151
|
Evaluation of the Feasibility of Screening Tau Radiotracers Using an Amyloid Biomathematical Screening Methodology. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2019; 2018:6287913. [PMID: 30662517 PMCID: PMC6314003 DOI: 10.1155/2018/6287913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/06/2018] [Accepted: 11/21/2018] [Indexed: 11/17/2022]
Abstract
The purpose of this study is to evaluate the feasibility of extending a previously developed amyloid biomathematical screening methodology to support the screening of tau radiotracers during compound development. 22 tau-related PET radiotracers were investigated. For each radiotracer, in silico MLogP, V x, and in vitro K D were input into the model to predict the in vivo K 1, k 2, and BPND under healthy control (HC), mild cognitive impaired (MCI), and Alzheimer's disease (AD) conditions. These kinetic parameters were used to simulate the time activity curves (TACs) in the target regions of HC, MCI, and AD and a reference region. Standardized uptake value ratios (SUVR) were determined from the integrated area under the TACs of the target region over the reference region within a default time window of 90-110 min. The predicted K 1, k 2, and BPND values were compared with the clinically observed values. The TACs and SUVR distributions were also simulated with population variations and noise. Finally, the clinical usefulness index (CUI) ranking was compared with clinical comparison results. The TACs and SUVR distributions differed for tau radiotracers with lower tau selectivity. The CUI values ranged from 0.0 to 16.2, with 6 out of 9 clinically applied tau radiotracers having CUI values higher than the recommend CUI value of 3.0. The differences between the clinically observed TACs and SUVR results showed that the evaluation of the clinical usefulness of tau radiotracer based on single target binding could not fully reflect in vivo tau binding. The screening methodology requires further study to improve the accuracy of screening tau radiotracers. However, the higher CUI rankings of clinically applied tau radiotracers with higher signal-to-noise ratio supported the use of the screening methodology in radiotracer development by allowing comparison of candidate radiotracers with clinically applied radiotracers based on SUVR, with respect to binding to a single target.
Collapse
|
152
|
Leuzy A, Chiotis K, Lemoine L, Gillberg PG, Almkvist O, Rodriguez-Vieitez E, Nordberg A. Tau PET imaging in neurodegenerative tauopathies-still a challenge. Mol Psychiatry 2019; 24:1112-1134. [PMID: 30635637 PMCID: PMC6756230 DOI: 10.1038/s41380-018-0342-8] [Citation(s) in RCA: 382] [Impact Index Per Article: 76.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/19/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
The accumulation of pathological misfolded tau is a feature common to a collective of neurodegenerative disorders known as tauopathies, of which Alzheimer's disease (AD) is the most common. Related tauopathies include progressive supranuclear palsy (PSP), corticobasal syndrome (CBS), Down's syndrome (DS), Parkinson's disease (PD), and dementia with Lewy bodies (DLB). Investigation of the role of tau pathology in the onset and progression of these disorders is now possible due the recent advent of tau-specific ligands for use with positron emission tomography (PET), including first- (e.g., [18F]THK5317, [18F]THK5351, [18F]AV1451, and [11C]PBB3) and second-generation compounds [namely [18F]MK-6240, [18F]RO-948 (previously referred to as [18F]RO69558948), [18F]PI-2620, [18F]GTP1, [18F]PM-PBB3, and [18F]JNJ64349311 ([18F]JNJ311) and its derivative [18F]JNJ-067)]. In this review we describe and discuss findings from in vitro and in vivo studies using both initial and new tau ligands, including their relation to biomarkers for amyloid-β and neurodegeneration, and cognitive findings. Lastly, methodological considerations for the quantification of in vivo ligand binding are addressed, along with potential future applications of tau PET, including therapeutic trials.
Collapse
Affiliation(s)
- Antoine Leuzy
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Chiotis
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden ,0000 0000 9241 5705grid.24381.3cTheme Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Laetitia Lemoine
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Per-Göran Gillberg
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ove Almkvist
- 0000 0004 1937 0626grid.4714.6Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden ,0000 0004 1936 9377grid.10548.38Department of Psychology, Stockholm University, Stockholm, Sweden
| | - Elena Rodriguez-Vieitez
- 0000 0004 1937 0626grid.4714.6Division 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 Aging, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
153
|
Hopewell R, Ross K, Kostikov A, Pascoal TA, Alberti T, Lacatus-Samoila M, Soucy JP, Bennacef I, Kobayashi E, Kang MS, Rosa-Neto P, Massarweh G. A simplified radiosynthesis of [18
F]MK-6240 for tau PET imaging. J Labelled Comp Radiopharm 2018; 62:109-114. [DOI: 10.1002/jlcr.3695] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/01/2018] [Accepted: 11/06/2018] [Indexed: 11/12/2022]
Affiliation(s)
- Robert Hopewell
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Karen Ross
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Alexey Kostikov
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Tharick A. Pascoal
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
- Translational Neuroimaging Laboratory; The McGill University Research Centre for Studies in Aging; Montreal Quebec Canada
| | - Thais Alberti
- Translational Neuroimaging Laboratory; The McGill University Research Centre for Studies in Aging; Montreal Quebec Canada
| | | | - Jean-Paul Soucy
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Idriss Bennacef
- Translational Biomarkers; Merck & Co, Inc; West Point Pennsylvania USA
| | - Eliane Kobayashi
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| | - Min Su Kang
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
- Translational Neuroimaging Laboratory; The McGill University Research Centre for Studies in Aging; Montreal Quebec Canada
| | - Pedro Rosa-Neto
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
- Translational Neuroimaging Laboratory; The McGill University Research Centre for Studies in Aging; Montreal Quebec Canada
| | - Gassan Massarweh
- Montreal Neurological Institute; McGill University; Montreal Quebec Canada
| |
Collapse
|
154
|
Schöll M, Maass A, Mattsson N, Ashton NJ, Blennow K, Zetterberg H, Jagust W. Biomarkers for tau pathology. Mol Cell Neurosci 2018; 97:18-33. [PMID: 30529601 PMCID: PMC6584358 DOI: 10.1016/j.mcn.2018.12.001] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/01/2018] [Indexed: 12/14/2022] Open
Abstract
