1
|
Vanderlinden G, Vandenberghe R, Vandenbulcke M, Van Laere K. The Current Role of Tau PET Imaging in Neurodegeneration. Semin Nucl Med 2025:S0001-2998(25)00031-5. [PMID: 40263023 DOI: 10.1053/j.semnuclmed.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Neurodegenerative tauopathies are characterized by the pathological hyperphosphorylation of tau proteins that subsequently form aggregates. Tau PET tracers with affinity to bind these pathological tau aggregates have been developed to measure disease progression and to support therapeutic drug development. In this review, we summarize the pathophysiology of tau throughout the range of neurodegenerative tauopathies. We outline the available first- and second-generation tau PET tracers, with a focus on new tau PET tracer developments, and discuss the quantification of tau PET images. Next, we summarize how tau PET relates to cerebrospinal fluid and plasma tau biomarkers. Finally, we review the current recommendations on the clinical use of tau PET versus fluid tau biomarkers in diagnosis, prognosis and treatment development.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Research Group Psychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Division of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Takasaki A, Nishio Y, Satake Y, Kobayashi M, Sakai M, Mori K, Ishii K, Ikeda M. Primary Tauopathy With Logopenic/Semantic Mixed Progressive Aphasia and Frontotemporal Dementia-like Behavior. Cogn Behav Neurol 2025:00146965-990000000-00086. [PMID: 40249666 DOI: 10.1097/wnn.0000000000000391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 02/24/2025] [Indexed: 04/20/2025]
Abstract
Here we present the cases of two individuals with language and behavioral symptoms indicative of the early clinical manifestations of both Alzheimer disease (AD) and frontotemporal lobar degeneration (FTLD). Phonological language symptoms similar to those evident in logopenic variant primary progressive aphasia suggested AD pathology, while semantic impairment and behavioral changes (ie, abnormal eating behavior and disinhibition) suggested a diagnosis of FTLD. Multimodal neuroimaging studies revealed underlying neuropathology indicative of primary tauopathy with presumable 3/4-repeat isoform, devoid of amyloid deposition. We suggest that these cases may represent a previously unrecognized syndrome associated with non-Alzheimer primary tauopathy.
Collapse
Affiliation(s)
- Akihiro Takasaki
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshiyuki Nishio
- Department of Behavioral Neurology and Neuropsychiatry, Osaka University United Graduate School of Child Development, Suita, Japan
| | - Yuto Satake
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Matasaburo Kobayashi
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Mariko Sakai
- Department of Rehabilitation, Yukoukai General Hospital, Ibaraki, Japan
| | - Kohji Mori
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazunari Ishii
- Department of Radiology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
3
|
Cummings JL, Teunissen CE, Fiske BK, Le Ber I, Wildsmith KR, Schöll M, Dunn B, Scheltens P. Biomarker-guided decision making in clinical drug development for neurodegenerative disorders. Nat Rev Drug Discov 2025:10.1038/s41573-025-01165-w. [PMID: 40185982 DOI: 10.1038/s41573-025-01165-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative disorders are characterized by complex neurobiological changes that are reflected in biomarker alterations detectable in blood, cerebrospinal fluid (CSF) and with brain imaging. As accessible proxies for processes that are difficult to measure, biomarkers are tools that hold increasingly important roles in drug development and clinical trial decision making. In the past few years, biomarkers have been the basis for accelerated approval of new therapies for Alzheimer disease and amyotrophic lateral sclerosis as surrogate end points reasonably likely to predict clinical benefit.Blood-based biomarkers are emerging for Alzheimer disease and other neurodegenerative disorders (for example, Parkinson disease, frontotemporal dementia), and some biomarkers may be informative across multiple disease states. Collection of CSF provides access to biomarkers not available in plasma, including markers of synaptic dysfunction and neuroinflammation. Molecular imaging is identifying an increasing array of targets, including amyloid plaques, neurofibrillary tangles, inflammation, mitochondrial dysfunction and synaptic density. In this Review, we consider how biomarkers can be implemented in clinical trials depending on their context of use, including providing information on disease risk and/or susceptibility, diagnosis, prognosis, pharmacodynamic outcomes, monitoring, prediction of response to therapy and safety. Informed choice of increasingly available biomarkers and rational deployment in clinical trials support drug development decision making and de-risk the drug development process for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Brian K Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Göteborg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Philip Scheltens
- Alzheimer's Center Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
- EQT Group, Dementia Fund, Stockholm, Sweden
| |
Collapse
|
4
|
Zeiss CJ, Huttner A, Nairn AC, Arnsten A, Datta D, Strittmatter SM, Wyk BV, Duque A. The neuropathologic basis for translational biomarker development in the macaque model of late-onset Alzheimer's disease. J Alzheimers Dis 2025; 104:1243-1258. [PMID: 40095666 DOI: 10.1177/13872877251323787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
BackgroundAccurate placement of the macaque within the Alzheimer's disease (AD) research framework is essential to discover early-stage predictive biomarkers.ObjectiveTo assess utility of the aging macaque in advancing translational biomarker development for preclinical AD, we evaluated relative signal strength of comparable neuropathologic phenomena in macaques and patients.MethodsWe compared pathology in patient and macaque formalin-fixed paraffin embedded (FFPE) tissues using identical criteria. We quantified expression of amyloid-β (Aβ), pTau, and inflammatory and senescence markers across species. Distribution of AD-relevant markers were compared in FFPE and perfused frozen macaque brain to assess expression of labile proteins that could inform in-life fluid biomarkers.ResultsAβ pathology in macaques closely approximated patient pathology. Complex plaque composition in macaques implied significant disruption of synaptic connectivity. In FFPE tissue, pretangle pTau immunoreactivity placed the macaque in Braak Stage 1b. In perfused frozen tissue, soluble pTau distribution approximated Braak Stage III-IV. In macaque, Aβ, pTau, and acetylcholinesterase labeling co-localized to AD-vulnerable circuits. Significant association of glial fibrillary acidic protein with Aβ occurred in humans only. The senescence marker p16 correlated positively with pTau expression and negatively with Aβ in patients only. Macaques lacked neuropathologic co-morbidities.ConclusionsAD-relevant neuropathologic signals in the macaque support biomarker discovery in the areas of Aβ plaque evolution and associated synaptic disruption as well as early-stage tau phosphorylation. Relative protection from accumulation of senescence markers, fibrillar tau and neuropathologic co-morbidities in macaque implicate species difference in rates of biological brain aging. We provide over 4000 digital slides for further study.
Collapse
Affiliation(s)
- Caroline J Zeiss
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Amy Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Dibyadeep Datta
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen M Strittmatter
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
5
|
Kling A, Kusche-Palenga J, Palleis C, Jäck A, Bernhardt AM, Frontzkowski L, Roemer SN, Slemann L, Zaganjori M, Scheifele M, Paeger L, Bischof GN, van Eimeren T, Drzezga A, Sabri O, Rullmann M, Barthel H, Levin J, Herms J, Franzmeier N, Höglinger G, Roeber S, Brendel M, Gnörich J. Exploring the origins of frequent tau-PET signal in vermal and adjacent regions. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07199-x. [PMID: 40100387 DOI: 10.1007/s00259-025-07199-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Off-target binding remains a significant challenge in tau-PET neuroimaging. While off-targets including monoamine oxidase enzymes and neuromelanin-containing cells have been identified, recent studies indicated a relevant binding of novel tau tracers to melanin-containing structures. To date, little is known about the effect of melanocytes in the meninges on tracer signals in brain PET data. Thus, we aimed to identify the target structure causal for the frequently observed [18F]PI-2620 PET signal in the vermis and adjacent cerebellar regions. METHODS 274 participants underwent dynamic [18F]PI-2620 tau-PET: 3/4R-tauopathies (n = 85), 4R-tauopathies (n = 147), tau-negative disease controls (n = 24), and healthy controls (n = 18). Standardized uptake value ratio (SUVR) and kinetic parameters including the distribution volume ratio (DVR), tracer clearance (k2) and relative perfusion (R1), were compared among the cohorts and sexes using the Automated Anatomical Labelling (AAL) atlas. Age and p-Tau levels in cerebrospinal fluid (CSF) were assessed for their relationship with vermal tau-PET signal. Furthermore, we combined autoradiographic and histochemical experiments on post-mortem brain tissue of deceased patients (n = 9). RESULTS Male participants revealed higher mean vermal [18F]PI-2620 DVR (0.95 ± 0.13; vs. females 0.88 ± 0.10, p < 0.0001). Sex-related differences were most pronounced in the 3/4R-tauopathy cohort (p < 0.0001). Mean SUVRVer/Cbl, k2 and correlation analyses of kinetic parameters did not differ among groups. Histological assessments revealed co-localization of leptomeningeal pigmented cells with strong autoradiography signal spots within the vermal fissures. Tau-related autoradiography signals, age or p-Tau levels did not correlate significantly with tau-PET signals. Iron deposits did not cause relevant autoradiography signals in the vermis. CONCLUSION Leptomeningeal melanocytes are the primary target structure for [18F]PI-2620 PET binding in anterior vermis, whereas iron and tau deposits do not contribute significantly.
Collapse
Affiliation(s)
- Agnes Kling
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Julia Kusche-Palenga
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Alexander Jäck
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Alexander M Bernhardt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Lukas Frontzkowski
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian N Roemer
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Luna Slemann
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Maximilian Scheifele
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Lars Paeger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Gérard N Bischof
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Research Center Jülich, Jülich, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Research Center Jülich, Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Research Center Jülich, Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Michael Rullmann
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Center of Neuropathology and Prion Research, University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, The Sahlgrenska Academy, Mölndal and Gothenburg, Sweden
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sigrun Roeber
- Center of Neuropathology and Prion Research, University of Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
| |
Collapse
|
6
|
Ghatamaneni S, Coleman C, Shin I, Bruinsma T, Scott N, Lee J, Fang P, Min HK, Moloney CM, Wood AC, Constantopoulos E, Reichard RR, Schwarz CG, Jones DT, Graff-Radford J, Knopman DS, Jack CR, Petersen RC, Dickson DW, Murray ME, Lowe VJ. High resolution autoradiography of [ 18F]MK-6240 and [ 18F]Flortaucipir shows similar neurofibrillary tangle binding patterns preferentially recognizing middling neurofibrillary tangle maturity. Acta Neuropathol 2025; 149:26. [PMID: 40085238 PMCID: PMC11909026 DOI: 10.1007/s00401-025-02864-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/08/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Recent developments in tau positron emission tomography (PET) radiotracers have enhanced the visualization of tau aggregates in Alzheimer's disease (AD). The maturity level of neurofibrillary tangles can affect its recognition by biomarkers. Early detection of tau aggregates regarding tangle pathology is of interest in early diagnosis and comparison of tau radiotracers in this aspect is important. This study focused on head to head pathologic comparison of [18F]MK-6240 and [18F]Flortaucipir postmortem binding as seen on high resolution autoradiography as compared to CP-13 (early tangle maturity) and PHF-1 (middling tangle maturity) immunohistochemistry (IHC) to evaluate the tangle maturity pathology specificity of binding for tau aggregates in AD, atypical AD and non-AD tauopathies. Analyses were performed on serial 5 μm formalin-fixed paraffin-embedded human brain sections acquired from the Mayo Clinic brain bank. Visual assessment of colocalization with IHC as well as quantitative analyses were used. Evaluation of the tracers' off-target binding profiles were performed. Both tracers had similar binding properties for tau aggregates with preference to middling tangle maturity as shown by comparison to immunohistochemical distributions. Both the tracers showed strong binding to AD tau aggregates and no or minimal binding to non-AD tauopathies which corroborates with other studies.
Collapse
Affiliation(s)
| | | | - Ian Shin
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Tyler Bruinsma
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nancy Scott
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jeyeon Lee
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biomedical Engineering, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Ping Fang
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Hoon-Ki Min
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Ashley C Wood
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Ross R Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
7
|
Alosco ML, Mejía Pérez J, Culhane JE, Shankar R, Nowinski CJ, Bureau S, Mundada N, Smith K, Amuiri A, Asken B, Groh JR, Miner A, Pettway E, Mosaheb S, Tripodis Y, Windon C, Mercier G, Stern RA, Grinberg LT, Soleimani-Meigooni DN, Christian BT, Betthauser TJ, Stein TD, McKee AC, Mathis CA, Abrahamson EE, Ikonomovic MD, Johnson SC, Mez J, La Joie R, Schonhaut D, Rabinovici GD. 18F-MK-6240 tau PET in patients at-risk for chronic traumatic encephalopathy. Mol Neurodegener 2025; 20:23. [PMID: 39994806 PMCID: PMC11852567 DOI: 10.1186/s13024-025-00808-1] [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: 08/26/2024] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Molecular biomarkers of chronic traumatic encephalopathy (CTE) are lacking. We evaluated 18F-MK-6240 tau PET as a biomarker for CTE. Two studies were done: (1) 3H-MK-6240 autoradiography and an in-vitro brain homogenate binding studies on postmortem CTE tissue, (2) an in-vivo 18F-MK-6240 tau PET study in former American football players. METHODS Autoradiography and in-vitro binding studies were done using 3H-MK-6240 on frozen temporal and frontal cortex tissue from six autopsy cases with stage III CTE compared to Alzheimer's disease. Thirty male former National Football League (NFL) players with cognitive concerns (mean age = 58.9, SD = 7.8) completed tau (18F-MK-6240) and Aβ (18F-Florbetapir) PET. Controls included 39 Aβ-PET negative, cognitively normal males (mean age = 65.7, SD = 6.3). 18F-MK-6240 SUVr images were created using 70-90 min post-injection data with inferior cerebellar gray matter as the reference. We compared SUVr between players and controls using voxelwise and region-of-interest approaches. Correlations between 18F-MK-6240 SUVr and cognitive scores were tested. RESULTS All six CTE stage III cases had Braak NFT stage III but no neuritic plaques. Two had Thal Phase 1 for Aβ; one showed a laminar pattern of 3H-MK-6240 autoradiography binding in the superior temporal cortex and less so in the dorsolateral frontal cortex, corresponding to tau-immunoreactive lesions detected using the AT8 antibody (pSer202/pThr205 tau) in adjacent tissue sections. The other CTE cases had low frequencies of cortical tau-immunoreactive deposits and no well-defined autoradiography binding. In-vitro 3H-MK-6240 binding studies to CTE brain homogenates in the case with autoradiography signal indicated high binding affinity (KD = 2.0 ± 0.9 nM, Bmax = 97 ± 24 nM, n = 3). All NFL players had negative Aβ-PET. There was variable, low-to-intermediate intensity 18F-MK-6240 uptake across participants: 16 had no cortical signal, 7 had medial temporal lobe (MTL) uptake, 2 had frontal uptake, and 4 had MTL and frontal uptake. NFL players had higher SUVr in the entorhinal cortex (d = 0.86, p = 0.001), and the parahippocampal gyrus (d = 0.39, p = 0.08). Voxelwise regressions showed increased uptake in NFL players in two bilateral anterior MTL clusters (p < 0.05 FWE). Higher parahippocampal and frontal-temporal SUVrs correlated with worse memory (r = -0.38, r = -0.40) and semantic fluency (r = -0.38, r = -0.48), respectively. CONCLUSION We present evidence of 3H-MK-6240 in-vitro binding to post-mortem CTE tissue homogenates and in vivo 18F-MK-6240 PET binding in the MTL among a subset of participants. Additional studies in larger samples and PET-to-autopsy correlations are required to further elucidate the potential of 18F-MK-6240 to detect tau pathology in CTE.
