabanel
- Department of Psychiatry and Psychotherapy, Philipps-University of Marburg, 35039 Marburg, Germany; (N.C.); (M.Z.); (T.K.)
- Marburg Center for Mind, Brain and Behavior—MCMBB, University of Marburg, 35032 Marburg, Germany
| | - Maxim Zavorotnyy
- Department of Psychiatry and Psychotherapy, Philipps-University of Marburg, 35039 Marburg, Germany; (N.C.); (M.Z.); (T.K.)
- Marburg Center for Mind, Brain and Behavior—MCMBB, University of Marburg, 35032 Marburg, Germany
- Department of Psychiatry and Psychotherapy, Psychiatric Services Aargau, Academic Hospital of the University of Zurich, 5210 Windisch, Switzerland
| | - Elisabeth Riehl
- Department of Nuclear Medicine, Philipps-University of Marburg, 35043 Marburg, Germany; (D.L.); (E.R.); (M.L.)
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University of Marburg, 35039 Marburg, Germany; (N.C.); (M.Z.); (T.K.)
- Marburg Center for Mind, Brain and Behavior—MCMBB, University of Marburg, 35032 Marburg, Germany
| | - Markus Luster
- Department of Nuclear Medicine, Philipps-University of Marburg, 35043 Marburg, Germany; (D.L.); (E.R.); (M.L.)
| | - Behrooz Hooshyar Yousefi
- Department of Nuclear Medicine, Philipps-University of Marburg, 35043 Marburg, Germany; (D.L.); (E.R.); (M.L.)
- Correspondence: ; Tel.: +49-6421-586-5806
| |
Collapse
|
19
|
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
|
20
|
Ahmed TF, Ahmed A, Imtiaz F. History in perspective: How Alzheimer's Disease came to be where it is? Brain Res 2021; 1758:147342. [PMID: 33548268 DOI: 10.1016/j.brainres.2021.147342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/18/2021] [Accepted: 01/28/2021] [Indexed: 01/03/2023]
Abstract
Treatment of Alzheimer's Disease (AD) remains an unsolved issue despite the pronounced global attention it has received from researchers over the last four decades. Determining the primary cause of the disease is challenging due to its long prodromal phase and multifactorial etiology. Regardless, academic disagreements amongst the scientific community have helped in making significant advancements in underpinning the molecular basis of disease pathogenesis. Substantial development in fluid and imaging biomarkers for AD led to a sharp turn in defining the disease as a molecular construct, dispensing its clinical definition. With conceptual progress, revisions in the diagnostic criteria of AD were made, culminating into the research framework proposed by National Institute on Aging and Alzheimer's Association in 2018 which unified different stages of the disease continuum, giving a common language of AT(N)1 classification to researchers. With realization that dementia is the final stage of AD spectrum, its early diagnosis by means of cerebrospinal fluid biomarkers, Positron Emission Tomography and Magnetic Resonance Imaging of the brain holds crucial importance in discovering ways of halting the disease progression. This article maps the insights into the pathogenesis as well as the diagnostic criteria and tests for AD as these have evolved over time. A contextualized timeline of how the understanding of AD has matured with advancing knowledge allows future research to be directed and unexplored avenues to be prioritized.
Collapse
Affiliation(s)
- Tehniat F Ahmed
- Department of Biochemistry, Institute of Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan.