The aggregation of fibrils of hyperphosphorylated and C-terminally truncated microtubule-associated tau protein characterizes 80% of all dementia disorders, the most common neurodegenerative disorders. These so-called tauopathies are hitherto not curable and their diagnosis, especially at early disease stages, has traditionally proven difficult. A keystone in the diagnosis of tauopathies was the development of methods to assess levels of tau protein in vivo in cerebrospinal fluid, which has significantly improved our knowledge about these conditions. Tau proteins have also been measured in blood, but the importance of tau-related changes in blood is still unclear. The recent addition of positron emission tomography ligands to visualize, map and quantify tau pathology has further contributed with information about the temporal and spatial characteristics of tau accumulation in the living brain. Together, the measurement of tau with fluid biomarkers and positron emission tomography constitutes the basis for a highly active field of research. This review describes the current state of biomarkers for tau biomarkers derived from neuroimaging and from the analysis of bodily fluids and their roles in the detection, diagnosis and prognosis of tau-associated neurodegenerative disorders, as well as their associations with neuropathological findings, and aims to provide a perspective on how these biomarkers might be employed prospectively in research and clinical settings. Biomarkers for tau pathology are now essential to the research framework in the diagnosis of Alzheimer's disease (AD) Measurement of t- and p-tau has been possible in cerebrospinal fluid (CSF) for some time, the recent development of positron emission tomography (PET) ligands binding to tau has added the possibility to map and quantify tau in the living brain First-generation tau PET ligands bind predominantly to AD-typical 3R/4R tau isoforms and exhibit off-target binding that can limit accurate ligand uptake quantification Second-generation tau PET ligands appear to bind to comparable binding sites but exhibit fewer issues with brain off-target binding Biomarkers for tau derived from CSF analysis and PET could provide complementary information about disease state and stage At this time, T-tau, but not p-tau, can be reliably measured in plasma using ultra-sensitive immunoassays.
Collapse
Affiliation(s)
- Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden; Clinical Memory Research Unit, Lund University, Malmö, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.
| | - Anne Maass
- German Center for Neurodegenerative Diseases, Magdeburg, Germany; Helen Wills Neuroscience Institute, University of California, Berkeley, USA
| | - Niklas Mattsson
- Clinical Memory Research Unit, Lund University, Malmö, Sweden; Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Nicholas J Ashton
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden; King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, UK
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, USA; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
155
|
Harada R, Okamura N, Furumoto S, Yanai K. Imaging Protein Misfolding in the Brain Using β-Sheet Ligands. Front Neurosci 2018; 12:585. [PMID: 30186106 PMCID: PMC6110819 DOI: 10.3389/fnins.2018.00585] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases characterized by pathological protein accumulation in cells are termed “proteinopathies.” Although various protein aggregates share cross-β-sheet structures, actual conformations vary among each type of protein deposit. Recent progress in the development of radiotracers for positron emission tomography (PET) has enabled the visualization of protein aggregates in living brains. Amyloid PET tracers have been developed, and are widely used for the diagnosis of Alzheimer’s disease and non-invasive assessment of amyloid burden in clinical trials of anti-dementia drugs. Furthermore, several tau PET tracers have been successfully developed and used in the clinical studies. However, recent studies have identified the presence of off-target binding of radiotracers in areas of tau deposition, suggesting that concomitant neuroinflammatory changes might affect tracer binding. In contrast to amyloid and tau PET, there are no established tracers for imaging Lewy bodies in the human brain. In this review, we describe lessons learned from the development of PET tracers and discuss the future direction of tracer development for protein misfolding diseases.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| |
Collapse
|
156
|
|
157
|
Pascoal TA, Shin M, Kang MS, Chamoun M, Chartrand D, Mathotaarachchi S, Bennacef I, Therriault J, Ng KP, Hopewell R, Bouhachi R, Hsiao HH, Benedet AL, Soucy JP, Massarweh G, Gauthier S, Rosa-Neto P. In vivo quantification of neurofibrillary tangles with [ 18F]MK-6240. ALZHEIMERS RESEARCH & THERAPY 2018; 10:74. [PMID: 30064520 PMCID: PMC6069775 DOI: 10.1186/s13195-018-0402-y] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/06/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Imaging agents capable of quantifying the brain's tau aggregates will allow a more precise staging of Alzheimer's disease (AD). The aim of the present study was to examine the in vitro properties as well as the in vivo kinetics, using gold standard methods, of the novel positron emission tomography (PET) tau imaging agent [18F]MK-6240. METHODS In vitro properties of [18F]MK-6240 were estimated with autoradiography in postmortem brain tissues of 14 subjects (seven AD patients and seven age-matched controls). In vivo quantification of [18F]MK-6240 binding was performed in 16 subjects (four AD patients, three mild cognitive impairment patients, six healthy elderly individuals, and three healthy young individuals) who underwent 180-min dynamic scans; six subjects had arterial sampling for metabolite correction. Simplified approaches for [18F]MK-6240 quantification were validated using full kinetic modeling with metabolite-corrected arterial input function. All participants also underwent amyloid-PET and structural magnetic resonance imaging. RESULTS In vitro [18F]MK-6240 uptake was higher in AD patients than in age-matched controls in brain regions expected to contain tangles such as the hippocampus, whereas no difference was found in the cerebellar gray matter. In vivo, [18F]MK-6240 displayed favorable kinetics with rapid brain delivery and washout. The cerebellar gray matter had low binding across individuals, showing potential for use as a reference region. A reversible two-tissue compartment model well described the time-activity curves across individuals and brain regions. Distribution volume ratios using the plasma input and standardized uptake value ratios (SUVRs) calculated after the binding approached equilibrium (90 min) were correlated and higher in mild cognitive impairment or AD dementia patients than in controls. Reliability analysis revealed robust SUVRs calculated from 90 to 110 min, while earlier time points provided inaccurate estimates. CONCLUSIONS This evaluation shows an [18F]MK-6240 distribution in concordance with postmortem studies and that simplified quantitative approaches such as the SUVR offer valid estimates of neurofibrillary tangle load 90 min post injection. [18F]MK-6240 is a promising tau tracer with the potential to be applied in the disease diagnosis and assessment of therapeutic interventions.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada.,Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Monica Shin
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada.,Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Mira Chamoun
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Daniel Chartrand
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada.,Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Idriss Bennacef
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, 19486, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Kok Pin Ng
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Robert Hopewell
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Reda Bouhachi
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Hung-Hsin Hsiao
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Andrea L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Jean-Paul Soucy
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Gassan Massarweh
- Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, 6825 LaSalle Boulevard, Verdun, QC, H4H 1R3, Canada. .,Montreal Neurological Institute, 3801 University Street, Montreal, QC, H3A 2B4, Canada. .,Douglas Hospital, McGill University, 6875 La Salle Blvd-FBC room 3149, Montreal, QC, H4H 1R3, Canada.