Collapse
Affiliation(s)
- Michael L Alosco
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston Medical Center, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jhony Mejía Pérez
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Julia E Culhane
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ranjani Shankar
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Nidhi Mundada
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Karen Smith
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Alinda Amuiri
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Breton Asken
- Department of Clinical & Health Psychology, 1Florida Alzheimer's Disease Research Center, Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Jenna R Groh
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Annalise Miner
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Erika Pettway
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sydney Mosaheb
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Charles Windon
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Gustavo Mercier
- Molecular Imaging and Nuclear Medicine, Boston Medical Center, Boston, MA, USA
| | - Robert A Stern
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lea T Grinberg
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - David N Soleimani-Meigooni
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Bradley T Christian
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Thor D Stein
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S.Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- Department of Psychiatry and Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ann C McKee
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S.Department of Veteran Affairs, VA Boston Healthcare System, Jamaica Plain, MA, USA
- Department of Psychiatry and Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric E Abrahamson
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
- School of Medicine and Public Health, Wisconsin Alzheimer's Institute, University of Wisconsin-Madison, Madison, USA
| | - Jesse Mez
- Department of Neurology, Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Renaud La Joie
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Daniel Schonhaut
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Department of Neurology, Alzheimer's Disease Research Center, Memory & Aging Center, University of California San Francisco, San Francisco, CA, USA.
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
- University of California, San Francisco (UCSF), Memory and Aging Center MC: 1207, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA.
| |
Collapse
|
8
|
Gogola A, Lopresti BJ, Minhas DS, Lopez O, Cohen A, Villemagne VL. Tau Imaging: Use and Implementation in New Diagnostic and Therapeutic Paradigms for Alzheimer's Disease. Geriatrics (Basel) 2025; 10:27. [PMID: 39997526 PMCID: PMC11855481 DOI: 10.3390/geriatrics10010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/24/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Alzheimer's disease (AD) affects an estimated 6.9 million older adults in the United States and is projected to impact as many as 13.8 million people by 2060. As studies continue to search for ways to combat the development and progression of AD, it is imperative to ensure that confident diagnoses can be made before the onset of severe clinical symptoms and new therapies can be evaluated effectively. Tau positron emission tomography (PET) has emerged as one method that may be capable of both, given its ability to recognize the presence of tau, a primary pathologic hallmark of AD; its usefulness in determining the spatial distribution of tau, which is necessary for differentiating AD from other tauopathies; and its association with measures of cognition. This review aims to evaluate the scope of tau PET's utility in clinical trials and practice. Firstly, the potential of using tau PET for differential diagnoses, distinguishing AD from other dementias, is considered. Next, the value of tau PET as a tool for staging disease progression is investigated. Finally, tau PET as a prognostic method for identifying the individuals most at risk of cognitive decline and, therefore, most in need of, and likely to benefit from, intervention, is discussed.
Collapse
Affiliation(s)
- Alexandra Gogola
- Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.J.L.); (D.S.M.)
| | - Brian J. Lopresti
- Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.J.L.); (D.S.M.)
| | - Davneet S. Minhas
- Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.J.L.); (D.S.M.)
| | - Oscar Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Ann Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (A.C.); (V.L.V.)
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (A.C.); (V.L.V.)
| |
Collapse
|
9
|
Olafson E, Tonietto M, Klein G, Teng E, Stephens AW, Russell DS, Pickthorn K, Sanabria Bohorquez S. In Vivo Head-to-Head Comparison of [ 18F]GTP1 with [ 18F]MK-6240 and [ 18F]PI-2620 in Alzheimer Disease. J Nucl Med 2025; 66:277-285. [PMID: 39746756 PMCID: PMC11800736 DOI: 10.2967/jnumed.124.268623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Alzheimer disease (AD) is characterized by the accumulation of tau neurofibrillary tangles that can be labeled with PET tracers. Multiple tau PET tracers have been used in clinical studies, including [18F]GTP1, [18F]PI-2620, and [18F]MK-6240. Standardized harmonization scales for comparing tau PET signals across tracers are currently under development and can be informed by comparisons of signals between tracers in both target and off-target regions of the brain. Methods: We conducted a head-to-head study comparing [18F]GTP1 with [18F]PI-2620 and [18F]MK-6240 in terms of dynamic range, magnitude of uptake, and correlation between tracers in participants with normal cognition and prodromal to mild AD. Results: [18F]GTP1 exhibited retention patterns that correlated with [18F]PI-2620 and [18F]MK-6240 for all Braak regions (except Braak II). Differences in tracer binding in AD target regions were relatively small, and off-target binding profiles were unique to each tracer. Conclusion: Our findings indicate that [18F]GTP1, [18F]PI-2620, and [18F]MK-6240 display similar uptake patterns in AD patients, suggesting that they detect the same tau pathology. However, the tracer-specific off-target signal distribution may impact their direct comparability, and for some use cases, tracer-specific considerations should be taken into account in the development of a standardized harmonization scale for tau PET.
Collapse
Affiliation(s)
- Emily Olafson
- gRED, Genentech, Inc., South San Francisco, California;
| | | | - Gregory Klein
- pRED, F. Hoffmann-La Roche, Ltd., Basel, Switzerland
| | - Edmond Teng
- gRED, Genentech, Inc., South San Francisco, California
| | | | | | | | | |
Collapse
|
10
|
Aumont E, Bedard MA, Bussy A, Arias JF, Tissot C, Hall BJ, Therriault J, Rahmouni N, Stevenson J, Servaes S, Macedo AC, Vitali P, Poltronetti NM, Fliaguine O, Trudel L, Gauthier S, Chakravarty MM, Rosa-Neto P. Hippocampal atrophy over two years in relation to tau, amyloid-β and memory in older adults. Neurobiol Aging 2025; 146:48-57. [PMID: 39631245 DOI: 10.1016/j.neurobiolaging.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/27/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
In this longitudinal brain imaging study, we aimed to characterize hippocampal tau accumulation and subfield atrophy relative to cortical amyloid-β and memory performance. We measured tau-PET in regions associated with Braak stages I to VI, global amyloid-PET burden, hippocampal subfield volumes and memory assessments from 173 participants aged 55-85. Eighty-six of these participants were tested again two years later. Tau-PET change in the Braak II region, corresponding to the hippocampus and the entorhinal cortex, was significantly associated with the cornu ammonis 1 (CA1) atrophy and memory score. This CA1 atrophy did not significantly mediate the association between tau and memory, nor did global amyloid-PET burden correlate with tau-PET changes in the Braak II region. Longitudinal hippocampal tau accumulation is amyloid-β-independent and co-localized with subfield atrophy. As tau-associated memory decline seems to be independent from hippocampal atrophy, other mechanisms could contribute to the deficit.
Collapse
Affiliation(s)
- Etienne Aumont
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3P2, Canada; McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Marc-André Bedard
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3P2, Canada; McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Aurélie Bussy
- Cerebral Imaging Center, Douglas Research Center, Montreal, QC H4H 1R3, Canada; Computational Brain Anatomy (CoBrALab) Laboratory, Montreal, QC H4H 1R3, Canada
| | - Jaime Fernandez Arias
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Cecile Tissot
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Brandon J Hall
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Joseph Therriault
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Nesrine Rahmouni
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Jenna Stevenson
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Stijn Servaes
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Arthur C Macedo
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Paolo Vitali
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | | | - Olga Fliaguine
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal, QC H2X 3P2, Canada
| | - Lydia Trudel
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Serge Gauthier
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada
| | - Mallar M Chakravarty
- Cerebral Imaging Center, Douglas Research Center, Montreal, QC H4H 1R3, Canada; Computational Brain Anatomy (CoBrALab) Laboratory, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
| | - Pedro Rosa-Neto
- McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC H4H 1R3, Canada; Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Department of neurology and neurosurgery, McGill University, Montréal, QC H3A 1A1, Canada.
| |
Collapse
|
11
|
Asken BM, Brett BL, Barr WB, Banks S, Wethe JV, Dams-O'Connor K, Stern RA, Alosco ML. Chronic traumatic encephalopathy: State-of-the-science update and narrative review. Clin Neuropsychol 2025:1-25. [PMID: 39834035 DOI: 10.1080/13854046.2025.2454047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE The long-recognized association of brain injury with increased risk of dementia has undergone significant refinement and more detailed study in recent decades. Chronic traumatic encephalopathy (CTE) is a specific neurodegenerative tauopathy related to prior exposure to repetitive head impacts (RHI). We aim to contextualize CTE within a historical perspective and among emerging data which highlights the scientific and conceptual evolution of CTE-related research in parallel with the broader field of neurodegenerative disease and dementia. METHODS We provide a narrative state-of-the-science update on CTE neuropathology, clinical manifestations, biomarkers, different types and patterns of head impact exposure relevant for CTE, and the complicated influence of neurodegenerative co-pathology on symptoms. CONCLUSIONS Now almost 20 years since the initial case report of CTE in a former American football player, the field of CTE continues evolving with increasing clarity but also several ongoing controversies. Our understanding of CTE neuropathology outpaces that of disease-specific clinical correlates or the development of in-vivo biomarkers. Diagnostic criteria for symptoms attributable to CTE are still being validated, but leveraging increasingly available biomarkers for other conditions like Alzheimer's disease may be helpful for informing the CTE differential diagnosis. As diagnostic refinement efforts advance, clinicians should provide care and/or referrals to providers best suited to treat an individual patient's clinical symptoms, many of which have evidence-based behavioral treatment options that are etiologically agnostic. Several ongoing research initiatives and the gradual accrual of gold standard clinico-pathological data will pay dividends for advancing the many existing gaps in the field of CTE.
Collapse
Affiliation(s)
- Breton M Asken
- Department of Clinical and Health Psychology, University of Florida, 1Florida Alzheimer's Disease Research Center, Gainesville, FL, USA
| | - Benjamin L Brett
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WS, USA
| | - William B Barr
- Department of Neurology, New York University Langone Health Medical Center, New York, NY, USA
| | - Sarah Banks
- Department of Neuroscience, University of California San Diego, La Jolla, CA, USA
| | - Jennifer V Wethe
- Departments of Psychiatry and Psychology, Mayo Clinic, Phoenix, AZ, USA
| | - Kristen Dams-O'Connor
- Departments of Rehabilitation Medicine and Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert A Stern
- Departments of Neurology, Neurosurgery, and Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston University CTE and Alzheimer's Disease Research Centers, Boston, MA, USA
| | - Michael L Alosco
- Departments of Neurology and Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston University CTE and Alzheimer's Disease Research Centers, Boston, MA, USA
| |
Collapse
|
12
|
Wongso H, Harada R, Furumoto S. Current Progress and Future Directions in Non-Alzheimer's Disease Tau PET Tracers. ACS Chem Neurosci 2025; 16:111-127. [PMID: 39762194 DOI: 10.1021/acschemneuro.4c00319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) and non-AD tauopathies are dominant public health issues driven by several factors, especially in the aging population. The discovery of first-generation radiotracers, including [18F]FDDNP, [11C]PBB3, [18F]flortaucipir, and the [18F]THK series, for the in vivo detection of tauopathies has marked a significant breakthrough in the fields of neuroscience and radiopharmaceuticals, creating a robust new category of labeled compounds: tau positron emission tomography (PET) tracers. Subsequently, other tau PET tracers with improved binding properties have been developed using various chemical scaffolds to target the three-repeat/four-repeat (3R/4R) tau folds in AD. In 2020, [18F]flortaucipir was approved by the U.S. Food and Drug Administration for PET imaging of tau pathology in adult patients with cognitive deficits undergoing evaluation for AD. Despite remarkable progress in the development of AD tau PET tracers, imaging agents for rare non-AD tauopathies (4R tauopathies [predominantly expressing a 4R tau isoform], involved in progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, and globular glial tauopathy, and 3R tauopathies [predominantly expressing a 3R tau isoform], such as Pick's disease) remain substantially underdeveloped. In this review, we discuss recent progress in tau PET tracer development, with particular emphasis on clinically validated tracers for AD and their potential use for non-AD tauopathies. Additionally, we highlight the critical need for further development of tau PET tracers specifically designed for non-AD tauopathies, an area that remains significantly underexplored despite its importance in advancing the understanding and diagnosis of these disorders.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten 15314, Indonesia
| | - Ryuichi Harada
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Shozo Furumoto
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Miyagi 980-0845, Japan
- Graduate School of Pharmaceutical Science, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
13
|
Vande Casteele T, Laroy M, Van Cauwenberge M, Vanderlinden G, Vansteelandt K, Koole M, Dupont P, Van Den Bossche M, Van den Stock J, Bouckaert F, Van Laere K, Emsell L, Vandenbulcke M. Late Life Depression is Not Associated With Alzheimer-Type Tau: Preliminary Evidence From a Next-Generation Tau Ligand PET-MR Study. Am J Geriatr Psychiatry 2025; 33:47-62. [PMID: 39107144 DOI: 10.1016/j.jagp.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVE To investigate whether tau accumulation is higher in late life depression (LLD) compared to non-depressed cognitively unimpaired (CU) older adults. To situate these findings in the neurodegeneration model of LLD by assessing group differences in tau and grey matter volume (GMV) between LLD, non-depressed CU and mild cognitive impairment due to Alzheimer's Disease (MCI). DESIGN Monocentric, cross-sectional study. SETTING University Psychiatric hospital, memory clinic and outpatient neurology practice. PARTICIPANTS A total of 102 adults over age 60, of whom 19 currently depressed participants with LLD, 19 with MCI and 36 non-depressed CU participants completed neuropsychological testing and tau PET-MR imaging. MEASUREMENTS PET-MRI: 18F-MK-6240 tracer SUVR for tau assessment; 3D T1-weighted structural MRI derived GMV in seven brain regions (temporal, cingulate, prefrontal and parietal regions); amyloid PET to assess amyloid positivity; Neuropsychological test scores: MMSE, RAVLT, GDS, MADRS. ANCOVA and Spearman's rank correlations to investigate group differences in tau and GMV, and correlations with neuropsychological test scores respectively. RESULTS Compared to non-depressed CU participants, LLD patients showed lower GMV in temporal and anterior cingulate regions but similar tau accumulation and amyloid positivity rate. In contrast, MCI patients had significantly higher tau accumulation in all regions. Tau did not correlate with any neuropsychological test scores in LLD. CONCLUSION Our findings suggest AD-type tau is not higher in LLD compared to non-depressed, cognitively unimpaired older adults and appears unlikely to contribute to lower gray matter volume in LLD, further underscoring the need to distinguish major depressive disorder from depressive symptoms occurring in early AD.