| | - Affan Ahmed
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Fauzia Imtiaz
- Department of Biochemistry, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
21
|
Giacomucci G, Mazzeo S, Bagnoli S, Casini M, Padiglioni S, Polito C, Berti V, Balestrini J, Ferrari C, Lombardi G, Ingannato A, Sorbi S, Nacmias B, Bessi V. Matching Clinical Diagnosis and Amyloid Biomarkers in Alzheimer's Disease and Frontotemporal Dementia. J Pers Med 2021; 11:jpm11010047. [PMID: 33466854 PMCID: PMC7830228 DOI: 10.3390/jpm11010047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The aims of this study were to compare the diagnostic accuracy, sensitivity, specificity, and positive and negative predictive values (PPV, NPV) of different cerebrospinal fluid (CSF) amyloid biomarkers and amyloid-Positron Emission Tomography (PET) in patients with a clinical diagnosis of Alzheimer's disease (AD) and Frontotemporal Dementia (FTD); to compare concordance between biomarkers; and to provide an indication of their use and interpretation. METHODS We included 148 patients (95 AD and 53 FTD), who underwent clinical evaluation, neuropsychological assessment, and at least one amyloid biomarker (CSF analysis or amyloid-PET). Thirty-six patients underwent both analyses. One-hundred-thirteen patients underwent Apolipoprotein E (ApoE) genotyping. RESULTS Amyloid-PET presented higher diagnostic accuracy, sensitivity, and NPV than CSF Aβ1-42 but not Aβ42/40 ratio. Concordance between CSF biomarkers and amyloid-PET was higher in FTD patients compared to AD cases. None of the AD patients presented both negative Aβ biomarkers. CONCLUSIONS CSF Aβ42/40 ratio significantly increased the diagnostic accuracy of CSF biomarkers. On the basis of our current and previous data, we suggest a flowchart to guide the use of biomarkers according to clinical suspicion: due to the high PPV of both amyloid-PET and CSF analysis including Aβ42/40, in cases of concordance between at least one biomarker and clinical diagnosis, performance of the other analysis could be avoided. A combination of both biomarkers should be performed to better characterize unclear cases. If the two amyloid biomarkers are both negative, an underlying AD pathology can most probably be excluded.
Collapse
Affiliation(s)
- Giulia Giacomucci
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
| | - Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
- IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143 Florence, Italy;
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
| | - Matteo Casini
- Faculty of Medicine and Surgery, University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Sonia Padiglioni
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
| | - Cristina Polito
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
- IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143 Florence, Italy;
| | - Valentina Berti
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, Via Giovanni Battista Morgagni 50, 50134 Florence, Italy;
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria Careggi, Largo Piero Palagi 1, 50139 Florence, Italy
| | - Juri Balestrini
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
| | - Gemma Lombardi
- IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143 Florence, Italy;
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
- IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143 Florence, Italy;
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
- IRCCS Fondazione Don Carlo Gnocchi, Via Scandicci 269, 50143 Florence, Italy;
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence (NEUROFARBA), Azienda Ospedaliera-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (G.G.); (S.M.); (S.B.); (S.P.); (C.P.); (J.B.); (C.F.); (A.I.); (S.S.); (B.N.)
- Correspondence: ; Tel.: +39-05-7948660; Fax: +39-05-7947484
| |
Collapse
|
22
|
Sala A, Nordberg A, Rodriguez-Vieitez E. Longitudinal pathways of cerebrospinal fluid and positron emission tomography biomarkers of amyloid-β positivity. Mol Psychiatry 2021; 26:5864-5874. [PMID: 33303945 PMCID: PMC8758501 DOI: 10.1038/s41380-020-00950-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 10/09/2020] [Accepted: 11/02/2020] [Indexed: 01/20/2023]
Abstract
Mismatch between CSF and PET amyloid-β biomarkers occurs in up to ≈20% of preclinical/prodromal Alzheimer's disease individuals. Factors underlying mismatching results remain unclear. In this study we hypothesized that CSF/PET discordance provides unique biological/clinical information. To test this hypothesis, we investigated non-demented and demented participants with CSF amyloid-β42 and [18F]Florbetapir PET assessments at baseline (n = 867) and at 2-year follow-up (n = 289). Longitudinal trajectories of amyloid-β positivity were tracked simultaneously for CSF and PET biomarkers. In the longitudinal cohort (n = 289), we found that participants with normal CSF/PET amyloid-β biomarkers progressed more frequently toward CSF/PET discordance than to full CSF/PET positivity (χ2(1) = 5.40; p < 0.05). Progression to CSF+/PET+ status was ten times more frequent in cases with discordant biomarkers, as compared to csf-/pet- cases (χ2(1) = 18.86; p < 0.001). Compared to the CSF+/pet- group, the csf-/PET+ group had lower APOE-ε4ε4 prevalence (χ2(6) = 197; p < 0.001; n = 867) and slower rate of brain amyloid-β accumulation (F(3,600) = 12.76; p < 0.001; n = 608). These results demonstrate that biomarker discordance is a typical stage in the natural history of amyloid-β accumulation, with CSF or PET becoming abnormal first and not concurrently. Therefore, biomarker discordance allows for identification of individuals with elevated risk of progression toward fully abnormal amyloid-β biomarkers, with subsequent risk of neurodegeneration and cognitive decline. Our results also suggest that there are two alternative pathways ("CSF-first" vs. "PET-first") toward established amyloid-β pathology, characterized by different genetic profiles and rates of amyloid-β accumulation. In conclusion, CSF and PET amyloid-β biomarkers provide distinct information, with potential implications for their use as biomarkers in clinical trials.