| |
Collapse
|
158
|
Okamura N, Harada R, Ishiki A, Kikuchi A, Nakamura T, Kudo Y. The development and validation of tau PET tracers: current status and future directions. Clin Transl Imaging 2018; 6:305-316. [PMID: 30148121 PMCID: PMC6096533 DOI: 10.1007/s40336-018-0290-y] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/03/2018] [Indexed: 01/02/2023]
Abstract
Purpose To provide an overview on positron emission tomography (PET) imaging of tau pathology in Alzheimer’s disease (AD) and other neurodegenerative disorders. Results Different classes of tau tracers such as flortaucipir, THK5317, and PBB3 have been developed and utilized in previous clinical studies. In AD, the topographical distribution of tracer binding follows the known distribution of neurofibrillary tangles and is closely associated with neurodegeneration as well as the clinical phenotype of dementia. Significant retention of tracers has also been observed in the frequent site of the 4-repeat (4R) tau isoform deposits in non-AD tauopathies, such as in progressive supranuclear palsy. However, in vitro binding studies indicate that most tau tracers are less sensitive to straight tau filaments, in contrast to their high binding affinity to paired helical filaments of tau (PHF-tau). The first-generation of tau tracers shows off-target binding in the basal ganglia, midbrain, thalamus, choroid plexus, and venous sinus. Off-target binding of THK5351 to monoamine oxidase B (MAO-B) has been observed in disease-associated brain regions linked to neurodegeneration and is associated with astrogliosis in areas of misfolded protein accumulation. The second generation of tau tracers, such as [18F]MK-6240, is highly selective to PHF-tau with little off-target binding and have enabled the reliable assessment of PHF-tau burden in aging and AD. Conclusions Tau PET tracers have enabled in vivo quantification of PHF-tau burden in human brains. Tau PET can help in understanding the underlying cause of dementia symptoms, and in patient selection for clinical trials of anti-dementia therapies.
Collapse
Affiliation(s)
- Nobuyuki Okamura
- 1Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan.,3Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Ryuichi Harada
- 2Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| | - Aiko Ishiki
- 3Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Akio Kikuchi
- 4Department of Neurology, Tohoku University School of Medicine, Sendai, Japan
| | - Tadaho Nakamura
- 1Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yukitsuka Kudo
- 3Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
159
|
Murugan NA, Nordberg A, Ågren H. Different Positron Emission Tomography Tau Tracers Bind to Multiple Binding Sites on the Tau Fibril: Insight from Computational Modeling. ACS Chem Neurosci 2018; 9:1757-1767. [PMID: 29630333 DOI: 10.1021/acschemneuro.8b00093] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Using the recently reported cryo-EM structure for the tau fibril [ Fitzpatrick et al. (2017) Nature 547, 185-190 ], which is a potential target concerning Alzheimer's disease, we present the first molecular modeling studies on its interaction with various positron emission tomography (PET) tracers. Experimentally, based on the binding assay studies, at least three different high-affinity binding sites have been reported for tracers in the tau fibril. Herein, through integrated modeling using molecular docking, molecular dynamics, and binding free energy calculations, we provide insight into the binding patterns of various tracers to the tau fibril. We suggest that there are four different high-affinity binding sites available for many of the studied tracers showing varying binding affinity to different binding sites. Thus, PBB3 binds most strongly to site 4, and interestingly, this site is not a preferable site for any other tracers. For THK5351, our data show that it strongly binds to sites 3 and 1, the former one being more preferable. We also find that MK6240 and T807 bind to site 1 specifically. The modeling data also give some insight into whether a tracer bound to a specific site can be replaced by others or not. For example, the displacement of T807 by PBB3 as reported experimentally can also be explained and attributed to the larger binding affinity of the latter compound in all binding sites. The binding free energy results explain very well the small binding affinity of THK523 compared to all the aryl quinoline moieties containing THK tracers. The ability of certain tau tracers, like FDDNP and THK523, to bind to amyloid fibrils has also been investigated. Furthermore, such off-target interaction of tau tracers with amyloid beta fibrils has been validated using a quantum mechanical fragmentation approach.
Collapse
Affiliation(s)
- N. Arul Murugan
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
| | - Agneta Nordberg
- Department of Neurobiology, Care Sciences and Society, Center of Alzheimer Research, Division of Clinical Geriatric, Karolinska Institutet, Huddinge, S-141 86 Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, S-141 86 Stockholm, Sweden
| | - Hans Ågren
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center, S-106 91 Stockholm, Sweden
- Department of Physics and Astronomy, Uppsala University, SE-751 20 Uppsala, Sweden
| |
Collapse
|
160
|
Ali F, Josephs K. The diagnosis of progressive supranuclear palsy: current opinions and challenges. Expert Rev Neurother 2018; 18:603-616. [DOI: 10.1080/14737175.2018.1489241] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Farwa Ali
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Keith Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
161
|
Ryan J, Fransquet P, Wrigglesworth J, Lacaze P. Phenotypic Heterogeneity in Dementia: A Challenge for Epidemiology and Biomarker Studies. Front Public Health 2018; 6:181. [PMID: 29971228 PMCID: PMC6018385 DOI: 10.3389/fpubh.2018.00181] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022] Open
Abstract
Dementia can result from a number of distinct diseases with differing etiology and pathophysiology. Even within the same disease, there is considerable phenotypic heterogeneity with varying symptoms and disease trajectories. Dementia diagnosis is thus very complex, time-consuming, and expensive and can only be made definitively post-mortem with histopathological confirmation. These inherent difficulties combined with the overlap of some symptoms and even neuropathological features, present a challenging problem for research in the field. This has likely hampered progress in epidemiological studies of risk factors and preventative interventions, as well as genetic and biomarker research. Resource limitations in large epidemiologically studies mean that limited diagnostic criteria are often used, which can result in phenotypically heterogeneous disease states being grouped together, potentially resulting in misclassification bias. When biomarkers are identified for etiologically heterogeneous diseases, they will have low specificity for any utility in clinical practice, even if their sensitivity is high. We highlight several challenges in in the field which must be addressed for the success of future genetic and biomarker studies, and may be key to the development of the most effective treatments. As a step toward achieving this goal, defining the dementia as a biological construct based on the presence of specific pathological features, rather than clinical symptoms, will enable more precise predictive models. It has the potential to lead to the discovery of novel genetic variants, as well as the identification of individuals at heightened risk of the disease, even prior to the appearance of clinical symptoms.