Collapse
Affiliation(s)
- Thomas Vande Casteele
- Department of Neurosciences, Neuropsychiatry (TVC, ML, MVC, MVDB, JVDS, FB, LE, MV), KU Leuven, Leuven Brain Institute, Leuven, Belgium.
| | - Maarten Laroy
- Department of Neurosciences, Neuropsychiatry (TVC, ML, MVC, MVDB, JVDS, FB, LE, MV), KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Margot Van Cauwenberge
- Department of Neurosciences, Neuropsychiatry (TVC, ML, MVC, MVDB, JVDS, FB, LE, MV), KU Leuven, Leuven Brain Institute, Leuven, Belgium; Department of Neurology (MVC), University Hospitals Leuven, Leuven, Belgium
| | - Greet Vanderlinden
- Department of Imaging and Pathology, Nuclear Medicine (GV, MK, KVL), KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Kristof Vansteelandt
- Geriatric Psychiatry (KV, MVDB, JVDS, FB, LE), University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Michel Koole
- Department of Imaging and Pathology, Nuclear Medicine (GV, MK, KVL), KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Patrick Dupont
- Department of Neurosciences, Laboratory for Cognitive Neurology (PD), KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Maarten Van Den Bossche
- Department of Neurosciences, Neuropsychiatry (TVC, ML, MVC, MVDB, JVDS, FB, LE, MV), KU Leuven, Leuven Brain Institute, Leuven, Belgium; Geriatric Psychiatry (KV, MVDB, JVDS, FB, LE), University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Jan Van den Stock
- Department of Neurosciences, Neuropsychiatry (TVC, ML, MVC, MVDB, JVDS, FB, LE, MV), KU Leuven, Leuven Brain Institute, Leuven, Belgium; Geriatric Psychiatry (KV, MVDB, JVDS, FB, LE), University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Filip Bouckaert
- Department of Neurosciences, Neuropsychiatry (TVC, ML, MVC, MVDB, JVDS, FB, LE, MV), KU Leuven, Leuven Brain Institute, Leuven, Belgium; Geriatric Psychiatry (KV, MVDB, JVDS, FB, LE), University Psychiatric Center KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Department of Imaging and Pathology, Nuclear Medicine (GV, MK, KVL), KU Leuven, Leuven Brain Institute, Leuven, Belgium; Department of Nuclear Medicine (KVL), University Hospitals Leuven, Leuven, Belgium
| | - Louise Emsell
- Department of Neurosciences, Neuropsychiatry (TVC, ML, MVC, MVDB, JVDS, FB, LE, MV), KU Leuven, Leuven Brain Institute, Leuven, Belgium; Geriatric Psychiatry (KV, MVDB, JVDS, FB, LE), University Psychiatric Center KU Leuven, Leuven, Belgium; Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Department of Neurosciences, Neuropsychiatry (TVC, ML, MVC, MVDB, JVDS, FB, LE, MV), KU Leuven, Leuven Brain Institute, Leuven, Belgium; Geriatric Psychiatry (KV, MVDB, JVDS, FB, LE), University Psychiatric Center KU Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Sakurai K, Kaneda D, Morimoto S, Uchida Y, Inui S, Shang C, Kimura Y, Cai C, Kato T, Ito K, Hashizume Y. Medial temporal atrophy predicts the limbic comorbidities in lewy body disease. Neuroradiology 2025; 67:65-77. [PMID: 39531077 DOI: 10.1007/s00234-024-03502-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Although neuropathological comorbidities, including Alzheimer's disease neuropathological change (AD-NC) and limbic-predominant age-related TAR DNA-binding protein 43encephalopathy neuropathological change (LATE-NC), are associated with medial temporal atrophy in patients with Lewy body disease (LBD), the diagnostic performance of magnetic resonance imaging (MRI)-derived indices remains unclear. This study aimed to investigate the diagnostic performance of MRI-derived indices representing medial temporal atrophy in differentiating between LBD with AD-NC and/or LATE-NC (mixed LBD [mLBD]) and without these comorbidities (pure LBD [pLBD]). METHODS This study included 24 and 16 patients with pathologically confirmed mLBD and pLBD, respectively. In addition to the well-known medial temporal atrophy and entorhinal cortex atrophy (ERICA) scores, the cross-sectional areas of the bilateral entorhinal cortices/parahippocampal gyri (ABEP) were segmented manually. RESULTS Even incorporating various covariates such as age at MRI examination, sex, argyrophilic grain, the MRI-derived indices, especially ABEP, significantly correlated with the severity of AD-NC, and showed a trend of correlation with LATE-NC. For the differentiation between all mLBD and pLBD, the ERICA score and ABEP demonstrated higher diagnostic performance (area under the receiver-operating-characteristic curve [AUC] of 0.80 and 0.87, respectively). Additionally, the highest diagnostic performance for ABEP (AUC, 0.94; sensitivity, 100%; specificity, 88.9%; accuracy, 96%) was observed in differentiating between pLBD and mLBD with two comorbidities (AD-NC and LATE-NC). CONCLUSION In patients with pathologically confirmed LBD, medial temporal atrophy was significantly correlated with AD-NC, and showed a trend of correlation with LATE-NC. Moreover, MRI-derived indices indicative of medial temporal atrophy were useful in diagnosing these comorbidities.
Collapse
Affiliation(s)
- Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan.
| | - Daita Kaneda
- Choju Medical Institute, Fukushimura Hospital, Aichi, Japan
| | - Satoru Morimoto
- Keio University Regenerative Medicine Research Center, Kanagawa, Japan
| | - Yuto Uchida
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shohei Inui
- Department of Radiology, The University of Tokyo, Tokyo, Japan
| | - Cong Shang
- Department of Radiology, Fujita Health University, Aichi, Japan
- Department of Radiology, Fujita Health University, Toyoake, Aichi, Japan
| | - Yasuyuki Kimura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Chang Cai
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Takashi Kato
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Kengo Ito
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | | |
Collapse
|
15
|
Keir G, Roytman M, Mashriqi F, Shahsavarani S, Franceschi AM. Atypical Parkinsonian Syndromes: Structural, Functional, and Molecular Imaging Features. AJNR Am J Neuroradiol 2024; 45:1865-1877. [PMID: 39209485 PMCID: PMC11630880 DOI: 10.3174/ajnr.a8313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/16/2024] [Indexed: 09/04/2024]
Abstract
Atypical parkinsonian syndromes, also known as Parkinson-plus syndromes, are a heterogeneous group of movement disorders, including dementia with Lewy bodies (DLB), progressive supranuclear palsy (PSP), multisystem atrophy (MSA), and corticobasal degeneration (CBD). This review highlights the characteristic structural, functional, and molecular imaging features of these complex disorders. DLB typically demonstrates parieto-occipital hypometabolism with involvement of the cuneus on FDG-PET, whereas dopaminergic imaging, such as [123I]-FP-CIT SPECT (DaTscan) or fluorodopa (FDOPA)-PET, can be utilized as an adjunct for diagnosis. PSP typically shows midbrain atrophy on structural imaging, whereas FDG-PET may be useful to depict frontal lobe hypometabolism and tau-PET confirms underlying tauopathy. MSA typically demonstrates putaminal or cerebellar atrophy, whereas FDG-PET highlights characteristic nigrostriatal or olivopontocerebellar hypometabolism, respectively. Finally, CBD typically shows asymmetric atrophy in the superior parietal lobules and corpus callosum, whereas FDG and tau-PET demonstrate asymmetric hemispheric and subcortical involvement contralateral to the side of clinical deficits. Additional advanced neuroimaging modalities and techniques described may assist in the diagnostic work-up or are promising areas of emerging research.
Collapse
Affiliation(s)
- Graham Keir
- From the Neuroradiology Division (G.K., M.R.), Department of Radiology, Weill Cornell Medical College, NY-Presbyterian Hospital, New York, New York
| | - Michelle Roytman
- From the Neuroradiology Division (G.K., M.R.), Department of Radiology, Weill Cornell Medical College, NY-Presbyterian Hospital, New York, New York
| | - Faizullah Mashriqi
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| | - Shaya Shahsavarani
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| | - Ana M Franceschi
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| |
Collapse
|
16
|
Wu X, Shirani H, Vidal R, Ghetti B, Ingelsson M, Klingstedt T, Nilsson KPR. Distinct Chemical Determinants are Essential for Achieving Ligands for Superior Optical Detection of Specific Amyloid-β Deposits in Alzheimer's Disease. ChemistryOpen 2024; 13:e202400186. [PMID: 39508558 PMCID: PMC11625938 DOI: 10.1002/open.202400186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Aggregated forms of different proteins are common hallmarks for several neurodegenerative diseases, including Alzheimer's disease, and ligands that selectively detect specific protein aggregates are vital. Herein, we investigate the molecular requirements of thiophene-vinyl-benzothiazole based ligands to detect a specific type of Aβ deposits found in individuals with dominantly inherited Alzheimer's disease caused by the Arctic APP E693G mutation. The staining of these Aβ deposits was alternated when switching the terminal heterocyclic moiety attached to the thiophene-vinyl-benzothiazole scaffold. The most prevalent staining was observed for ligands having a terminal 3-methyl-1H-indazole moiety or a terminal 1,2-dimethoxybenzene moiety, verifying that specific molecular interactions between these ligands and the aggregates were necessary. The synthesis of additional thiophene-vinyl-benzothiazole ligands aided in pinpointing additional crucial chemical determinants, such as positioning of nitrogen atoms and methyl substituents, for achieving optimal staining of Aβ aggregates. When combining the optimized thiophene-vinyl-benzothiazole based ligands with a conventional ligand, CN-PiB, distinct staining patterns were observed for sporadic Alzheimer's disease versus dominantly inherited Alzheimer's disease caused by the Arctic APP E693G mutation. Our findings provide chemical insights for developing novel ligands that allow for a more precise assignment of Aβ deposits, and might also aid in creating novel agents for clinical imaging of distinct Aβ aggregates in AD.
Collapse
Affiliation(s)
- Xiongyu Wu
- Department of Physics, Chemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| | - Hamid Shirani
- Department of Physics, Chemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| | - Ruben Vidal
- Department of Pathology and Laboratory MedicineIndiana University School of Medicine46202Indianapolis, IndianaUSA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory MedicineIndiana University School of Medicine46202Indianapolis, IndianaUSA
| | - Martin Ingelsson
- Krembil Brain InstituteUniversity Health NetworkM5T 1 M8Toronto, OntarioCanada
- Tanz Centre for Research in Neurodegenerative DiseasesDepartments of Medicine and Laboratory Medicine & PathobiologyUniversity of TorontoM5T 0S8Toronto, OntarioCanada
- Molecular GeriatricsDepartment of Public Health and Caring SciencesUppsala UniversitySE-751 85UppsalaSweden
| | - Therése Klingstedt
- Department of Physics, Chemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| | - K. Peter R. Nilsson
- Department of Physics, Chemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| |
Collapse
|
17
|
Slemann L, Gnörich J, Hummel S, Bartos LM, Klaus C, Kling A, Kusche-Palenga J, Kunte ST, Kunze LH, Englert AL, Li Y, Vogler L, Katzdobler S, Palleis C, Bernhardt A, Jäck A, Zwergal A, Hopfner F, Roemer-Cassiano SN, Biechele G, Stöcklein S, Bischof G, van Eimeren T, Drzezga A, Sabri O, Barthel H, Respondek G, Grimmer T, Levin J, Herms J, Paeger L, Willroider M, Beyer L, Höglinger GU, Roeber S, Franzmeier N, Brendel M. Neuronal and oligodendroglial, but not astroglial, tau translates to in vivo tau PET signals in individuals with primary tauopathies. Acta Neuropathol 2024; 148:70. [PMID: 39580770 PMCID: PMC11586312 DOI: 10.1007/s00401-024-02834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Tau PET has attracted increasing interest as an imaging biomarker for 4-repeat (4R)-tauopathy progressive supranuclear palsy (PSP). However, the translation of in vitro 4R-tau binding to in vivo tau PET signals is still unclear. Therefore, we performed a translational study using a broad spectrum of advanced methodologies to investigate the sources of [18F]PI-2620 tau PET signals in individuals with 4R-tauopathies, including a pilot PET autopsy study in patients. First, we conducted a longitudinal [18F]PI-2620 PET/MRI study in a 4-repeat-tau mouse model (PS19) and detected elevated [18F]PI-2620 PET signals in the presence of high levels of neuronal tau. An innovative approach involving cell sorting after radiotracer injection in vivo revealed higher tracer uptake in single neurons than in the astrocytes of PS19 mice. Regional [18F]PI-2620 tau PET signals during the lifetime correlated with the abundance of fibrillary tau and with autoradiography signal intensity in PSP patients and disease controls who underwent autopsy 2-63 months after tau PET. In autoradiography, tau-positive neurons and oligodendrocytes with a high AT8 density, but not tau-positive astrocytes, were the drivers of [18F]PI-2620 autoradiography signals in individuals with PSP. The high tau abundance in oligodendrocytes at the boundary of gray and white matter facilitated the identification of an optimized frontal lobe target region to detect the tau burden in patients with PSP. In summary, neuronal and oligodendroglial tau constitutes the dominant source of tau PET radiotracer binding in 4-repeat-tauopathies, translating to an in vivo signal.
Collapse
Affiliation(s)
- Luna Slemann
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Selina Hummel
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Carolin Klaus
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Agnes Kling
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Julia Kusche-Palenga
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Sebastian T Kunte
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Lea H Kunze
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Amelie L Englert
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Yunlei Li
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Letizia Vogler
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Sabrina Katzdobler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Alexander Bernhardt
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Alexander Jäck
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- German Center for Vertigo and Balance Disorders, DSGZ, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Franziska Hopfner
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sebastian N Roemer-Cassiano
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU Hospital, LMU Munich, Munich, Germany
| | - Gloria Biechele
- Department of Radiology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sophia Stöcklein
- Department of Radiology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Gerard Bischof
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Thilo van Eimeren
- Cognitive Neuroscience, Institute for Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- Department of Neurology, University Hospital Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Gesine Respondek
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Timo Grimmer
- Center for Cognitive Disorders, Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Center of Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lars Paeger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Marie Willroider
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Department of Neurology, LMU Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sigrun Roeber
- Center of Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Nicolai Franzmeier
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Stroke and Dementia Research, LMU Hospital, LMU Munich, Munich, Germany
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, , University of Gothenburg, The Sahlgrenska Academy, Mölndal, Gothenburg, Sweden
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU Hospital, Ludwig Maximilian University of Munich, Marchioninstraße 15, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
18
|
Studart-Neto A, Barbosa BJAP, Coutinho AM, de Souza LC, Schilling LP, da Silva MNM, Castilhos RM, Bertolucci PHF, Borelli WV, Gomes HR, Fernandes GBP, Barbosa MT, Balthazar MLF, Frota NAF, Forlenza OV, Smid J, Brucki SMD, Caramelli P, Nitrini R, Engelhardt E, Resende EDPF. Guidelines for the use and interpretation of Alzheimer's disease biomarkers in clinical practice in Brazil: recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2024; 18:e2024C001. [PMID: 39534442 PMCID: PMC11556292 DOI: 10.1590/1980-5764-dn-2024-c001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, the diagnostic accuracy of Alzheimer's disease has been enhanced by the development of different types of biomarkers that indicate the presence of neuropathological processes. In addition to improving patient selection for clinical trials, biomarkers can assess the effects of new treatments on pathological processes. However, there is concern about the indiscriminate and poorly supported use of biomarkers, especially in asymptomatic individuals or those with subjective cognitive decline. Difficulties interpreting these tests, high costs, and unequal access make this scenario even more challenging in healthcare. This article presents the recommendations from the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology (Departamento Científico de Neurologia Cognitiva e Envelhecimento da Academia Brasileira de Neurologia) regarding the rational use and interpretation of Alzheimer's disease biomarkers in clinical practice. The clinical diagnosis of cognitive-behavioral syndrome is recommended as the initial step to guide the request for biomarkers.