Collapse
Affiliation(s)
- Arianna Sala
- grid.4714.60000 0004 1937 0626Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden ,grid.15496.3f0000 0001 0439 0892Vita-Salute San Raffaele University, Milan, Italy ,grid.18887.3e0000000417581884In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Agneta Nordberg
- grid.4714.60000 0004 1937 0626Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Theme Aging, The Aging Brain, Karolinska University Hospital, Stockholm, Sweden
| | - Elena Rodriguez-Vieitez
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
23
|
Sörensen A, Blazhenets G, Schiller F, Meyer PT, Frings L. Amyloid biomarkers as predictors of conversion from mild cognitive impairment to Alzheimer's dementia: a comparison of methods. ALZHEIMERS RESEARCH & THERAPY 2020; 12:155. [PMID: 33213489 PMCID: PMC7678323 DOI: 10.1186/s13195-020-00721-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/05/2020] [Indexed: 11/30/2022]
Abstract
Background Amyloid-β (Aβ) PET is an established predictor of conversion from mild cognitive impairment (MCI) to Alzheimer’s dementia (AD). We compared three PET (including an approach based on voxel-wise Cox regression) and one cerebrospinal fluid (CSF) outcome measures in their predictive power. Methods Datasets were retrieved from the ADNI database. In a training dataset (N = 159), voxel-wise Cox regression and principal component analyses were used to identify conversion-related regions (Cox-VOI and AD conversion-related pattern (ADCRP), respectively). In a test dataset (N = 129), the predictive value of mean normalized 18F-florbetapir uptake (SUVR) in AD-typical brain regions (composite SUVR) or the Cox-VOI and the pattern expression score (PES) of ADCRP and CSF Aβ42/Aβ40 as predictors were compared by Cox models (corrected for age and sex). Results All four Aβ measures were significant predictors (p < 0.001). Prediction accuracies (Harrell’s c) showed step-wise significant increases from Cox-SUVR (c = 0.71; HR = 1.84 per Z-score increase), composite SUVR (c = 0.73; HR = 2.18), CSF Aβ42/Aβ40 (c = 0.75; HR = 3.89) to PES (c = 0.77; HR = 2.71). Conclusion The PES of ADCRP is the most predictive Aβ PET outcome measure, comparable to CSF Aβ42/Aβ40, with a slight but statistically significant advantage.