Collapse
Affiliation(s)
- Joanne Ryan
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Peter Fransquet
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Jo Wrigglesworth
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Paul Lacaze
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
162
|
Lohith TG, Bennacef I, Vandenberghe R, Vandenbulcke M, Salinas CA, Declercq R, Reynders T, Telan-Choing NF, Riffel K, Celen S, Serdons K, Bormans G, Tsai K, Walji A, Hostetler ED, Evelhoch JL, Van Laere K, Forman M, Stoch A, Sur C, Struyk A. Brain Imaging of Alzheimer Dementia Patients and Elderly Controls with 18F-MK-6240, a PET Tracer Targeting Neurofibrillary Tangles. J Nucl Med 2018; 60:107-114. [PMID: 29880509 DOI: 10.2967/jnumed.118.208215] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/14/2018] [Indexed: 11/16/2022] Open
Abstract
18F-MK-6240 (18F-labeled 6-(fluoro)-3-(1H-pyrrolo[2,3-c]pyridin-1-yl)isoquinolin-5-amine) is a highly selective, subnanomolar-affinity PET tracer for imaging neurofibrillary tangles (NFTs). Plasma kinetics, brain uptake, and preliminary quantitative analysis of 18F-MK-6240 in healthy elderly (HE) subjects, subjects with clinically probable Alzheimer disease (AD), and subjects with amnestic mild cognitive impairment were characterized in a study that is, to our knowledge, the first to be performed on humans. Methods: Dynamic PET scans of up to 150 min were performed on 4 cognitively normal HE subjects, 4 AD subjects, and 2 amnestic mild cognitive impairment subjects after a bolus injection of 152-169 MBq of 18F-MK-6240 to evaluate tracer kinetics and distribution in brain. Regional SUV ratio (SUVR) and distribution volume ratio were determined using the cerebellar cortex as a reference region. Total distribution volume was assessed by compartmental modeling using radiometabolite-corrected input function in a subgroup of 6 subjects. Results: 18F-MK-6240 had rapid brain uptake with a peak SUV of 3-5, followed by a uniformly quick washout from all brain regions in HE subjects; slower clearance was observed in regions commonly associated with NFT deposition in AD subjects. In AD subjects, SUVR between 60 and 90 min after injection was high (approximately 2-4) in regions associated with NFT deposition, whereas in HE subjects, SUVR was approximately 1 across all brain regions, suggesting high tracer selectivity for binding NFTs in vivo. 18F-MK-6240 total distribution volume was approximately 2- to 3-fold higher in neocortical and medial temporal brain regions of AD subjects than in HE subjects and stabilized by 60 min in both groups. Distribution volume ratio estimated by the Logan reference tissue model or compartmental modeling correlated well (R 2 > 0.9) to SUVR from 60 to 90 min for AD subjects. Conclusion: 18F-MK-6240 exhibited favorable kinetics and high binding levels to brain regions with a plausible pattern for NFT deposition in AD subjects. In comparison, negligible tracer binding was observed in HE subjects. This pilot study suggests that simplified ratio methods such as SUVR can be used to quantify NFT binding. These results support further clinical development of 18F-MK-6240 for potential application in longitudinal studies.
Collapse
Affiliation(s)
- Talakad G Lohith
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | - Idriss Bennacef
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | - Rik Vandenberghe
- Neurology Department, University Hospitals Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium; Alzheimer Research Centre, KU Leuven, Leuven Institute for Neuroscience and Disease, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Neurology Department, University Hospitals Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium; Alzheimer Research Centre, KU Leuven, Leuven Institute for Neuroscience and Disease, Leuven, Belgium
| | | | - Ruben Declercq
- Translational Pharmacology Europe, Merck Sharp & Dohme Corp., Brussels, Belgium
| | - Tom Reynders
- Translational Pharmacology Europe, Merck Sharp & Dohme Corp., Brussels, Belgium
| | | | - Kerry Riffel
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | - Sofie Celen
- Radiopharmaceutical Research, KU Leuven, Leuven, Belgium
| | - Kim Serdons
- Division of Nuclear Medicine, University Hospitals Leuven and UZ Leuven, Leuven, Belgium
| | - Guy Bormans
- Radiopharmaceutical Research, KU Leuven, Leuven, Belgium
| | - Kuenhi Tsai
- Early Clinical Statistics, Merck & Co., Inc., West Point, Pennsylvania
| | - Abbas Walji
- Discovery Chemistry, Merck & Co., Inc., West Point, Pennsylvania; and
| | - Eric D Hostetler
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | | | - Koen Van Laere
- Division of Nuclear Medicine, University Hospitals Leuven and UZ Leuven, Leuven, Belgium
| | - Mark Forman
- Translational Pharmacology, Merck & Co., Inc., North Wales, Pennsylvania
| | - Aubrey Stoch
- Translational Pharmacology, Merck & Co., Inc., North Wales, Pennsylvania
| | - Cyrille Sur
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | - Arie Struyk
- Translational Pharmacology, Merck & Co., Inc., North Wales, Pennsylvania
| |
Collapse
|
163
|
Betthauser TJ, Cody KA, Zammit MD, Murali D, Converse AK, Barnhart TE, Stone CK, Rowley HA, Johnson SC, Christian BT. In Vivo Characterization and Quantification of Neurofibrillary Tau PET Radioligand 18F-MK-6240 in Humans from Alzheimer Disease Dementia to Young Controls. J Nucl Med 2018; 60:93-99. [PMID: 29777006 DOI: 10.2967/jnumed.118.209650] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/14/2018] [Indexed: 11/16/2022] Open
Abstract