Collapse
Affiliation(s)
- Adalberto Studart-Neto
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Breno José Alencar Pires Barbosa
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Pernambuco, Hospital das Clínicas, Recife, Centro de Ciências Médicas, Recife PE, Brazil
- Universidade Federal de Pernambuco, Empresa Brasileira de Serviços Hospitalares, Hospital das Clínicas, Departamento de Neurologia, Recife PE, Brazil
| | - Artur Martins Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Radiologia, Centro de Medicina Nuclear, Laboratório de Investigação Médica (LIM 43), São Paulo SP, Brazil
- Hospital Sírio-Libanês, Medicina Nuclear e Serviço de PET-CT, São Paulo SP, Brazil
| | - Leonardo Cruz de Souza
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Lucas Porcello Schilling
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brazil
| | - Mari Nilva Maia da Silva
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Nina Rodrigues, Serviço de Neuropsiquiatria, São Luís MA, Brazil
| | - Raphael Machado Castilhos
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Centro de Neurologia Cognitiva e Comportamental, Porto Alegre RS, Brazil
| | - Paulo Henrique Ferreira Bertolucci
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, São Paulo SP, Brazil
| | - Wyllians Vendramini Borelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Departamento de Ciências Morfológicas, Porto Alegre RS, Brazil
| | - Hélio Rodrigues Gomes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Laboratório de Líquido Cefalorraquidiano, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Laboratório de Investigação Médica (LIM 15), São Paulo SP, Brazil
- Departamento Científico de Líquido Cefalorraquiano, Academia Brasileira de Neurologia, São Paulo SP, Brazil
| | | | - Maira Tonidandel Barbosa
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Marcio Luiz Figueredo Balthazar
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brazil
| | - Norberto Anízio Ferreira Frota
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Hospital Geral de Fortaleza, Serviço de Neurologia, Fortaleza CE, Brazil
- Universidade de Fortaleza, Fortaleza, CE, Brazil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brazil
| | - Jerusa Smid
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Sonia Maria Dozzi Brucki
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Paulo Caramelli
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| | - Ricardo Nitrini
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Eliasz Engelhardt
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Neurologia Deolindo Couto, Rio de Janeiro RJ, Brazil
- Universidade Federal do Rio de Janeiro, Instituto de Psiquiatria, Rio de Janeiro RJ, Brazil
| | - Elisa de Paula França Resende
- Academia Brasileira de Neurologia, Departamento Científico de Neurologia Cognitiva e do Envelhecimento, São Paulo SP, Brazil
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Unidade de Neurologia Cognitiva e do Comportamento, Belo Horizonte MG, Brazil
| |
Collapse
|
19
|
Sheng L, Bhalla R. Biomarkers and Target-Specific Small-Molecule Drugs in Alzheimer's Diagnostic and Therapeutic Research: From Amyloidosis to Tauopathy. Neurochem Res 2024; 49:2273-2302. [PMID: 38844706 PMCID: PMC11310295 DOI: 10.1007/s11064-024-04178-w] [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: 01/16/2024] [Revised: 03/31/2024] [Accepted: 05/22/2024] [Indexed: 08/09/2024]
Abstract
Alzheimer's disease (AD) is the most common type of human dementia and is responsible for over 60% of diagnosed dementia cases worldwide. Abnormal deposition of β-amyloid and the accumulation of neurofibrillary tangles have been recognised as the two pathological hallmarks targeted by AD diagnostic imaging as well as therapeutics. With the progression of pathological studies, the two hallmarks and their related pathways have remained the focus of researchers who seek for AD diagnostic and therapeutic strategies in the past decades. In this work, we reviewed the development of the AD biomarkers and their corresponding target-specific small molecule drugs for both diagnostic and therapeutic applications, underlining their success, failure, and future possibilities.
Collapse
Affiliation(s)
- Li Sheng
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
20
|
Arabi H, Manesh AS, Zaidi H. Innovations in dedicated PET instrumentation: from the operating room to specimen imaging. Phys Med Biol 2024; 69:11TR03. [PMID: 38744305 DOI: 10.1088/1361-6560/ad4b92] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 05/14/2024] [Indexed: 05/16/2024]
Abstract
This review casts a spotlight on intraoperative positron emission tomography (PET) scanners and the distinctive challenges they confront. Specifically, these systems contend with the necessity of partial coverage geometry, essential for ensuring adequate access to the patient. This inherently leans them towards limited-angle PET imaging, bringing along its array of reconstruction and geometrical sensitivity challenges. Compounding this, the need for real-time imaging in navigation systems mandates rapid acquisition and reconstruction times. For these systems, the emphasis is on dependable PET image reconstruction (without significant artefacts) while rapid processing takes precedence over the spatial resolution of the system. In contrast, specimen PET imagers are unburdened by the geometrical sensitivity challenges, thanks to their ability to leverage full coverage PET imaging geometries. For these devices, the focus shifts: high spatial resolution imaging takes precedence over rapid image reconstruction. This review concurrently probes into the technical complexities of both intraoperative and specimen PET imaging, shedding light on their recent designs, inherent challenges, and technological advancements.
Collapse
Affiliation(s)
- Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva 4, Switzerland
| | - Abdollah Saberi Manesh
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva 4, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva 4, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, 500 Odense, Denmark
- University Research and Innovation Center, Óbuda University, Budapest, Hungary
| |
Collapse
|
21
|
Jaisa-Aad M, Muñoz-Castro C, Healey MA, Hyman BT, Serrano-Pozo A. Characterization of monoamine oxidase-B (MAO-B) as a biomarker of reactive astrogliosis in Alzheimer's disease and related dementias. Acta Neuropathol 2024; 147:66. [PMID: 38568475 PMCID: PMC10991006 DOI: 10.1007/s00401-024-02712-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/03/2024] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
Reactive astrogliosis accompanies the two neuropathological hallmarks of Alzheimer's disease (AD)-Aβ plaques and neurofibrillary tangles-and parallels neurodegeneration in AD and AD-related dementias (ADRD). Thus, there is growing interest in developing imaging and fluid biomarkers of reactive astrogliosis for AD/ADRD diagnosis and prognostication. Monoamine oxidase-B (MAO-B) is emerging as a target for PET imaging radiotracers of reactive astrogliosis. However, a thorough characterization of MAO-B expression in postmortem control and AD/ADRD brains is lacking. We sought to: (1) identify the primary cell type(s) expressing MAO-B in control and AD brains; (2) quantify MAO-B immunoreactivity in multiple brain regions of control and AD donors as a proxy for PET radiotracer uptake; (3) correlate MAO-B level with local AD neuropathological changes, reactive glia, and cortical atrophy; (4) determine whether the MAOB rs1799836 SNP genotype impacts MAO-B expression level; (5) compare MAO-B immunoreactivity across AD/ADRD, including Lewy body diseases (LBD) and frontotemporal lobar degenerations with tau (FTLD-Tau) and TDP-43 (FTLD-TDP). We found that MAO-B is mainly expressed by subpial and perivascular cortical astrocytes as well as by fibrous white matter astrocytes in control brains, whereas in AD brains, MAO-B is significantly upregulated by both cortical reactive astrocytes and white matter astrocytes across temporal, frontal, and occipital lobes. By contrast, MAO-B expression level was unchanged and lowest in cerebellum. Cortical MAO-B expression was independently associated with cortical atrophy and local measures of reactive astrocytes and microglia, and significantly increased in reactive astrocytes surrounding Thioflavin-S+ dense-core Aβ plaques. MAO-B expression was not affected by the MAOB rs1799836 SNP genotype. MAO-B expression was also significantly increased in the frontal cortex and white matter of donors with corticobasal degeneration, Pick's disease, and FTLD-TDP, but not in LBD or progressive supranuclear palsy. These findings support ongoing efforts to develop MAO-B-based PET radiotracers to image reactive astrogliosis in AD/ADRD.
Collapse
Affiliation(s)
- Methasit Jaisa-Aad
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Clara Muñoz-Castro
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Molly A Healey
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
22
|
Ishibashi K, Kurihara M, Toyohara J, Ishii K, Iwata A. Pitfalls of Amyloid-Beta PET: Comparisons With 18 F-MK-6240 and 18 F-THK5351 PET. Clin Nucl Med 2024; 49:319-321. [PMID: 38363815 DOI: 10.1097/rlu.0000000000005097] [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: 02/18/2024]
Abstract
ABSTRACT We present 3 patients as pitfalls of amyloid-beta (Aβ) PET, who underwent 11 C-PiB (Aβ), 18 F-MK-6240 (Alzheimer disease [AD]-tau), and 18 F-THK5351 (astrogliosis) PET examinations. Despite negligible or tiny Aβ pathology, patients 1 and 2 were diagnosed with AD as the cause of symptoms. Despite widespread Aβ pathology, patient 3 was not diagnosed with AD as the cause of symptoms. However, if we had only conducted Aβ PET, patients 1 and 2 might not have been diagnosed with AD, whereas patient 3 might have been diagnosed with AD. Hence, both Aβ and AD-tau assessments are necessary to relate clinical symptoms to AD pathology.
Collapse
Affiliation(s)
| | - Masanori Kurihara
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | | | | | - Atsushi Iwata
- Department of Neurology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| |
Collapse
|
23
|
Ishibashi K. Clinical application of MAO-B PET using 18F-THK5351 in neurological disorders. Geriatr Gerontol Int 2024; 24 Suppl 1:31-43. [PMID: 37973072 PMCID: PMC11503588 DOI: 10.1111/ggi.14729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
Monoamine oxidase B (MAO-B) is an enzyme localized to the outer mitochondrial membrane and highly concentrated in astrocytes. Temporal changes in regional MAO-B levels can be used as an index of astrocytic proliferation, known as activated astrocytes or astrogliosis. MAO-B is a marker to evaluate the degree of astrogliosis. Therefore, MAO-B positron emission tomography (PET) is a powerful imaging technique for visualizing and quantifying ongoing astrogliosis through the estimate of regional MAO-B levels. Each neurodegenerative disorder generally has a characteristic distribution pattern of astrogliosis secondary to neuronal loss and pathological protein aggregation. Therefore, by imaging astrogliosis, MAO-B PET can be used as a neurodegeneration marker for identifying degenerative lesions. Any inflammation in the brain usually accompanies astrogliosis starting from an acute phase to a chronic phase. Therefore, by imaging astrogliosis, MAO-B PET can be used as a neuroinflammation marker for identifying inflammatory lesions. MAO-B levels are high in gliomas originating from astrocytes but low in lymphoid tumors. Therefore, MAO-B PET can be used as a brain tumor marker for identifying astrocytic gliomas by imaging MAO-B levels and distinguishing between astrocytic and lymphoid tumors. This review summarizes the clinical application of MAO-B PET using 18F-THK5351 as markers for neurodegeneration, neuroinflammation, and brain tumors in neurological disorders. Because we assume that MAO-B PET is clinically applied to an individual patient, we focus on visual inspection of MAO-B images at the individual patient level. Geriatr Gerontol Int 2024; 24: 31-43.
Collapse
Affiliation(s)
- Kenji Ishibashi
- Diagnostic Neuroimaging Research, Research Team for NeuroimagingTokyo Metropolitan Institute for Geriatrics and GerontologyTokyoJapan
| |
Collapse
|
24
|
Kubota M, Endo H, Takahata K, Tagai K, Suzuki H, Onaya M, Sano Y, Yamamoto Y, Kurose S, Matsuoka K, Seki C, Shinotoh H, Kawamura K, Zhang MR, Takado Y, Shimada H, Higuchi M. In vivo PET classification of tau pathologies in patients with frontotemporal dementia. Brain Commun 2024; 6:fcae075. [PMID: 38510212 PMCID: PMC10953627 DOI: 10.1093/braincomms/fcae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/23/2023] [Accepted: 02/29/2024] [Indexed: 03/22/2024] Open
Abstract
Frontotemporal dementia refers to a group of neurodegenerative disorders with diverse clinical and neuropathological features. In vivo neuropathological assessments of frontotemporal dementia at an individual level have hitherto not been successful. In this study, we aim to classify patients with frontotemporal dementia based on topologies of tau protein aggregates captured by PET with 18F-florzolotau (aka 18F-APN-1607 and 18F-PM-PBB3), which allows high-contrast imaging of diverse tau fibrils in Alzheimer's disease as well as in non-Alzheimer's disease tauopathies. Twenty-six patients with frontotemporal dementia, 15 with behavioural variant frontotemporal dementia and 11 with other frontotemporal dementia phenotypes, and 20 age- and sex-matched healthy controls were included in this study. They underwent PET imaging of amyloid and tau depositions with 11C-PiB and 18F-florzolotau, respectively. By combining visual and quantitative analyses of PET images, the patients with behavioural variant frontotemporal dementia were classified into the following subgroups: (i) predominant tau accumulations in frontotemporal and frontolimbic cortices resembling three-repeat tauopathies (n = 3), (ii) predominant tau accumulations in posterior cortical and subcortical structures indicative of four-repeat tauopathies (n = 4); (iii) amyloid and tau accumulations consistent with Alzheimer's disease (n = 4); and (iv) no overt amyloid and tau pathologies (n = 4). Despite these distinctions, clinical symptoms and localizations of brain atrophy did not significantly differ among the identified behavioural variant frontotemporal dementia subgroups. The patients with other frontotemporal dementia phenotypes were also classified into similar subgroups. The results suggest that PET with 18F-florzolotau potentially allows the classification of each individual with frontotemporal dementia on a neuropathological basis, which might not be possible by symptomatic and volumetric assessments.
Collapse
Affiliation(s)
- Manabu Kubota
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Sakyo-ku Kyoto 606-8507, Japan
| | - Hironobu Endo
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Keisuke Takahata
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenji Tagai
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Psychiatry, Jikei University Graduate School of Medicine, Tokyo 105-8461, Japan
| | - Hisaomi Suzuki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Psychiatry, National Hospital OrganizationShimofusa Psychiatric Center, Chiba 266-0007, Japan
| | - Mitsumoto Onaya
- Department of Psychiatry, National Hospital OrganizationShimofusa Psychiatric Center, Chiba 266-0007, Japan
| | - Yasunori Sano
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yasuharu Yamamoto
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shin Kurose
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kiwamu Matsuoka
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Psychiatry, Nara Medical University, Nara 634-8521, Japan
| | - Chie Seki
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Hitoshi Shinotoh
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Kazunori Kawamura
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
- Department of Functional Neurology and Neurosurgery, Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
25
|
Vanderlinden G, Michiels L, Koole M, Lemmens R, Liessens D, Van Walleghem J, Depreitere B, Vandenbulcke M, Van Laere K. Tau Imaging in Late Traumatic Brain Injury: A [ 18F]MK-6240 Positron Emission Tomography Study. J Neurotrauma 2024; 41:420-429. [PMID: 38038357 DOI: 10.1089/neu.2023.0085] [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: 12/02/2023] Open
Abstract
Epidemiological studies have identified prior traumatic brain injury (TBI) as a risk factor for developing Alzheimer's disease (AD). Neurofibrillary tangles (NFTs) are common to AD and chronic traumatic encephalopathy following repetitive mild TBI. However, it is unclear if a single TBI is sufficient to cause accumulation of NFTs. We performed a [18F]MK-6240 positron emission tomography (PET) imaging study to assess NFTs in patients who had sustained a single TBI at least 2 years prior to study inclusion. Fourteen TBI patients (49 ± 20 years; 5 M/9 F; 8 moderate-severe, 1 mild-probable, 5 symptomatic-possible TBI) and 40 demographically similar controls (57 ± 19 years; 19 M/21 F) underwent simultaneous [18F]MK-6240 PET and magnetic resonance imaging (MRI) as well as neuropsychological assessment including the Cambridge Neuropsychological Test Automated Battery (CANTAB). A region-based voxelwise partial volume correction was applied, using parcels obtained by FreeSurfer v6.0, and standardized uptake value ratios (SUVR) were calculated relative to the cerebellar gray matter. Group differences were assessed on both a voxel- and a volume-of-interest-based level and correlations of [18F]MK-6240 SUVR with time since injury as well as with clinical outcomes were calculated. Visual assessment of TBI images did not show global or focal increases in tracer uptake in any subject. On a group level, [18F]MK-6240 SUVR was not significantly different in patients versus controls or between subgroups of moderate-severe TBI versus less severe TBI. Within the TBI group, One Touch Stockings problem solving and spatial working memory (executive function), reaction time (attention), and Mini-Mental State Examination (MMSE) (global cognition) were associated with [18F]MK-6240 SUVR. We found no group-based increase of [18F]MK-6240 brain uptake in patients scanned at least 2 years after a single TBI compared with healthy volunteers, which suggests that no NFTs are building up in the first years after a single TBI. Nonetheless, correlations with cognitive outcomes were found that warrant further investigation.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Laura Michiels
- Leuven Brain Institute, Leuven, Belgium
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium
- VIB, Center for Brain and Disease Research, Laboratory of Neurobiology, Belgium
- Neurosciences, and Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Robin Lemmens
- Leuven Brain Institute, Leuven, Belgium
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium
- VIB, Center for Brain and Disease Research, Laboratory of Neurobiology, Belgium
- Neurosciences, and Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Dirk Liessens
- Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | | | - Bart Depreitere
- Department of Neurosurgery, and University Hospitals UZ Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
- Neuropsychiatry, Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Imaging and Pathology, and Departments of Research Group Psychiatry, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
- Department of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Aguero C, Dhaynaut M, Amaral AC, Moon SH, Neelamegam R, Scapellato M, Carazo-Casas C, Kumar S, El Fakhri G, Johnson K, Frosch MP, Normandin MD, Gómez-Isla T. Head-to-head comparison of [ 18F]-Flortaucipir, [ 18F]-MK-6240 and [ 18F]-PI-2620 postmortem binding across the spectrum of neurodegenerative diseases. Acta Neuropathol 2024; 147:25. [PMID: 38280071 PMCID: PMC10822013 DOI: 10.1007/s00401-023-02672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/29/2024]
Abstract
We and others have shown that [18F]-Flortaucipir, the most validated tau PET tracer thus far, binds with strong affinity to tau aggregates in Alzheimer's (AD) but has relatively low affinity for tau aggregates in non-AD tauopathies and exhibits off-target binding to neuromelanin- and melanin-containing cells, and to hemorrhages. Several second-generation tau tracers have been subsequently developed. [18F]-MK-6240 and [18F]-PI-2620 are the two that have garnered most attention. Our recent data indicated that the binding pattern of [18F]-MK-6240 closely parallels that of [18F]-Flortaucipir. The present study aimed at the direct comparison of the autoradiographic binding properties and off-target profile of [18F]-Flortaucipir, [18F]-MK-6240 and [18F]-PI-2620 in human tissue specimens, and their potential binding to monoamine oxidases (MAO). Phosphor-screen and high resolution autoradiographic patterns of the three tracers were studied in the same postmortem tissue material from AD and non-AD tauopathies, cerebral amyloid angiopathy, synucleopathies, transactive response DNA-binding protein 43 (TDP-43)-frontotemporal lobe degeneration and controls. Our results show that the three tracers show nearly identical autoradiographic binding profiles. They all strongly bind to neurofibrillary tangles in AD but do not seem to bind to a significant extent to tau aggregates in non-AD tauopathies pointing to their limited utility for the in vivo detection of non-AD tau lesions. None of them binds to lesions containing β-amyloid, α-synuclein or TDP-43 but they all show strong off-target binding to neuromelanin and melanin-containing cells, as well as weaker binding to areas of hemorrhage. The autoradiographic binding signals of the three tracers are only weakly displaced by competing concentrations of selective MAO-B inhibitor deprenyl but not by MAO-A inhibitor clorgyline suggesting that MAO enzymes do not appear to be a significant binding target of any of them. These findings provide relevant insights for the correct interpretation of the in vivo behavior of these three tau PET tracers.