Collapse
Affiliation(s)
- Arnd Sörensen
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
| | - Ganna Blazhenets
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Florian Schiller
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Philipp Tobias Meyer
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Lars Frings
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | | |
Collapse
|
24
|
Chételat G, Arbizu J, Barthel H, Garibotto V, Law I, Morbelli S, van de Giessen E, Agosta F, Barkhof F, Brooks DJ, Carrillo MC, Dubois B, Fjell AM, Frisoni GB, Hansson O, Herholz K, Hutton BF, Jack CR, Lammertsma AA, Landau SM, Minoshima S, Nobili F, Nordberg A, Ossenkoppele R, Oyen WJG, Perani D, Rabinovici GD, Scheltens P, Villemagne VL, Zetterberg H, Drzezga A. Amyloid-PET and 18F-FDG-PET in the diagnostic investigation of Alzheimer's disease and other dementias. Lancet Neurol 2020; 19:951-962. [PMID: 33098804 DOI: 10.1016/s1474-4422(20)30314-8] [Citation(s) in RCA: 278] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 07/22/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
Abstract
Various biomarkers are available to support the diagnosis of neurodegenerative diseases in clinical and research settings. Among the molecular imaging biomarkers, amyloid-PET, which assesses brain amyloid deposition, and 18F-fluorodeoxyglucose (18F-FDG) PET, which assesses glucose metabolism, provide valuable and complementary information. However, uncertainty remains regarding the optimal timepoint, combination, and an order in which these PET biomarkers should be used in diagnostic evaluations because conclusive evidence is missing. Following an expert panel discussion, we reached an agreement on the specific use of the individual biomarkers, based on available evidence and clinical expertise. We propose a diagnostic algorithm with optimal timepoints for these PET biomarkers, also taking into account evidence from other biomarkers, for early and differential diagnosis of neurodegenerative diseases that can lead to dementia. We propose three main diagnostic pathways with distinct biomarker sequences, in which amyloid-PET and 18F-FDG-PET are placed at different positions in the order of diagnostic evaluations, depending on clinical presentation. We hope that this algorithm can support diagnostic decision making in specialist clinical settings with access to these biomarkers and might stimulate further research towards optimal diagnostic strategies.
Collapse
Affiliation(s)
- Gaël Chételat
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Unité 1237, Groupement d'Intérêt Public Cyceron, Caen, France.
| | - Javier Arbizu
- Department of Nuclear Medicine, University of Navarra, Clinica Universidad de Navarra, Pamplona, Spain
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig, Germany
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and NIMTlab, Geneva University, Geneva, Switzerland
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Silvia Morbelli
- Nuclear Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genova, Italy
| | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Institutes of Neurology and Healthcare Engineering, University College London, London, UK
| | - David J Brooks
- Institute of Neuroscience, Newcastle University, Newcastle, UK; Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | | | - Bruno Dubois
- Centre des Maladies Cognitives et Comportementales, University Hospital of Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne-Université, Paris, France
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway, Oslo; Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Giovanni B Frisoni
- Memory Clinic, Department of Rehabilitation and Geriatrics, Geneva University and University Hospitals, Geneva, Switzerland
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Brian F Hutton
- Institute of Nuclear Medicine, University College London, London, UK
| | | | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, USA
| | - Flavio Nobili
- UO Clinica Neurologica, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale Policlinico San Martino, Genova, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Child and Mother Health, University of Genoa, Genova, Italy
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Rik Ossenkoppele
- Department of Neurology, Alzheimer Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Wim J G Oyen
- Humanitas University and Humanitas Clinical and Research Center, Department of Nuclear Medicine, Milan, Italy; Rijnstate, Department of Radiology and Nuclear Medicine, Arnhem, Netherlands; Radboud UMC, Department of Radiology and Nuclear Medicine, Nijmegen, Netherlands
| | - Daniela Perani
- Vita-Salute San Raffaele University, Nuclear Medicine Unit, San Raffaele Hospital, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Gil D Rabinovici
- Departments of Neurology, Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Philip Scheltens
- Department of Neurology, Alzheimer Center, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC, Australia; School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at University College London, London, UK