Tau PET imaging has potential for elucidating changes in the deposition of neuropathological tau aggregates that are occurring during the progression of Alzheimer disease (AD). This work investigates in vivo kinetics, quantification strategies, and imaging characteristics of a novel tau PET radioligand 18F-MK-6240 in humans. Methods: Fifty-one individuals ranging from cognitively normal young controls to persons with dementia underwent T1-weighted MRI as well as 11C-PiB and 18F-MK-6240 PET imaging. PET data were coregistered to the MRI, and time-activity curves were extracted from regions of interest to assess 18F-MK-6240 kinetics. The pons and inferior cerebellum were investigated as potential reference regions. Reference tissue methods (Logan graphical analysis [LGA] and multilinear reference tissue method [MRTM2]) were investigated for quantification of 18F-MK-6240 distribution volume ratios (DVRs) in a subset of 19 participants. Stability of DVR methods was evaluated using truncated scan durations. SUV ratio (SUVR) estimates were compared with DVR estimates to determine the optimal timing window for SUVR analysis. Parametric SUVR images were used to identify regions of potential off-target binding and to compare binding patterns with neurofibrillary tau staging established in neuropathology literature. Results: SUVs in the pons and the inferior cerebellum indicated consistent clearance across all 51 subjects. LGA and MRTM2 DVR estimates were similar, with LGA slightly underestimating DVR compared with MRTM2. DVR estimates remained stable when truncating the scan duration to 60 min. SUVR determined 70-90 min after injection of 18F-MK-6240 indicated linearity near unity when compared with DVR estimates and minimized potential spill-in from uptake outside the brain. 18F-MK-6240 binding patterns in target regions were consistent with neuropathological neurofibrillary tau staging. Off-target binding regions included the ethmoid sinus, clivus, meninges, substantia nigra, but not the basal ganglia or choroid plexus. Conclusion: 18F-MK-6240 is a promising PET radioligand for in vivo imaging of neurofibrillary tau aggregates in AD with minimal off-target binding in the human brain.
Collapse
Affiliation(s)
- Tobey J Betthauser
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin .,Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Karly A Cody
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Matthew D Zammit
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin.,Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Dhanabalan Murali
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Alexander K Converse
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin.,Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Charles K Stone
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Howard A Rowley
- Department of Radiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Sterling C Johnson
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin; and.,Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Bradley T Christian
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin.,Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
164
|
Wong DF, Comley RA, Kuwabara H, Rosenberg PB, Resnick SM, Ostrowitzki S, Vozzi C, Boess F, Oh E, Lyketsos CG, Honer M, Gobbi L, Klein G, George N, Gapasin L, Kitzmiller K, Roberts J, Sevigny J, Nandi A, Brasic J, Mishra C, Thambisetty M, Mogekar A, Mathur A, Albert M, Dannals RF, Borroni E. Characterization of 3 Novel Tau Radiopharmaceuticals, 11C-RO-963, 11C-RO-643, and 18F-RO-948, in Healthy Controls and in Alzheimer Subjects. J Nucl Med 2018; 59:1869-1876. [PMID: 29728519 DOI: 10.2967/jnumed.118.209916] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/27/2018] [Indexed: 01/01/2023] Open
Abstract
11C-RO-963, 11C-RO-643, and 18F-RO-948 (previously referred to as 11C-RO6924963, 11C-RO6931643, and 18F-RO6958948, respectively) have been reported as promising PET tracers for tau imaging based on in vitro and preclinical PET data. Here we describe the first, to our knowledge, human evaluation of these novel radiotracers. Methods: Amyloid PET-positive Alzheimer disease (AD) subjects and younger controls each received 2 different tau tracers. Dynamic 90-min scans were obtained after bolus injection of 11C-RO-963, 11C-RO-643, or 18F-RO-948. Arterial blood sampling was performed on 11 healthy controls and 11 AD subjects. Regions were defined on MR images, and PET data were quantified by plasma reference graphical analysis (for total distribution volume) and target cerebellum ratio (SUV ratios of 60- to 90-min frames). SUV ratio images were also analyzed voxelwise. Five older controls each underwent 2 scans with 18F-RO-948 for evaluation of test-retest variability. Four AD subjects underwent a repeated 18F-RO-948 scan 6-22 mo after the first scan. Six additional healthy controls (3 men and 3 women; age range, 41-67 y) each underwent 1 whole-body dosimetry scan with 18F-RO-948. Results: In younger controls, SUVpeak was observed in the temporal lobe with values of approximately 3.0 for 11C-RO-963, 1.5 for 11C-RO-643, and 3.5 for 18F-RO-948. Over all brain regions and subjects, the trend was for 18F-RO-948 to have the highest SUVpeak, followed by 11C-RO-963 and then 11C-RO-643. Regional analysis of SUV ratio and total distribution volume for 11C-RO-643 and 18F-RO-948 clearly discriminated the AD group from the healthy control groups. Compartmental modeling confirmed that 11C-RO-643 had lower brain entry than either 11C-RO-963 or 18F-RO-948 and that 18F-RO-948 showed better contrast between (predicted) areas of high versus low tau accumulation. Thus, our subsequent analysis focused on 18F-RO-948. Both voxelwise and region-based analysis of 18F-RO-948 binding in healthy controls versus AD subjects revealed multiple areas where AD subjects significantly differed from healthy controls. Of 22 high-binding regions, 13 showed a significant group difference (after ANOVA, F (1,21) = 45, P < 10-5). Voxelwise analysis also revealed a set of symmetric clusters where AD subjects had higher binding than healthy controls (threshold of P < 0.001, cluster size > 50). Conclusion: 18F-RO-948 demonstrates characteristics superior to 11C-RO-643 and 11C-RO-963 for characterization of tau pathology in AD. Regional binding data and kinetic properties of 18F-RO-948 compare favorably with other existing tau PET tracers.