Collapse
Affiliation(s)
- Cinthya Aguero
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ana C Amaral
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - S-H Moon
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Margaret Scapellato
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Carlos Carazo-Casas
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Sunny Kumar
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew P Frosch
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gómez-Isla
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA.
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA.
| |
Collapse
|
27
|
Malotaux V, Colmant L, Quenon L, Huyghe L, Gérard T, Dricot L, Ivanoiu A, Lhommel R, Hanseeuw B. Suspecting Non-Alzheimer's Pathologies and Mixed Pathologies: A Comparative Study Between Brain Metabolism and Tau Images. J Alzheimers Dis 2024; 97:421-433. [PMID: 38108350 PMCID: PMC10789317 DOI: 10.3233/jad-230696] [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] [Accepted: 10/27/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) pathology can be disclosed in vivo using amyloid and tau imaging, unlike non-AD neuropathologies for which no specific markers exist. OBJECTIVE We aimed to compare brain hypometabolism and tauopathy to unveil non-AD pathologies. METHODS Sixty-one patients presenting cognitive complaints (age 48-90), including 32 with positive AD biomarkers (52%), performed [18F]-Fluorodeoxyglucose (FDG)-PET (brain metabolism) and [18F]-MK-6240-PET (tau). We normalized these images using data from clinically normal individuals (n = 30), resulting in comparable FDG and tau z-scores. We computed between-patients correlations to evaluate regional associations. For each patient, a predominant biomarker (i.e., Hypometabolism > Tauopathy or Hypometabolism≤Tauopathy) was determined in the temporal and frontoparietal lobes. We computed within-patient correlations between tau and metabolism and investigated their associations with demographics, cognition, cardiovascular risk factors (CVRF), CSF biomarkers, and white matter hypointensities (WMH). RESULTS We observed negative associations between tau and FDG in 37 of the 68 cortical regions-of-interest (average Pearson's r = -0.25), mainly in the temporal lobe. Thirteen patients (21%) had Hypometabolism > Tauopathy whereas twenty-five patients (41%) had Hypometabolism≤Tauopathy. Tau-predominant patients were more frequently females and had greater amyloid burden. Twenty-three patients (38%) had Hypometabolism≤Tauopathy in the temporal lobe, but Hypometabolism > Tauopathy in the frontoparietal lobe. This group was older and had higher CVRF than Tau-predominant patients. Patients with more negative associations between tau and metabolism were younger, had worse cognition, and greater amyloid and WMH burdens. CONCLUSIONS Tau-FDG comparison can help suspect non-AD pathologies in patients presenting cognitive complaints. Stronger Tau-FDG correlations are associated with younger age, worse cognition, and greater amyloid and WMH burdens.
Collapse
Affiliation(s)
- Vincent Malotaux
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Lise Colmant
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
- Department of Neurology, Saint-Luc University Hospital, Brussels, Belgium
| | - Lisa Quenon
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
- Department of Neurology, Saint-Luc University Hospital, Brussels, Belgium
| | - Lara Huyghe
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Thomas Gérard
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
- Department of Nuclear Medicine, Saint-Luc University Hospital, Brussels, Belgium
| | - Laurence Dricot
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Adrian Ivanoiu
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
- Department of Neurology, Saint-Luc University Hospital, Brussels, Belgium
| | - Renaud Lhommel
- Department of Nuclear Medicine, Saint-Luc University Hospital, Brussels, Belgium
| | - Bernard Hanseeuw
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
- Department of Neurology, Saint-Luc University Hospital, Brussels, Belgium
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- WEL Research Institute, Welbio department, Wavre, Belgium
| |
Collapse
|
28
|
Wang Y, Zhang Y, Yu E. Targeted examination of amyloid beta and tau protein accumulation via positron emission tomography for the differential diagnosis of Alzheimer's disease based on the A/T(N) research framework. Clin Neurol Neurosurg 2024; 236:108071. [PMID: 38043158 DOI: 10.1016/j.clineuro.2023.108071] [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: 06/08/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/05/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases among the older population. Its main pathological features include the abnormal deposition of extracellular amyloid-β plaques and the intracellular neurofibrillary tangles of tau proteins. Its clinical presentation is complex. This review introduces the pathological processes in AD and other common neurodegenerative diseases. It then discusses the positron emission tomography (PET) probes that target amyloid-β plaques and tau proteins for diagnosing AD. According to the A/T(N) research framework, combined targeted amyloid-β and tau protein detection via PET to further improve the diagnostic accuracy of AD. In particular, the properties of the 18F-flortaucipir and 18F-MK6240 tracers-may be more beneficial in helping to differentiate AD from other common neurodegenerative diseases, such as dementia with Lewy bodies, Parkinson's disease dementia, and frontotemporal dementia. Furthermore, the A/T(N) research framework should be used as the clinical diagnosis model of AD in the future.
Collapse
Affiliation(s)
- Ye Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Psychiatry, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), 310022, China
| | - Yuhan Zhang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Enyan Yu
- Department of Psychiatry, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), 310022, China.
| |
Collapse
|
29
|
Dybing KM, Vetter CJ, Dempsey DA, Chaudhuri S, Saykin AJ, Risacher SL. Traumatic brain injury and Alzheimer's Disease biomarkers: A systematic review of findings from amyloid and tau positron emission tomography (PET). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.30.23298528. [PMID: 38077068 PMCID: PMC10705648 DOI: 10.1101/2023.11.30.23298528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Traumatic brain injury (TBI) has been discussed as a risk factor for Alzheimer's disease (AD) due to its association with dementia risk and earlier cognitive symptom onset. However, the mechanisms behind this relationship are unclear. Some studies have suggested TBI may increase pathological protein deposition in an AD-like pattern; others have failed to find such associations. This review covers literature that uses positron emission tomography (PET) of amyloid-β and/or tau to examine subjects with history of TBI who are at risk for AD due to advanced age. A comprehensive literature search was conducted on January 9, 2023, and 24 resulting citations met inclusion criteria. Common methodological concerns included small samples, limited clinical detail about subjects' TBI, recall bias due to reliance on self-reported TBI, and an inability to establish causation. For both amyloid and tau, results were widespread but inconsistent. The regions which showed the most compelling evidence for increased amyloid deposition were the cingulate gyrus, cuneus/precuneus, and parietal lobe. Evidence for increased tau was strongest in the medial temporal lobe, entorhinal cortex, precuneus, and frontal, temporal, parietal, and occipital lobes. However, conflicting findings across most regions of interest in both amyloid- and tau-PET studies indicate the critical need for future work in expanded samples and with greater clinical detail to offer a clearer picture of the relationship between TBI and protein deposition in older subjects at risk for AD.
Collapse
Affiliation(s)
- Kaitlyn M. Dybing
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cecelia J. Vetter
- Ruth Lilly Medical Library, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Desarae A. Dempsey
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Soumilee Chaudhuri
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
30
|
Burnham SC, Iaccarino L, Pontecorvo MJ, Fleisher AS, Lu M, Collins EC, Devous MD. A review of the flortaucipir literature for positron emission tomography imaging of tau neurofibrillary tangles. Brain Commun 2023; 6:fcad305. [PMID: 38187878 PMCID: PMC10768888 DOI: 10.1093/braincomms/fcad305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 01/09/2024] Open
Abstract
Alzheimer's disease is defined by the presence of β-amyloid plaques and neurofibrillary tau tangles potentially preceding clinical symptoms by many years. Previously only detectable post-mortem, these pathological hallmarks are now identifiable using biomarkers, permitting an in vivo definitive diagnosis of Alzheimer's disease. 18F-flortaucipir (previously known as 18F-T807; 18F-AV-1451) was the first tau positron emission tomography tracer to be introduced and is the only Food and Drug Administration-approved tau positron emission tomography tracer (Tauvid™). It has been widely adopted and validated in a number of independent research and clinical settings. In this review, we present an overview of the published literature on flortaucipir for positron emission tomography imaging of neurofibrillary tau tangles. We considered all accessible peer-reviewed literature pertaining to flortaucipir through 30 April 2022. We found 474 relevant peer-reviewed publications, which were organized into the following categories based on their primary focus: typical Alzheimer's disease, mild cognitive impairment and pre-symptomatic populations; atypical Alzheimer's disease; non-Alzheimer's disease neurodegenerative conditions; head-to-head comparisons with other Tau positron emission tomography tracers; and technical considerations. The available flortaucipir literature provides substantial evidence for the use of this positron emission tomography tracer in assessing neurofibrillary tau tangles in Alzheimer's disease and limited support for its use in other neurodegenerative disorders. Visual interpretation and quantitation approaches, although heterogeneous, mostly converge and demonstrate the high diagnostic and prognostic value of flortaucipir in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Ming Lu
- Avid, Eli Lilly and Company, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
31
|
Aumont E, Bussy A, Bedard MA, Bezgin G, Therriault J, Savard M, Fernandez Arias J, Sziklas V, Vitali P, Poltronetti NM, Pallen V, Thomas E, Gauthier S, Kobayashi E, Rahmouni N, Stevenson J, Tissot C, Chakravarty MM, Rosa-Neto P. Hippocampal subfield associations with memory depend on stimulus modality and retrieval mode. Brain Commun 2023; 5:fcad309. [PMID: 38035364 PMCID: PMC10681971 DOI: 10.1093/braincomms/fcad309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/26/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
Hippocampal atrophy is a well-known feature of age-related memory decline, and hippocampal subfields may contribute differently to this decline. In this cross-sectional study, we investigated the associations between hippocampal subfield volumes and performance in free recall and recognition memory tasks in both verbal and visual modalities in older adults without dementia. We collected MRIs from 97 (41 males) right-handed participants aged over 60. We segmented the right and left hippocampi into (i) dentate gyrus and cornu ammonis 4 (DG/CA4); (ii) CA2 and CA3 (CA2/CA3); (iii) CA1; (iv) strata radiatum, lacunosum and moleculare; and (v) subiculum. Memory was assessed with verbal free recall and recognition tasks, as well as visual free recall and recognition tasks. Amyloid-β and hippocampal tau positivity were assessed using [18F]AZD4694 and [18F]MK6240 PET tracers, respectively. The verbal free recall and verbal recognition performances were positively associated with CA1 and strata radiatum, lacunosum and moleculare volumes. The verbal free recall and visual free recall were positively correlated with the right DG/CA4. The visual free recall, but not verbal free recall, was also associated with the right CA2/CA3. The visual recognition was not significantly associated with any subfield volume. Hippocampal tau positivity, but not amyloid-β positivity, was associated with reduced DG/CA4, CA2/CA3 and strata radiatum, lacunosum and moleculare volumes. Our results suggest that memory performances are linked to specific subfields. CA1 appears to contribute to the verbal modality, irrespective of the free recall or recognition mode of retrieval. In contrast, DG/CA4 seems to be involved in the free recall mode, irrespective of verbal or visual modalities. These results are concordant with the view that DG/CA4 plays a primary role in encoding a stimulus' distinctive attributes, and that CA2/CA3 could be instrumental in recollecting a visual memory from one of its fragments. Overall, we show that hippocampal subfield segmentation can be useful for detecting early volume changes and improve our understanding of the hippocampal subfields' roles in memory.
Collapse
Affiliation(s)
- Etienne Aumont
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal H2X 3P2, Canada
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Aurélie Bussy
- Cerebral Imaging Center, Douglas Research Center, Montreal, QC H4H 1R3, Canada
- Computational Brain Anatomy (CoBrALab) Laboratory, Montreal, QC H4H 1R2, Canada
| | - Marc-André Bedard
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal H2X 3P2, Canada
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Gleb Bezgin
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Joseph Therriault
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Melissa Savard
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Jaime Fernandez Arias
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Viviane Sziklas
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Paolo Vitali
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | | | - Vanessa Pallen
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
| | - Emilie Thomas
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
| | - Serge Gauthier
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Eliane Kobayashi
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Nesrine Rahmouni
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Jenna Stevenson
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Cecile Tissot
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| | - Mallar M Chakravarty
- Cerebral Imaging Center, Douglas Research Center, Montreal, QC H4H 1R3, Canada
- Computational Brain Anatomy (CoBrALab) Laboratory, Montreal, QC H4H 1R2, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
| | - Pedro Rosa-Neto
- NeuroQAM Research Centre, Université du Québec à Montréal (UQAM), Montreal H2X 3P2, Canada
- McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC H4H 1R3, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 1A1, Canada
| |
Collapse
|
32
|
Björk L, Shirani H, Todarwal Y, Linares M, Vidal R, Ghetti B, Norman P, Klingstedt T, Nilsson KPR. Distinct Heterocyclic Moieties Govern the Selectivity of Thiophene-Vinylene-Based Ligands Towards Aβ or Tau Pathology in Alzheime's Disease. European J Org Chem 2023; 26:e202300583. [PMID: 38585413 PMCID: PMC10997339 DOI: 10.1002/ejoc.202300583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Indexed: 04/09/2024]
Abstract
Distinct aggregated proteins are correlated with numerous neurodegenerative diseases and the development of ligands that selectively detect these pathological hallmarks is vital. Recently, the synthesis of thiophene-based optical ligands, denoted bi-thiophene-vinyl-benzothiazoles (bTVBTs), that could be utilized for selective assignment of tau pathology in brain tissue with Alzheime's disease (AD) pathology, was reported. Herein, we investigate the ability of these ligands to selectively distinguish tau deposits from aggregated amyloid-β (Aβ), the second AD associated pathological hallmark, when replacing the terminal thiophene moiety with other heterocyclic motifs. The selectivity for tau pathology was reduced when introducing specific heterocyclic motifs, verifying that specific molecular interactions between the ligands and the aggregates are necessary for selective detection of tau deposits. In addition, ligands having certain heterocyclic moieties attached to the central thiophene-vinylene building block displayed selectivity to aggregated Aβ pathology. Our findings provide chemical insights for the development of ligands that can distinguish between aggregated proteinaceous species consisting of different proteins and might also aid in creating novel agents for clinical imaging of tau pathology in AD.