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; German Center for Neurodegenerative Diseases, Bonn-Cologne, Germany; Institute of Neuroscience and Medicine, Molecular Organization of the Brain, Forschungszentrum Jülich, Germany
| |
Collapse
|
25
|
Perini G, Rodriguez-Vieitez E, Kadir A, Sala A, Savitcheva I, Nordberg A. Clinical impact of 18F-FDG-PET among memory clinic patients with uncertain diagnosis. Eur J Nucl Med Mol Imaging 2020; 48:612-622. [PMID: 32734458 PMCID: PMC7835147 DOI: 10.1007/s00259-020-04969-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
Purpose To assess the clinical impact and incremental diagnostic value of 18F-fluorodeoxyglucose (FDG-PET) among memory clinic patients with uncertain diagnosis. Methods The study population consisted of 277 patients who, despite extensive baseline cognitive assessment, MRI, and CSF analyses, had an uncertain diagnosis of mild cognitive impairment (MCI) (n = 177) or dementia (n = 100). After baseline diagnosis, each patient underwent an FDG-PET, followed by a post-FDG-PET diagnosis formulation. We evaluated (i) the change in diagnosis (baseline vs. post-FDG-PET), (ii) the change in diagnostic accuracy when comparing each baseline and post-FDG-PET diagnosis to a long-term follow-up (3.6 ± 1.8 years) diagnosis used as reference, and (iii) comparative FDG-PET performance testing in MCI and dementia conditions. Results FDG-PET led to a change in diagnosis in 86 of 277 (31%) patients, in particular in 57 of 177 (32%) MCI and in 29 of 100 (29%) dementia patients. Diagnostic change was greater than two-fold in the sub-sample of cases with dementia “of unclear etiology” (change in diagnosis in 20 of 32 (63%) patients). In the dementia group, after results of FDG-PET, diagnostic accuracy improved from 77 to 90% in Alzheimer’s disease (AD) and from 85 to 94% in frontotemporal lobar degeneration (FTLD) patients (p < 0.01). FDG-PET performed better in dementia than in MCI (positive likelihood ratios >5 and < 5, respectively). Conclusion Within a selected clinical population, FDG-PET has a significant clinical impact, both in early and differential diagnosis of uncertain dementia. FDG-PET provides significant incremental value to detect AD and FTLD over a clinical diagnosis of uncertain dementia. Electronic supplementary material The online version of this article (10.1007/s00259-020-04969-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Giulia Perini
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, 141 52, Stockholm, Sweden.,Center for Cognitive and Behavioral Disorders, IRCCS Mondino Foundation and Dept of Brain and Behavior, University of Pavia, 27100, Pavia, Italy
| | - Elena Rodriguez-Vieitez
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Ahmadul Kadir
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, 141 52, Stockholm, Sweden.,Theme Aging, The Aging Brain Unit, Karolinska University Hospital, 141 86, Stockholm, Sweden
| | - Arianna Sala
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Irina Savitcheva
- Medical Radiation Physics and Nuclear Medicine Imaging, Section for Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, 141 52, Stockholm, Sweden. .,Theme Aging, The Aging Brain Unit, Karolinska University Hospital, 141 86, Stockholm, Sweden.
| |
Collapse
|
26
|
Cacabelos R. Pharmacogenomics of Cognitive Dysfunction and Neuropsychiatric Disorders in Dementia. Int J Mol Sci 2020; 21:E3059. [PMID: 32357528 PMCID: PMC7246738 DOI: 10.3390/ijms21093059] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
Symptomatic interventions for patients with dementia involve anti-dementia drugs to improve cognition, psychotropic drugs for the treatment of behavioral disorders (BDs), and different categories of drugs for concomitant disorders. Demented patients may take >6-10 drugs/day with the consequent risk for drug-drug interactions and adverse drug reactions (ADRs >80%) which accelerate cognitive decline. The pharmacoepigenetic machinery is integrated by pathogenic, mechanistic, metabolic, transporter, and pleiotropic genes redundantly and promiscuously regulated by epigenetic mechanisms. CYP2D6, CYP2C9, CYP2C19, and CYP3A4/5 geno-phenotypes are involved in the metabolism of over 90% of drugs currently used in patients with dementia, and only 20% of the population is an extensive metabolizer for this tetragenic cluster. ADRs associated with anti-dementia drugs, antipsychotics, antidepressants, anxiolytics, hypnotics, sedatives, and antiepileptic drugs can be minimized by means of pharmacogenetic screening prior to treatment. These drugs are substrates, inhibitors, or inducers of 58, 37, and 42 enzyme/protein gene products, respectively, and are transported by 40 different protein transporters. APOE is the reference gene in most pharmacogenetic studies. APOE-3 carriers are the best responders and APOE-4 carriers are the worst responders; likewise, CYP2D6-normal metabolizers are the best responders and CYP2D6-poor metabolizers are the worst responders. The incorporation of pharmacogenomic strategies for a personalized treatment in dementia is an effective option to optimize limited therapeutic resources and to reduce unwanted side-effects.
Collapse
Affiliation(s)
- Ramon Cacabelos
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, 15165-Bergondo, Corunna, Spain
| |
Collapse
|