Collapse
Affiliation(s)
- Dean F Wong
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland .,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland.,Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland.,Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - Robert A Comley
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Hiroto Kuwabara
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Susan M Resnick
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland; and
| | - Susanne Ostrowitzki
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Cristina Vozzi
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Frank Boess
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Esther Oh
- Lab of Behavior and Neuroscience, NIH-NIA IRP, Baltimore, Maryland
| | - Constantine G Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Michael Honer
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Luca Gobbi
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Gregory Klein
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Noble George
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Lorena Gapasin
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kelly Kitzmiller
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Josh Roberts
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Jeff Sevigny
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| | - Ayon Nandi
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - James Brasic
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Chakradhar Mishra
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | - Abhay Mogekar
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - Anil Mathur
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Marilyn Albert
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland.,Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - Robert F Dannals
- Section of High Resolution Brain PET, Department of Radiology and Radiological Sciences and Department of Nuclear Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Edilio Borroni
- Pharma Research and Early Development, Hoffmann-La Roche, Basel, Switzerland
| |
Collapse
|
165
|
Large inter- and intra-case variability of first generation tau PET ligand binding in neurodegenerative dementias. Acta Neuropathol Commun 2018; 6:34. [PMID: 29716656 PMCID: PMC5928586 DOI: 10.1186/s40478-018-0535-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 12/03/2022] Open
Abstract
Imaging of pathological tau with positron emission tomography (PET) has the potential to allow early diagnosis of the dementias and monitoring of disease progression, including assessment of therapeutic interventions, in vivo. The first generation of tau PET tracers, including the carbazole flortaucipir and the 2-arylquinolines of the THK series, are now used in clinical research; however, concerns have been raised about off-target binding and low sensitivity. With the aim to determine the nature of tau pathology depicted by structurally distinct tau ligands we carried out a microscopic neuropathological evaluation in post-mortem human brain tissue of cases with primary and secondary tauopathies. Carbazole and 2-arylquinoline binding was only observed in cases with Alzheimer’s disease and one case with frontotemporal dementia and parkinsonism linked to chromosome 17 exhibiting a R406W MAPT mutation. In end stage Alzheimer’s disease cases, fluorescent imaging with the carbazole T726 and the 2-arylquinoline THK-5117 revealed high inter- and intra-case variability of tracer binding, and this was corroborated by quantitative phosphorimaging with the PET tracer [18F]THK-5117. Microscopic analysis of the pathological inclusions revealed that the fluorescent tracers preferentially bind to premature tau aggregates. Whilst T726 binding was limited to neuronal tau, THK-5117 additionally depicted neuritic tau. Neither tracer depicted tau in pre-symptomatic disease. Our results highlight limitations of the first generation of tau PET tracers, in particular lack of correlation between pathological tau load and tracer binding, limited sensitivity to tau in early disease, and high variability in tracer binding between and within cases. Concerns remain that these limitations may also affect the next generation tracers as they target the same high affinity binding site. Therefore, it is crucial to assess inter- and intra-subject correlation of tracer binding with pathological tau load in post-mortem tissue studies, and to rigorously assess novel tau PET tracers before translation into clinical studies.
Collapse
|
166
|
Lemoine L, Leuzy A, Chiotis K, Rodriguez-Vieitez E, Nordberg A. Tau positron emission tomography imaging in tauopathies: The added hurdle of off-target binding. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2018; 10:232-236. [PMID: 29780868 PMCID: PMC5956931 DOI: 10.1016/j.dadm.2018.01.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ligands targeting tau for use with positron emission tomography have rapidly been developed during the past several years, enabling the in vivo study of tau pathology in patients with Alzheimer's disease and related non-Alzheimer's disease tauopathies. Several candidate compounds have been developed, showing good in vitro characteristics with respect to their ability to bind tau deposits; off-target binding, however, has also been observed. In this short commentary, we briefly summarize the available in vivo and in vitro evidence pertaining to their off-target binding and discuss the different approaches that are needed for the future development of tau positron emission tomography tracers.
Collapse
Affiliation(s)
- Laetitia Lemoine
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- Corresponding author. Tel.: +46 8 524 83 527; Fax: +46 8 585 85470 .
| | - Antoine Leuzy
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Konstantinos Chiotis
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Elena Rodriguez-Vieitez
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
167
|
Patel S, Schmidt K, Hesterman J, Hoppin J. Advancing Drug Discovery and Development Using Molecular Imaging (ADDMI): an Interest Group of the World Molecular Imaging Society and an Inaugural Session on Positron Emission Tomography (PET). Mol Imaging Biol 2018; 19:348-356. [PMID: 28417265 DOI: 10.1007/s11307-017-1085-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multi-modality molecular imaging techniques have expanded the role of imaging biomarkers in the pharmaceutical industry and are beginning to streamline the drug discovery and development process. The World Molecular Imaging Society (WMIS) serves as a forum for discussing innovative and exploratory multi-modal, interdisciplinary molecular imaging research with a mission of bridging the gap between pathology and in vivo imaging. To formalize the role of the WMIS in pharmaceutical research efforts, members of the society have formed an interest group entitled Advancing Drug Discovery and Development using Molecular Imaging (ADDMI). The ADDMI interest group launched their efforts at the 2016 World Molecular Imaging Congress by hosting a session of invited lectures on translational positron emission tomography (PET) imaging in the central nervous system. This article provides a synopsis of those lectures and frames the role of translational imaging biomarker strategies in the drug discovery and development process.