Collapse
Affiliation(s)
- Linnea Björk
- Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - Hamid Shirani
- Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - Yogesh Todarwal
- Division of Theoretical Chemistry and Biology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - Mathieu Linares
- Division of Theoretical Chemistry and Biology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - Ruben Vidal
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, 46202 Indiana, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, 46202 Indiana, USA
| | - Patrick Norman
- Division of Theoretical Chemistry and Biology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| |
Collapse
|
33
|
Lepinay E, Cicchetti F. Tau: a biomarker of Huntington's disease. Mol Psychiatry 2023; 28:4070-4083. [PMID: 37749233 DOI: 10.1038/s41380-023-02230-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023]
Abstract
Developing effective treatments for patients with Huntington's disease (HD)-a neurodegenerative disorder characterized by severe cognitive, motor and psychiatric impairments-is proving extremely challenging. While the monogenic nature of this condition enables to identify individuals at risk, robust biomarkers would still be extremely valuable to help diagnose disease onset and progression, and especially to confirm treatment efficacy. If measurements of cerebrospinal fluid neurofilament levels, for example, have demonstrated use in recent clinical trials, other proteins may prove equal, if not greater, relevance as biomarkers. In fact, proteins such as tau could specifically be used to detect/predict cognitive affectations. We have herein reviewed the literature pertaining to the association between tau levels and cognitive states, zooming in on Alzheimer's disease, Parkinson's disease and traumatic brain injury in which imaging, cerebrospinal fluid, and blood samples have been interrogated or used to unveil a strong association between tau and cognition. Collectively, these areas of research have accrued compelling evidence to suggest tau-related measurements as both diagnostic and prognostic tools for clinical practice. The abundance of information retrieved in this niche of study has laid the groundwork for further understanding whether tau-related biomarkers may be applied to HD and guide future investigations to better understand and treat this disease.
Collapse
Affiliation(s)
- Eva Lepinay
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
34
|
Matsuda H, Yamao T. Tau positron emission tomography in patients with cognitive impairment and suspected Alzheimer's disease. Fukushima J Med Sci 2023; 69:85-93. [PMID: 37302841 PMCID: PMC10480511 DOI: 10.5387/fms.2023-08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
Alzheimer's disease (AD) is diagnosed by the presence of both amyloid β and tau proteins. Recent advances in molecular PET imaging have made it possible to assess the accumulation of these proteins in the living brain. PET ligands have been developed that bind to 3R/4R tau in AD, but not to 3R tau or 4R tau alone. Of the first-generation PET ligands, 18F-flortaucipir has recently been approved by the Food and Drug Administration. Several second-generation PET probes with less off-target binding have been developed and are being applied clinically. Visual interpretation of tau PET should be based on neuropathological neurofibrillary tangle staging instead of a simple positive or negative classification. Four visual read classifications have been proposed: "no uptake," "medial temporal lobe (MTL) only," "MTL AND," and "outside MTL." As an adjunct to visual interpretation, quantitative analysis has been proposed using MRI-based native space FreeSurfer parcellations. The standardized uptake value ratio of the target area is measured using the cerebellar gray matter as a reference region. In the near future, the Centiloid scale of tau PET is expected to be used as a harmonized value for standardizing each analytical method or PET ligand used, similar to amyloid PET.
Collapse
Affiliation(s)
- Hiroshi Matsuda
- Department of Biofunctional Imaging, Fukushima Medical University
| | - Tensho Yamao
- Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University
| |
Collapse
|
35
|
Graham TJA, Lindberg A, Tong J, Stehouwer JS, Vasdev N, Mach RH, Mathis CA. In Silico Discovery and Subsequent Characterization of Potent 4R-Tauopathy Positron Emission Tomography Radiotracers. J Med Chem 2023; 66:10628-10638. [PMID: 37487189 PMCID: PMC10424182 DOI: 10.1021/acs.jmedchem.3c00775] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Indexed: 07/26/2023]
Abstract
A chemical fingerprint search identified Z3777013540 (1-(5-(6-fluoro-1H-indol-2-yl)pyrimidin-2-yl)piperidin-4-ol; 1) as a potential 4R-tau binding ligand. Binding assays in post-mortem Alzheimer's disease (AD), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD) brain with [3H]1 provided KD (nM) values in AD = 4.0, PSP = 5.1, and CBD = 4.5. In vivo positron emission tomography (PET) imaging in rats with [18F]1 demonstrated good brain penetration and rapid clearance from normal brain tissues. A subsequent molecular similarity search using 1 as the query revealed an additional promising compound, Z4169252340 (4-(5-(6-fluoro-1H-indol-2-yl)pyrimidin-2-yl)morpholine; 21). Binding assays with [3H]21 provided KD (nM) values in AD = 1.2, PSP = 1.6, and CBD = 1.7 and lower affinities for binding aggregated α-synuclein and amyloid-beta. PET imaging in rats with [18F]21 demonstrated a higher brain penetration than [18F]1 and rapid clearance from normal brain tissues. We anticipate that 1 and 21 will be useful for the identification of other potent novel 4R-tau radiotracers.
Collapse
Affiliation(s)
- Thomas J. A. Graham
- Department
of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United
States
| | - Anton Lindberg
- Azrieli
Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
| | - Junchao Tong
- Azrieli
Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
| | - Jeffrey S. Stehouwer
- Department
of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Neil Vasdev
- Azrieli
Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department
of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada
| | - Robert H. Mach
- Department
of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United
States
| | - Chester A. Mathis
- Department
of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
36
|
Lu J, Ma X, Zhang H, Xiao Z, Li M, Wu J, Ju Z, Chen L, Zheng L, Ge J, Liang X, Bao W, Wu P, Ding D, Yen TC, Guan Y, Zuo C, Zhao Q. Head-to-head comparison of plasma and PET imaging ATN markers in subjects with cognitive complaints. Transl Neurodegener 2023; 12:34. [PMID: 37381042 PMCID: PMC10308642 DOI: 10.1186/s40035-023-00365-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Gaining more information about the reciprocal associations between different biomarkers within the ATN (Amyloid/Tau/Neurodegeneration) framework across the Alzheimer's disease (AD) spectrum is clinically relevant. We aimed to conduct a comprehensive head-to-head comparison of plasma and positron emission tomography (PET) ATN biomarkers in subjects with cognitive complaints. METHODS A hospital-based cohort of subjects with cognitive complaints with a concurrent blood draw and ATN PET imaging (18F-florbetapir for A, 18F-Florzolotau for T, and 18F-fluorodeoxyglucose [18F-FDG] for N) was enrolled (n = 137). The β-amyloid (Aβ) status (positive versus negative) and the severity of cognitive impairment served as the main outcome measures for assessing biomarker performances. RESULTS Plasma phosphorylated tau 181 (p-tau181) level was found to be associated with PET imaging of ATN biomarkers in the entire cohort. Plasma p-tau181 level and PET standardized uptake value ratios of AT biomarkers showed a similarly excellent diagnostic performance for distinguishing between Aβ+ and Aβ- subjects. An increased tau burden and glucose hypometabolism were significantly associated with the severity of cognitive impairment in Aβ+ subjects. Additionally, glucose hypometabolism - along with elevated plasma neurofilament light chain level - was related to more severe cognitive impairment in Aβ- subjects. CONCLUSION Plasma p-tau181, as well as 18F-florbetapir and 18F-Florzolotau PET imaging can be considered as interchangeable biomarkers in the assessment of Aβ status in symptomatic stages of AD. 18F-Florzolotau and 18F-FDG PET imaging could serve as biomarkers for the severity of cognitive impairment. Our findings have implications for establishing a roadmap to identifying the most suitable ATN biomarkers for clinical use.
Collapse
Affiliation(s)
- Jiaying Lu
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoxi Ma
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Huiwei Zhang
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhenxu Xiao
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ming Li
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Wu
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zizhao Ju
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Chen
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Zheng
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingjie Ge
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoniu Liang
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiqi Bao
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Wu
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Ding Ding
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Yihui Guan
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China.
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China.
| | - Chuantao Zuo
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai, China.
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China.
- Human Phenome Institute, Fudan University, Shanghai, China.
| | - Qianhua Zhao
- National Clinical Research Center for Aging and Medicine and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China.
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Van Egroo M, Riphagen JM, Ashton NJ, Janelidze S, Sperling RA, Johnson KA, Yang HS, Bennett DA, Blennow K, Hansson O, Zetterberg H, Jacobs HIL. Ultra-high field imaging, plasma markers and autopsy data uncover a specific rostral locus coeruleus vulnerability to hyperphosphorylated tau. Mol Psychiatry 2023; 28:2412-2422. [PMID: 37020050 PMCID: PMC10073793 DOI: 10.1038/s41380-023-02041-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/07/2023]
Abstract
Autopsy data indicate that the locus coeruleus (LC) is one of the first sites in the brain to accumulate hyperphosphorylated tau pathology, with the rostral part possibly being more vulnerable in the earlier stages of the disease. Taking advantage of recent developments in ultra-high field (7 T) imaging, we investigated whether imaging measures of the LC also reveal a specific anatomic correlation with tau using novel plasma biomarkers of different species of hyperphosphorylated tau, how early in adulthood these associations can be detected and if are associated with worse cognitive performance. To validate the anatomic correlations, we tested if a rostro-caudal gradient in tau pathology is also detected at autopsy in data from the Rush Memory and Aging Project (MAP). We found that higher plasma measures of phosphorylated tau, in particular ptau231, correlated negatively with dorso-rostral LC integrity, whereas correlations for neurodegenerative plasma markers (neurofilament light, total tau) were scattered throughout the LC including middle to caudal sections. In contrast, the plasma Aβ42/40 ratio, associated with brain amyloidosis, did not correlate with LC integrity. These findings were specific to the rostral LC and not observed when using the entire LC or the hippocampus. Furthermore, in the MAP data, we observed higher rostral than caudal tangle density in the LC, independent of the disease stage. The in vivo LC-phosphorylated tau correlations became significant from midlife, with the earliest effect for ptau231, starting at about age 55. Finally, interactions between lower rostral LC integrity and higher ptau231 concentrations predicted lower cognitive performance. Together, these findings demonstrate a specific rostral vulnerability to early phosphorylated tau species that can be detected with dedicated magnetic resonance imaging measures, highlighting the promise of LC imaging as an early marker of AD-related processes.
Collapse
Grants
- R01 AG017917 NIA NIH HHS
- R01 AG068398 NIA NIH HHS
- R21 AG074220 NIA NIH HHS
- K23 AG062750 NIA NIH HHS
- R01 AG068062 NIA NIH HHS
- K01 AG001016 NIA NIH HHS
- ZEN-21-848495 Alzheimer's Association
- P01 AG036694 NIA NIH HHS
- R01 AG062559 NIA NIH HHS
- R01 AG015819 NIA NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- Alzheimer Nederland WE.03-2019-02
- BrightFocus Foundation (BrightFocus)
- Alzheimer’s Association
- Alzheimer’s Drug Discovery Foundation (ADDF)
- Swedish Research Council (#2017-00915), the Alzheimer Drug Discovery Foundation (ADDF), USA (#RDAPB-201809-2016615), the Swedish Alzheimer Foundation (#AF-930351, #AF-939721 and #AF-968270), Hjärnfonden, Sweden (#FO2017-0243 and #ALZ2022-0006), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986 and #ALFGBG-965240), the European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236)
- Cure Alzheimer’s Fund (Alzheimer’s Disease Research Foundation)
- Swedish Research Council (2016-00906), the Knut and Alice Wallenberg foundation (2017-0383), the Marianne and Marcus Wallenberg foundation (2015.0125), the Strategic Research Area MultiPark (Multidisciplinary Research in Parkinson’s disease) at Lund University, the Swedish Alzheimer Foundation (AF-939932), the Swedish Brain Foundation (FO2021-0293), The Parkinson foundation of Sweden (1280/20), the Cure Alzheimer’s fund, the Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse, the Skåne University Hospital Foundation (2020-O000028), Regionalt Forskningsstöd (2020-0314) and the Swedish federal government under the ALF agreement (2018-Projekt0279)
- HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Research Council (#681712 and #101053962), Swedish State Support for Clinical Research (#ALFGBG-71320), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), the AD Strategic Fund and the Alzheimer’s Association (#ADSF-21-831376-C, #ADSF-21-831381-C, and #ADSF-21-831377-C), the Bluefield Project, the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2022-0270), the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 860197 (MIRIADE), the European Union Joint Programme – Neurodegenerative Disease Research (JPND2021-00694), and the UK Dementia Research Institute at UCL (UKDRI-1003).
Collapse
Affiliation(s)
- Maxime Van Egroo
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Reisa A Sperling
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith A Johnson
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Hyun-Sik Yang
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Kaj Blennow
- 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
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Heidi I L Jacobs
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands.
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Björk L, Klingstedt T, Nilsson KPR. Thiophene-Based Ligands: Design, Synthesis and Their Utilization for Optical Assignment of Polymorphic-Disease-Associated Protein Aggregates. Chembiochem 2023; 24:e202300044. [PMID: 36891883 PMCID: PMC10404026 DOI: 10.1002/cbic.202300044] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/10/2023]
Abstract
The development of ligands for detecting protein aggregates is of great interest, as these aggregated proteinaceous species are the pathological hallmarks of several devastating diseases, including Alzheimer's disease. In this regard, thiophene-based ligands have emerged as powerful tools for fluorescent assessment of these pathological entities. The intrinsic conformationally sensitive photophysical properties of poly- and oligothiophenes have allowed optical assignment of disease-associated protein aggregates in tissue sections, as well as real-time in vivo imaging of protein deposits. Herein, we recount the chemical evolution of different generations of thiophene-based ligands, and exemplify their use for the optical distinction of polymorphic protein aggregates. Furthermore, the chemical determinants for achieving a superior fluorescent thiophene-based ligand, as well as the next generation of thiophene-based ligands targeting distinct aggregated species are described. Finally, the directions for future research into the chemical design of thiophene-based ligands that can aid in resolving the scientific challenges around protein aggregation diseases are discussed.
Collapse
Affiliation(s)
- Linnea Björk
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| |
Collapse
|
39
|
Ozsahin I, Onakpojeruo EP, Uzun B, Uzun Ozsahin D, Butler TA. A Multi-Criteria Decision Aid Tool for Radiopharmaceutical Selection in Tau PET Imaging. Pharmaceutics 2023; 15:1304. [PMID: 37111789 PMCID: PMC10147085 DOI: 10.3390/pharmaceutics15041304] [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: 02/20/2023] [Revised: 04/09/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
The accumulation of pathologically misfolded tau is a feature shared by a group of neurodegenerative disorders collectively referred to as tauopathies. Alzheimer's disease (AD) is the most prevalent of these tauopathies. Immunohistochemical evaluation allows neuropathologists to visualize paired-helical filaments (PHFs)-tau pathological lesions, but this is possible only after death and only shows tau in the portion of brain sampled. Positron emission tomography (PET) imaging allows both the quantitative and qualitative analysis of pathology over the whole brain of a living subject. The ability to detect and quantify tau pathology in vivo using PET can aid in the early diagnosis of AD, provide a way to monitor disease progression, and determine the effectiveness of therapeutic interventions aimed at reducing tau pathology. Several tau-specific PET radiotracers are now available for research purposes, and one is approved for clinical use. This study aims to analyze, compare, and rank currently available tau PET radiotracers using the fuzzy preference ranking organization method for enrichment of evaluations (PROMETHEE), which is a multi-criteria decision-making (MCDM) tool. The evaluation is based on relatively weighted criteria, such as specificity, target binding affinity, brain uptake, brain penetration, and rates of adverse reactions. Based on the selected criteria and assigned weights, this study shows that a second-generation tau tracer, [18F]RO-948, may be the most favorable. This flexible method can be extended and updated to include new tracers, additional criteria, and modified weights to help researchers and clinicians select the optimal tau PET tracer for specific purposes. Additional work is needed to confirm these results, including a systematic approach to defining and weighting criteria and clinical validation of tracers in different diseases and patient populations.
Collapse
Affiliation(s)
- Ilker Ozsahin
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA
- Operational Research Center in Healthcare, Near East University, Nicosia 99138, TRNC, Turkey
| | | | - Berna Uzun
- Operational Research Center in Healthcare, Near East University, Nicosia 99138, TRNC, Turkey
- Department of Statistics, Carlos III University of Madrid, Getafe, 28903 Madrid, Spain
| | - Dilber Uzun Ozsahin
- Operational Research Center in Healthcare, Near East University, Nicosia 99138, TRNC, Turkey
- Medical Diagnostic Imaging Department, College of Health Sciences & Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Tracy A. Butler
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
40
|
Fu JF, Lois C, Sanchez J, Becker JA, Rubinstein ZB, Thibault E, Salvatore AN, Sari H, Farrell ME, Guehl NJ, Normandin MD, Fakhri GE, Johnson KA, Price JC. Kinetic evaluation and assessment of longitudinal changes in reference region and extracerebral [ 18F]MK-6240 PET uptake. J Cereb Blood Flow Metab 2023; 43:581-594. [PMID: 36420769 PMCID: PMC10063833 DOI: 10.1177/0271678x221142139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/17/2022] [Accepted: 11/06/2022] [Indexed: 11/25/2022]
Abstract
[18F]MK-6240 meningeal/extracerebral off-target binding may impact tau quantification. We examined the kinetics and longitudinal changes of extracerebral and reference regions. [18F]MK-6240 PET was performed in 24 cognitively-normal and eight cognitively-impaired subjects, with arterial samples in 13 subjects. Follow-up scans at 6.1 ± 0.5 (n = 25) and 13.3 ± 0.9 (n = 16) months were acquired. Extracerebral and reference region (cerebellar gray matter (CerGM)-based, cerebral white matter (WM), pons) uptake were evaluated using standardized uptake values (SUV90-110), spectral analysis, and distribution volume. Longitudinal changes in SUV90-110 were examined. The impact of reference region on target region outcomes, partial volume correction (PVC) and regional erosion were evaluated. Eroded WM and pons showed lower variability, lower extracerebral contamination, and lower longitudinal changes than CerGM-based regions. CerGM-based regions resulted larger cross-sectional effect sizes for group differentiation. Extracerebral signal was high in 50% of subjects and exhibited irreversible kinetics and nonsignificant longitudinal changes over one-year but was highly variable at subject-level. PVC resulted in higher variability in reference region uptake and longitudinal changes. Our results suggest that eroded CerGM may be preferred for cross-sectional, whilst eroded WM or pons may be preferred for longitudinal [18F]MK-6240 studies. For CerGM, erosion was necessary (preferred over PVC) to address the heterogenous nature of extracerebral signal.
Collapse
Affiliation(s)
- Jessie Fanglu Fu
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Department of Radiology, Boston, MA, USA
| | - Cristina Lois
- Harvard Medical School, Department of Radiology, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Justin Sanchez
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - J Alex Becker
- Harvard Medical School, Department of Radiology, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Zoe B Rubinstein
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Emma Thibault
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew N Salvatore
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Hasan Sari
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Department of Radiology, Boston, MA, USA
| | | | - Nicolas J Guehl
- Harvard Medical School, Department of Radiology, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Marc D Normandin
- Harvard Medical School, Department of Radiology, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Georges El Fakhri
- Harvard Medical School, Department of Radiology, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Keith A Johnson
- Harvard Medical School, Department of Radiology, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Julie C Price
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Department of Radiology, Boston, MA, USA
| |
Collapse
|
41
|
Mondal R, Sandhu YK, Kamalia VM, Delaney BA, Syed AU, Nguyen GAH, Moran TR, Limpengco RR, Liang C, Mukherjee J. Measurement of Aβ Amyloid Plaques and Tau Protein in Postmortem Human Alzheimer’s Disease Brain by Autoradiography Using [18F]Flotaza, [125I]IBETA, [124/125I]IPPI and Immunohistochemistry Analysis Using QuPath. Biomedicines 2023; 11:biomedicines11041033. [PMID: 37189652 DOI: 10.3390/biomedicines11041033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
High-resolution scans of immunohistochemical (IHC) stains of Alzheimer’s disease (AD) brain slices and radioligand autoradiography both provide information about the distribution of Aβ plaques and Tau, the two common proteinopathies in AD. Accurate assessment of the amount and regional location of Aβ plaques and Tau is essential to understand the progression of AD pathology. Our goal was to develop a quantitative method for the analysis of IHC–autoradiography images. Postmortem anterior cingulate (AC) and corpus callosum (CC) from AD and control (CN) subjects were IHC stained with anti-Aβ for Aβ plaques and autoradiography with [18F]flotaza and [125I]IBETA for Aβ plaques. For Tau, [124I]IPPI, a new radiotracer, was synthesized and evaluated in the AD brain. For Tau imaging, brain slices were IHC stained with anti-Tau and autoradiography using [125I]IPPI and [124I]IPPI. Annotations for Aβ plaques and Tau using QuPath for training and pixel classifiers were generated to measure the percent of the area of Aβ plaques and Tau in each slice. The binding of [124I]IPPI was observed in all AD brains with an AC/CC ratio > 10. Selectivity to Tau was shown by blocking [124I]IPPI with MK-6240. Percent positivity for Aβ plaques was 4–15%, and for Tau, it was 1.3 to 35%. All IHC Aβ plaque-positive subjects showed [18F]flotaza and [125I]IBETA binding with a positive linear correlation (r2 > 0.45). Tau-positive subjects showed [124/125I]IPPI binding with a stronger positive linear correlation (r2 > 0.80). This quantitative IHC–autoradiography approach provides an accurate measurement of Aβ plaques and Tau within and across subjects.
Collapse
|
42
|
Ennis GE, Betthauser TJ, Koscik RL, Chin NA, Christian BT, Asthana S, Johnson SC, Bendlin BB. The relationship of insulin resistance and diabetes to tau PET SUVR in middle-aged to older adults. Alzheimers Res Ther 2023; 15:55. [PMID: 36932429 PMCID: PMC10022314 DOI: 10.1186/s13195-023-01180-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/31/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND Insulin resistance (IR) and type 2 diabetes have been found to increase the risk for Alzheimer's clinical syndrome in epidemiologic studies but have not been associated with tau tangles in neuropathological research and have been inconsistently associated with cerebrospinal fluid P-tau181. IR and type 2 diabetes are well-recognized vascular risk factors. Some studies suggest that cardiovascular risk may act synergistically with cortical amyloid to increase tau measured using tau PET. Utilizing data from largely nondemented middle-aged and older adult cohorts enriched for AD risk, we investigated the association of IR and diabetes to tau PET and whether amyloid moderated those relationships. METHODS Participants were enrolled in either the Wisconsin Registry for Alzheimer's Prevention (WRAP) or Wisconsin Alzheimer's Disease Research Center (WI-ADRC) Clinical Core. Two partially overlapping samples were studied: a sample characterized using HOMA-IR (n=280 WRAP participants) and a sample characterized on diabetic status (n=285 WRAP and n=109 WI-ADRC). IR was measured using the homeostasis model assessment of insulin resistance (HOMA-IR). Tau PET employing the radioligand 18F-MK-6240 was used to detect AD-specific aggregated tau. Linear regression tested the relationship of IR and diabetic status to tau PET standardized uptake value ratio (SUVR) within the entorhinal cortex and whether relationships were moderated by amyloid assessed by amyloid PET distribution volume ratio (DVR) and amyloid PET positivity status. RESULTS Neither HOMA-IR nor diabetic status was significantly associated with tau PET SUVR. The relationship between IR and tau PET SUVR was not moderated by amyloid PET DVR or positivity status. The association between diabetic status and tau PET SUVR was not significantly moderated by amyloid PET DVR but was significantly moderated by amyloid PET positivity status. Among the amyloid PET-positive participants, the estimated marginal tau PET SUVR mean was higher in the diabetic (n=6) relative to the nondiabetic group (n=88). CONCLUSION Findings indicate that IR may not be related to tau in generally healthy middle-aged and older adults who are in the early stages of the AD clinicopathologic continuum but suggest the need for additional research to investigate whether a synergistic relationship between type 2 diabetes and amyloid is associated with increased tau levels.
Collapse
Affiliation(s)
- Gilda E Ennis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca Langhough Koscik
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Nathaniel A Chin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Bradley T Christian
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| |
Collapse
|
43
|
Malarte ML, Gillberg PG, Kumar A, Bogdanovic N, Lemoine L, Nordberg A. Discriminative binding of tau PET tracers PI2620, MK6240 and RO948 in Alzheimer's disease, corticobasal degeneration and progressive supranuclear palsy brains. Mol Psychiatry 2023; 28:1272-1283. [PMID: 36447011 PMCID: PMC10005967 DOI: 10.1038/s41380-022-01875-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 12/02/2022]
Abstract
Recent mechanistic and structural studies have challenged the classical tauopathy classification approach and revealed the complexity and heterogeneity of tau pathology in Alzheimer's disease (AD) and primary tauopathies such as corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP), progressing beyond distinct tau isoforms. In this multi-tau tracer study, we focused on the new second-generation tau PET tracers PI2620, MK6240 and RO948 to investigate this tau complexity in AD, CBD, and PSP brains using post-mortem radioligand binding studies and autoradiography of large and small frozen brain sections. Saturation binding studies indicated multiple binding sites for 3H-PI2620 in AD, CBD and PSP brains with different binding affinities (Kd ranging from 0.2 to 0.7 nM) and binding site densities (following the order: BmaxAD > BmaxCBD > BmaxPSP). Competitive binding studies complemented these findings, demonstrating the presence of two binding sites [super-high affinity (SHA): IC50(1) = 8.1 pM; and high affinity (HA): IC50(2) = 4.9 nM] in AD brains. Regional binding distribution studies showed that 3H-PI2620 could discriminate between AD (n = 6) and control cases (n = 9), especially in frontal cortex and temporal cortex tissue (p < 0.001) as well as in the hippocampal region (p = 0.02). 3H-PI2620, 3H-MK6240 and 3H-RO948 displayed similar binding behaviour in AD brains (in both homogenate competitive studies and one large frozen hemispherical brain section autoradiography studies) in terms of binding affinities, number of sites and regional patterns. Our small section autoradiography studies in the frontal cortex of CBD (n = 3) and PSP brains (n = 2) showed high specificity for 3H-PI2620 but not for 3H-MK6240 or 3H-RO948. Our findings clearly demonstrate different binding properties among the second-generation tau PET tracers, which may assist in further understanding of tau heterogeneity in AD versus non-AD tauopathies and suggests potential for development of pure selective 4R tau PET tracers.
Collapse
Grants
- Stiftelsen för Strategisk Forskning (Swedish Foundation for Strategic Research)
- Stiftelsen Olle Engkvist Byggmästare
- Svenska Forskningsrådet Formas (Swedish Research Council Formas)
- Stockholms Läns Landsting (Stockholm County Council)
- Hjärnfonden (Swedish Brain Foundation)
- Stockholm County Council -Karolinska Institute regional agreement on medical training and clinical research (ALF grant),the Swedish Alzheimer Foundation, the Foundation for Old Servants, Gun and Bertil Stohne’s Foundation, the KI Foundation for Geriatric Diseases, the Swedish Dementia Foundation, the Center for Innovative Medicine (CIMED) Region Stockholm, the Michael J Fox Foundation (MJFF-019728), the Alzheimer Association USA (AARF -21-848395), and the Recherche sur Alzheimer Foundation (Paris, France).
Collapse
Affiliation(s)
- Mona-Lisa Malarte
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Per-Göran Gillberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Nenad Bogdanovic
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Laëtitia Lemoine
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
44
|
Varlow C, Vasdev N. Evaluation of Tau Radiotracers in Chronic Traumatic Encephalopathy. J Nucl Med 2023; 64:460-465. [PMID: 36109185 PMCID: PMC10071800 DOI: 10.2967/jnumed.122.264404] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a neurologic disorder associated with head injuries, diagnosed by the perivascular accumulation of hyperphosphorylated tau protein (phospho-tau) identified at autopsy. Tau PET radiopharmaceuticals developed for imaging Alzheimer disease are under evaluation for brain injuries. The goal of this study was to conduct a head-to-head in vitro evaluation of 5 tau PET radiotracers in subjects pathologically diagnosed with CTE. Methods: Autoradiography was used to assess the specific binding and distribution of 3H-flortaucipir (also known as Tauvid, AV-1451, and T807), 3H-MK-6240 (also known as florquinitau), 3H-PI-2620, 3H-APN-1607 (also known as PM-PBB3 and florzolotau), and 3H-CBD-2115 (also known as 3H-OXD-2115) in fresh-frozen human postmortem CTE brain tissue (stages I-IV). Immunohistochemistry was performed for phospho-tau with AT8, 3R tau with RD3, 4R tau with RD4 and amyloid-β with 6F/3D antibodies. Tau target density (maximum specific binding) was quantified by saturation analysis with 3H-flortaucipir in tissue sections. Results: 3H-flortaucipir demonstrated a positive signal in all CTE cases examined, with varying degrees of specific binding (68.7% ± 10.5%; n = 12) defined by homologous blockade and to a lesser extent by heterologous blockade with MK-6240 (27.3% ± 13.6%; n = 12). The 3H-flortaucipir signal was also displaced by the monoamine oxidase (MAO)-A inhibitor clorgyline (43.9% ± 4.6%; n = 3), indicating off-target binding to MAO-A. 3H-APN-1607 was moderately displaced in homologous blocking studies and was not displaced by 3H-flortaucipir; however, substantial displacement was observed when blocking with the β-amyloid-targeting compound NAV-4694. 3H-MK-6240 and 3H-PI-2620 had negligible binding in all but 2 CTE IV cases, and binding may be attributed to pathology severity or mixed Alzheimer disease/CTE pathology. 3H-CBD-2115 showed moderate binding, displaced under homologous blockade, and aligned with 4R-tau immunostaining. Conclusion: In human CTE tissues, 3H-flortaucipir and 3H-APN-1607 revealed off-target binding to MAO-A and amyloid-β, respectively, and should be considered if these radiotracers are used in PET imaging studies of patients with brain injuries. 3H-MK-6240 and 3H-PI-2620 bind to CTE tau in severe- or mixed-pathology cases, and their respective 18F PET radiotracers warrant further evaluation in patients with severe suspected CTE.