Collapse
Affiliation(s)
- Shil Patel
- Eisai AiM Institute, 4 Corporate Drive, Andover, MA, USA.
| | | | | | | |
Collapse
|
168
|
Goedert M, Yamaguchi Y, Mishra SK, Higuchi M, Sahara N. Tau Filaments and the Development of Positron Emission Tomography Tracers. Front Neurol 2018; 9:70. [PMID: 29497399 PMCID: PMC5818396 DOI: 10.3389/fneur.2018.00070] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/30/2018] [Indexed: 12/15/2022] Open
Abstract
A pathological pathway leading from soluble, monomeric to insoluble, filamentous Tau, is believed to underlie human Tauopathies. Cases of frontotemporal dementia are caused by dominantly inherited mutations in MAPT, the Tau gene. They show that dysfunction of Tau protein is sufficient to cause neurodegeneration and dementia. Extrapolation to the more common sporadic Tauopathies leads one to conclude that the pathological pathway is central to the development of all cases of disease, even if there are multiple reasons for Tau assembly. These findings are conceptually similar to those reported for beta-amyloid, alpha-synuclein and prion protein. Here, we provide an overview of Tau filaments and their positron emission tomography ligands.
Collapse
Affiliation(s)
- Michel Goedert
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | | | - Makoto Higuchi
- National Institute of Radiological Sciences, Chiba, Japan
| | | |
Collapse
|
169
|
Lyoo CH, Cho H, Choi JY, Ryu YH, Lee MS. Tau Positron Emission Tomography Imaging in Degenerative Parkinsonisms. J Mov Disord 2018; 11:1-12. [PMID: 29381890 PMCID: PMC5790630 DOI: 10.14802/jmd.17071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 11/20/2017] [Indexed: 01/08/2023] Open
Abstract
In recent years, several radiotracers that selectively bind to pathological tau proteins have been developed. Evidence is emerging that binding patterns of in vivo tau positron emission tomography (PET) studies in Alzheimer’s disease (AD) patients closely resemble the distribution patterns of known neurofibrillary tangle pathology, with the extent of tracer binding reflecting the clinical and pathological progression of AD. In Lewy body diseases (LBD), tau PET imaging has clearly revealed cortical tau burden with a distribution pattern distinct from AD and increased cortical binding within the LBD spectrum. In progressive supranuclear palsy, the globus pallidus and midbrain have shown increased binding most prominently. Tau PET patterns in patients with corticobasal syndrome are characterized by asymmetrical uptake in the motor cortex and underlying white matter, as well as in the basal ganglia. Even in the patients with multiple system atrophy, which is basically a synucleinopathy, 18F-flortaucipir, a widely used tau PET tracer, also binds to the atrophic posterior putamen, possibly due to off-target binding. These distinct patterns of tau-selective radiotracer binding in the various degenerative parkinsonisms suggest its utility as a potential imaging biomarker for the differential diagnosis of parkinsonisms.
Collapse
Affiliation(s)
- Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Yong Choi
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Division of RI-Convergence Research, Korea Institute Radiological and Medical Sciences, Seoul, Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Myung Sik Lee
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
170
|
Vermeiren C, Motte P, Viot D, Mairet-Coello G, Courade JP, Citron M, Mercier J, Hannestad J, Gillard M. The tau positron-emission tomography tracer AV-1451 binds with similar affinities to tau fibrils and monoamine oxidases. Mov Disord 2017; 33:273-281. [DOI: 10.1002/mds.27271] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/01/2017] [Accepted: 11/22/2017] [Indexed: 12/25/2022] Open
Affiliation(s)
| | - Philippe Motte
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| | - Delphine Viot
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| | | | | | - Martin Citron
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| | - Joël Mercier
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| | - Jonas Hannestad
- Denali Therapeutics Inc.; South San Francisco California USA
| | - Michel Gillard
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| |
Collapse
|
171
|
Lemoine L, Gillberg PG, Svedberg M, Stepanov V, Jia Z, Huang J, Nag S, Tian H, Ghetti B, Okamura N, Higuchi M, Halldin C, Nordberg A. Comparative binding properties of the tau PET tracers THK5117, THK5351, PBB3, and T807 in postmortem Alzheimer brains. ALZHEIMERS RESEARCH & THERAPY 2017; 9:96. [PMID: 29229003 PMCID: PMC5725799 DOI: 10.1186/s13195-017-0325-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/22/2017] [Indexed: 11/10/2022]
Abstract
Background The aim of this study was to compare the binding properties of several tau positron emission tomography tracers—THK5117, THK5351, T807 (also known as AV1451; flortaucipir), and PBB3—head to head in the same human brain tissue. Methods Binding assays were performed to compare the regional distribution of 3H-THK5117 and 3H-THK5351 in postmortem tissue from three Alzheimer’s disease (AD) cases and three control subjects in frontal and temporal cortices as well as in the hippocampus. Competition binding assays between THK5351, THK5117, PBB3, and T807, as well as off-target binding of THK5117 and T807 toward monoamine oxidase B (MAO-B), were performed using binding assays in brain homogenates and autoradiography of three AD cases. Results Regional binding of 3H-THK5117 and 3H-THK5351 was similar, except in the temporal cortex, which showed higher 3H-THK5117 binding. Saturation studies demonstrated two binding sites for 3H-THK5351 (Kd1 = 5.6 nM, Bmax = 76 pmol/g; Kd2 = 1 nM, Bmax = 40 pmol/g). Competition studies in the hippocampus between 3H-THK5351 and unlabeled THK5351, THK5117, and T807 revealed super-high-affinity sites for all three tracers (THK5351 Ki = 0.1 pM; THK5117 Ki = 0.3 pM; T807 Ki = 0.2 pM) and an additional high-affinity site (THK5351 Ki = 16 nM; THK5117 Ki = 20 nM; T807 Ki = 78nM). 18F-T807, 11C-THK5351, and 11C-PBB3 autoradiography of large frozen sections from three AD brains showed similar regional binding for the three tracers, with lower binding intensity for 11C-PBB3. Unlabeled THK5351 and T807 displaced 11C-THK5351 to a similar extent and a lower extent, respectively, compared with 11C-PBB3. Competition with the MAO-B inhibitor 3H-l-deprenyl was observed for THK5117 and T807 in the hippocampus (THK5117 Ki = 286 nM; T807 Ki = 227 nM) and the putamen (THK5117 Ki = 148 nM; T807 Ki = 135 nM). 3H-THK5351 binding was displaced using autoradiography competition with unlabeled THK5351 and T807 in cortical areas by 70–80% and 60–77%, respectively, in the basal ganglia, whereas unlabeled deprenyl displaced 3H-THK5351 binding by 40% in the frontal cortex and 50% in the basal ganglia. Conclusions THK5351, THK5117, and T807 seem to target similar binding sites, but with different affinities, whereas PBB3 seems to target its own binding site. Both THK5117 and T807 demonstrated off-target binding in the hippocampus and putamen with a ten times lower binding affinity to the MAO-B inhibitor deprenyl compared with 3H-THK5351. Electronic supplementary material The online version of this article (doi:10.1186/s13195-017-0325-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laetitia Lemoine