Collapse
Affiliation(s)
- Cassis Varlow
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Tissot C, Servaes S, Lussier FZ, Ferrari-Souza JP, Therriault J, Ferreira PCL, Bezgin G, Bellaver B, Leffa DT, Mathotaarachchi SS, Chamoun M, Stevenson J, Rahmouni N, Kang MS, Pallen V, Margherita-Poltronetti N, Wang YT, Fernandez-Arias J, Benedet AL, Zimmer ER, Soucy JP, Tudorascu DL, Cohen AD, Sharp M, Gauthier S, Massarweh G, Lopresti B, Klunk WE, Baker SL, Villemagne VL, Rosa-Neto P, Pascoal TA. The Association of Age-Related and Off-Target Retention with Longitudinal Quantification of [ 18F]MK6240 Tau PET in Target Regions. J Nucl Med 2023; 64:452-459. [PMID: 36396455 PMCID: PMC10071794 DOI: 10.2967/jnumed.122.264434] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022] Open
Abstract
6-(fluoro-18F)-3-(1H-pyrrolo[2,3-c]pyridin-1-yl)isoquinolin-5-amine ([18F]MK6240) tau PET tracer quantifies the brain tau neurofibrillary tangle load in Alzheimer disease. The aims of our study were to test the stability of common reference region estimates in the cerebellum over time and across diagnoses and evaluate the effects of age-related and off-target retention on the longitudinal quantification of [18F]MK6240 in target regions. Methods: We assessed reference, target, age-related, and off-target regions in 125 individuals across the aging and Alzheimer disease spectrum with longitudinal [18F]MK6240 SUVs and SUV ratios (SUVRs) (mean ± SD, 2.25 ± 0.40 y of follow-up). We obtained SUVR from meninges, exhibiting frequent off-target retention with [18F]MK6240. Additionally, we compared tracer uptake between 37 cognitively unimpaired young (CUY) (mean age, 23.41 ± 3.33 y) and 27 cognitively unimpaired older (CU) adults (amyloid-β-negative and tau-negative, 58.50 ± 9.01 y) to identify possible nonvisually apparent, age-related signal. Two-tailed t testing and Pearson correlation testing were used to determine the difference between groups and associations between changes in region uptake, respectively. Results: Inferior cerebellar gray matter SUV did not differ on the basis of diagnosis and amyloid-β status, cross-sectionally and over time. [18F]MK6240 uptake significantly differed between CUY and CU adults in the putamen or pallidum (affecting ∼75% of the region) and in the Braak II region (affecting ∼35%). Changes in meningeal and putamen or pallidum SUVRs did not significantly differ from zero, nor did they vary across diagnostic groups. We did not observe significant correlations between longitudinal changes in age-related or meningeal off-target retention and changes in target regions, whereas changes in all target regions were strongly correlated. Conclusion: Inferior cerebellar gray matter was similar across diagnostic groups cross-sectionally and stable over time and thus was deemed a suitable reference region for quantification. Despite not being visually perceptible, [18F]MK6240 has age-related retention in subcortical regions, at a much lower magnitude but topographically colocalized with significant off-target signal of the first-generation tau tracers. The lack of correlation between changes in age-related or meningeal and target retention suggests little influence of possible off-target signals on longitudinal tracer quantification. Nevertheless, the age-related retention in the Braak II region needs to be further investigated. Future postmortem studies should elucidate the source of the newly reported age-related [18F]MK6240 signal, and in vivo studies should further explore its impact on tracer quantification.
Collapse
Affiliation(s)
- Cécile Tissot
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Stijn Servaes
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Firoza Z Lussier
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - João Pedro Ferrari-Souza
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Porto Alegre, Brazil
| | - Joseph Therriault
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Pâmela C L Ferreira
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gleb Bezgin
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Bruna Bellaver
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Graduate Program in Biological Sciences: Biochemistry, Porto Alegre, Brazil
| | - Douglas Teixeira Leffa
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sulantha S Mathotaarachchi
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Mira Chamoun
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Jenna Stevenson
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Min Su Kang
- Artificial Intelligence and Computational Neurosciences Lab, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- L.C. Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Vanessa Pallen
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Nina Margherita-Poltronetti
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Yi-Ting Wang
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | - Jaime Fernandez-Arias
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
| | | | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Porto Alegre, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Dana L Tudorascu
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Annie D Cohen
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Serge Gauthier
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
- Douglas Mental Health Institute, Montreal, Quebec, Canada
| | - Gassan Massarweh
- Department of Radiochemistry, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Brian Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - William E Klunk
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Victor L Villemagne
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Pedro Rosa-Neto
- McGill University, Montreal, Quebec, Canada
- McGill University Research Center for Studies in Aging, Montreal, Quebec, Canada
- Montreal Neurological Institute, Montreal, Quebec, Canada
- Douglas Mental Health Institute, Montreal, Quebec, Canada
| | - Tharick A Pascoal
- Departments of Psychiatry and Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
| |
Collapse
|
46
|
Mohammadi Z, Alizadeh H, Marton J, Cumming P. The Sensitivity of Tau Tracers for the Discrimination of Alzheimer's Disease Patients and Healthy Controls by PET. Biomolecules 2023; 13:290. [PMID: 36830659 PMCID: PMC9953528 DOI: 10.3390/biom13020290] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Hyperphosphorylated tau aggregates, also known as neurofibrillary tangles, are a hallmark neuropathological feature of Alzheimer's disease (AD). Molecular imaging of tau by positron emission tomography (PET) began with the development of [18F]FDDNP, an amyloid β tracer with off-target binding to tau, which obtained regional specificity through the differing distributions of amyloid β and tau in AD brains. A concerted search for more selective and affine tau PET tracers yielded compounds belonging to at least eight structural categories; 18F-flortaucipir, known variously as [18F]-T807, AV-1451, and Tauvid®, emerged as the first tau tracer approved by the American Food and Drug Administration. The various tau tracers differ concerning their selectivity over amyloid β, off-target binding at sites such as monoamine oxidase and neuromelanin, and degree of uptake in white matter. While there have been many reviews of molecular imaging of tau in AD and other conditions, there has been no systematic comparison of the fitness of the various tracers for discriminating between AD patient and healthy control (HC) groups. In this narrative review, we endeavored to compare the binding properties of the various tau tracers in vitro and the effect size (Cohen's d) for the contrast by PET between AD patients and age-matched HC groups. The available tracers all gave good discrimination, with Cohen's d generally in the range of two-three in culprit brain regions. Overall, Cohen's d was higher for AD patient groups with more severe illness. Second-generation tracers, while superior concerning off-target binding, do not have conspicuously higher sensitivity for the discrimination of AD and HC groups. We suppose that available pharmacophores may have converged on a maximal affinity for tau fibrils, which may limit the specific signal imparted in PET studies.
Collapse
Affiliation(s)
- Zohreh Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hadi Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Straße 10-14, D-01454 Radeberg, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Freiburgstraße 18, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
47
|
Krishnadas N, Doré V, Robertson JS, Ward L, Fowler C, Masters CL, Bourgeat P, Fripp J, Villemagne VL, Rowe CC. Rates of regional tau accumulation in ageing and across the Alzheimer's disease continuum: an AIBL 18F-MK6240 PET study. EBioMedicine 2023; 88:104450. [PMID: 36709581 PMCID: PMC9900352 DOI: 10.1016/j.ebiom.2023.104450] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 12/15/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Tau positron emission tomography (PET) imaging enables longitudinal observation of tau accumulation in Alzheimer's disease (AD). 18F-MK6240 is a high affinity tracer for the paired helical filaments of tau in AD, widely used in clinical trials, despite sparse longitudinal natural history data. We aimed to evaluate the natural history of tau accumulation, and the impact of disease stage and reference region on the magnitude and effect size of regional change. METHODS One hundred and eighty-four participants: 89 cognitively unimpaired (CU) beta-amyloid negative (Aβ-), 44 CU Aβ+, 51 cognitively impaired Aβ+ (26 with mild cognitive impairment [MCI] and 25 with dementia) had follow-up 18F-MK6240 PET for one to four years (median 1.48). Regional standardised uptake value ratios (SUVR) were generated. Two reference regions were examined: cerebellar cortex and eroded subcortical white matter. FINDINGS CU Aβ- participants had very low rates of tau accumulation in the mesial temporal lobe (MTL). In CU Aβ+, significantly higher rate of accumulation was seen in the MTL (particularly the amygdala), extending into the inferior temporal lobes. In MCI Aβ+, the rate of accumulation was greatest in the lateral temporal, parietal and lateral occipital cortex, and plateaued in the MTL. Accumulation was global in AD Aβ+, except for a plateau in the MTL. The eroded subcortical white matter reference region showed no significant advantage over the cerebellar cortex and appeared prone to spill-over in AD participants. Data fitting suggested approximately 15-20 years to accumulate tau to typical AD levels. INTERPRETATION Tau accumulation occurs slowly. Rates vary according to brain region, disease stage and tend to plateau at high levels. Rates of tau accumulation are best measured in the MTL and inferior temporal cortex in preclinical AD and in large neocortical areas, in MCI and AD. FUNDING NHMRC; Cerveau Technologies.
Collapse
Affiliation(s)
- Natasha Krishnadas
- Florey Department of Neurosciences & Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia; Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, 3084, Australia.
| | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, 3084, Australia; Health and Biosecurity Flagship, The Australian eHealth Research Centre, Melbourne, Victoria, Australia
| | - Joanne S Robertson
- Florey Institute of Neurosciences & Mental Health, Parkville, Victoria, 3010, Australia
| | - Larry Ward
- Florey Institute of Neurosciences & Mental Health, Parkville, Victoria, 3010, Australia
| | - Christopher Fowler
- Florey Institute of Neurosciences & Mental Health, Parkville, Victoria, 3010, Australia
| | - Colin L Masters
- Florey Institute of Neurosciences & Mental Health, Parkville, Victoria, 3010, Australia
| | - Pierrick Bourgeat
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Brisbane, Queensland, Australia
| | - Jurgen Fripp
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Brisbane, Queensland, Australia
| | | | - Christopher C Rowe
- Florey Department of Neurosciences & Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia; Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, 3084, Australia; Florey Institute of Neurosciences & Mental Health, Parkville, Victoria, 3010, Australia
| |
Collapse
|
48
|
Abstract
Imaging of mild traumatic brain injury (TBI) using conventional techniques such as CT or MRI often results in no specific imaging correlation that would explain cognitive and clinical symptoms. Molecular imaging of mild TBI suggests that secondary events after injury can be detected using PET. However, no single specific pattern emerges that can aid in diagnosing the injury or determining the prognosis of the long-term behavioral profiles, indicating the heterogeneous and diffuse nature of TBI. Chronic traumatic encephalopathy, a primary tauopathy, has been shown to be strongly associated with repetitive TBI. In vivo data on the available tau PET tracers, however, have produced mixed results and overall low retention profiles in athletes with a history of repetitive mild TBI. Here, we emphasize that the lack of a mechanistic understanding of chronic TBI has posed a challenge when interpreting the results of molecular imaging biomarkers. We advocate for better target identification, improved analysis techniques such as machine learning or artificial intelligence, and novel tracer development.
Collapse
Affiliation(s)
- Gérard N. Bischof
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany;,Institute for Neuroscience and Medicine II–Molecular Organization of the Brain, Research Center Juelich, Juelich, Germany; and
| | - Donna J. Cross
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| |
Collapse
|
49
|
Engels-Domínguez N, Koops EA, Prokopiou PC, Van Egroo M, Schneider C, Riphagen JM, Singhal T, Jacobs HIL. State-of-the-art imaging of neuromodulatory subcortical systems in aging and Alzheimer's disease: Challenges and opportunities. Neurosci Biobehav Rev 2023; 144:104998. [PMID: 36526031 PMCID: PMC9805533 DOI: 10.1016/j.neubiorev.2022.104998] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
Primary prevention trials have shifted their focus to the earliest stages of Alzheimer's disease (AD). Autopsy data indicates that the neuromodulatory subcortical systems' (NSS) nuclei are specifically vulnerable to initial tau pathology, indicating that these nuclei hold great promise for early detection of AD in the context of the aging brain. The increasing availability of new imaging methods, ultra-high field scanners, new radioligands, and routine deep brain stimulation implants has led to a growing number of NSS neuroimaging studies on aging and neurodegeneration. Here, we review findings of current state-of-the-art imaging studies assessing the structure, function, and molecular changes of these nuclei during aging and AD. Furthermore, we identify the challenges associated with these imaging methods, important pathophysiologic gaps to fill for the AD NSS neuroimaging field, and provide future directions to improve our assessment, understanding, and clinical use of in vivo imaging of the NSS.
Collapse
Affiliation(s)
- Nina Engels-Domínguez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Elouise A Koops
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Prokopis C Prokopiou
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maxime Van Egroo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Christoph Schneider
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tarun Singhal
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
50
|
Butler T, Chiang GC, Niogi SN, Wang XH, Skudin C, Tanzi E, Wickramasuriya N, Spiegel J, Maloney T, Pahlajani S, Zhou L, Morim S, Rusinek H, Normandin M, Dyke JP, Fung EK, Li Y, Glodzik L, Razlighi QR, Shah SA, de Leon M. Tau PET following acute TBI: Off-target binding to blood products, tauopathy, or both? FRONTIERS IN NEUROIMAGING 2022; 1:958558. [PMID: 36876118 PMCID: PMC9979975 DOI: 10.3389/fnimg.2022.958558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/15/2022] [Indexed: 11/07/2022]
Abstract
Repeated mild Traumatic Brain Injury (TBI) is a risk factor for Chronic Traumatic Encephalopathy (CTE), characterized pathologically by neurofibrillary tau deposition in the depths of brain sulci and surrounding blood vessels. The mechanism by which TBI leads to CTE remains unknown but has been posited to relate to axonal shear injury leading to release and possibly deposition of tau at the time of injury. As part of an IRB-approved study designed to learn how processes occurring acutely after TBI may predict later proteinopathy and neurodegeneration, we performed tau PET using 18F-MK6240 and MRI within 14 days of complicated mild TBI in three subjects. PET radiotracer accumulation was apparent in regions of traumatic hemorrhage in all subjects, with prominent intraparenchymal PET signal in one young subject with a history of repeated sports-related concussions. These results are consistent with off-target tracer binding to blood products as well as possible on-target binding to chronically and/or acutely-deposited neurofibrillary tau. Both explanations are highly relevant to applying tau PET to understanding TBI and CTE. Additional study is needed to assess the potential utility of tau PET in understanding how processes occurring acutely after TBI, such as release and deposition of tau and blood from damaged axons and blood vessels, may relate to development CTE years later.
Collapse
Affiliation(s)
- Tracy Butler
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Gloria C. Chiang
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Sumit Narayan Niogi
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Xiuyuan Hugh Wang
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Carly Skudin
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Emily Tanzi
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | | | - Jonathan Spiegel
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Thomas Maloney
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Silky Pahlajani
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Liangdong Zhou
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Simon Morim
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Henry Rusinek
- New York University Grossman School of Medicine, New York, NY, United States
| | - Marc Normandin
- Department of Radiology Harvard Medical School, Boston, MA, United States
| | - Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Edward K. Fung
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Yi Li
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Lidia Glodzik
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | | | - Sudhin A. Shah
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| | - Mony de Leon
- Department of Radiology, Weill Cornell Medicine,, New York, NY, United States
| |
Collapse
|