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Per-Göran Gillberg
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Marie Svedberg
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Vladimir Stepanov
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Zhisheng Jia
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Jinghai Huang
- Institute of Fine Chemicals, East China University of Science and Technology, Shanghai, China
| | - Sangram Nag
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - He Tian
- Institute of Fine Chemicals, East China University of Science and Technology, Shanghai, China
| | - Bernardino Ghetti
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Tohoku, Japan
| | - Makoto Higuchi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden. .,Department of Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
172
|
Jang YK, Lyoo CH, Park S, Oh SJ, Cho H, Oh M, Ryu YH, Choi JY, Rabinovici GD, Kim HJ, Moon SH, Jang H, Lee JS, Jagust WJ, Na DL, Kim JS, Seo SW. Head to head comparison of [ 18F] AV-1451 and [ 18F] THK5351 for tau imaging in Alzheimer's disease and frontotemporal dementia. Eur J Nucl Med Mol Imaging 2017; 45:432-442. [PMID: 29143870 DOI: 10.1007/s00259-017-3876-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/03/2017] [Indexed: 11/25/2022]
Abstract
PURPOSE Tau accumulation is a core pathologic change in various neurodegenerative diseases including Alzheimer's disease and frontotemporal lobar degeneration-tau. Recently, tau positron emission tomography tracers such as [18F] AV-1451 and [18F] THK5351 have been developed to detect tau deposition in vivo. In the present study, we performed a head to head comparison of these two tracers in Alzheimer's disease and frontotemporal dementia cases and aimed to investigate which tracers are better suited to image tau in these disorders. METHODS A cross-sectional study was conducted using a hospital-based sample at a tertiary referral center. We recruited eight participants (two Alzheimer's disease, four frontotemporal dementia and two normal controls) who underwent magnetic resonance image, amyloid positron emission tomography with [18F]-Florbetaben and tau positron emission tomography with both THK5351 and AV-1451. To measure regional AV1451 and THK5351 uptakes, we used the standardized uptake value ratios by dividing mean activity in target volume of interest by mean activity in the cerebellar hemispheric gray matter. RESULTS Although THK5351 and AV-1451 uptakes were highly correlated, cortical uptake of AV-1451 was more striking in Alzheimer's disease, while cortical uptake of THK5351 was more prominent in frontotemporal dementia. THK5351 showed higher off-target binding than AV-1451 in the white matter, midbrain, thalamus, and basal ganglia. CONCLUSIONS AV-1451 is more sensitive and specific to Alzheimer's disease type tau and shows lower off-target binding, while THK5351 may mirror non-specific neurodegeneration.
Collapse
Affiliation(s)
- Young Kyoung Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Kangnam-ku, Seoul, 06351, South Korea
- Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Seongbeom Park
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Kangnam-ku, Seoul, 06351, South Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-736, South Korea
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-736, South Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Yong Choi
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Gil D Rabinovici
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Kangnam-ku, Seoul, 06351, South Korea
- Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Kangnam-ku, Seoul, 06351, South Korea
- Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Jin San Lee
- Department of Neurology, Kyung Hee University Hospital, Seoul, South Korea
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
- Center of Functional Imaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Kangnam-ku, Seoul, 06351, South Korea
- Neuroscience Center, Samsung Medical Center, Seoul, South Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-736, South Korea.
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Kangnam-ku, Seoul, 06351, South Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, South Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
173
|
Andrés JI, Schmidt M. Medicinal Chemistry strategies for PET tracer discovery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2017; 25:11-17. [PMID: 29233262 DOI: 10.1016/j.ddtec.2017.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/29/2017] [Accepted: 10/10/2017] [Indexed: 06/07/2023]
Abstract
The detection of gamma rays, resulting from decay of positron emitting isotopes, allows exquisitely sensitive detection of probes radiolabeled with such isotopes. These probes can be designed for high affinity binding to specific molecular targets and be used as tools in the early development of drugs, particularly for neuropsychiatric disorders. Availability of novel tracers requires dedicated resources and selection assays. Many of the selection assays are similar to those used for discovery of clinical compounds, although the distribution and clearance of target specific radioligands requires different in vitro and in vivo methods and new derivatives.
Collapse
Affiliation(s)
- José Ignacio Andrés
- Discovery Sciences, Janssen Research & Development, Janssen-Cilag S. A., C/Jarama 75A, 45007 Toledo, Spain.
| | - Mark Schmidt
- Neuroscience Therapeutic Area, Janssen Research & Development, Division of Janssen Pharmaceutica, NV, Turnhoutseweg 30, Beerse 2340, Belgium
| |
Collapse
|
174
|
Bernard-Gauthier V, Collier TL, Liang SH, Vasdev N. Discovery of PET radiopharmaceuticals at the academia-industry interface. DRUG DISCOVERY TODAY. TECHNOLOGIES 2017; 25:19-26. [PMID: 29233263 DOI: 10.1016/j.ddtec.2017.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 09/18/2017] [Indexed: 01/24/2023]
Abstract
Project-specific collaborations between academia and pharmaceutical partners are a growing phenomenon within molecular imaging and in particular in the positron emission tomography (PET) radiopharmaceutical community. This cultural shift can be attributed in part to decreased public funding in academia in conjunction with the increased reliance on outsourcing of chemistry, radiochemistry, pharmacology and molecular imaging studies by the pharmaceutical industry. This account highlights some of our personal experiences working with industrial partners to develop new PET radiochemistry methodologies for drug discovery and neuro-PET research studies. These symbiotic academic-industrial partnerships have not only led to novel radiotracers for new targets but also to the application of new carbon-11 and fluorine-18 labeling methodologies and technologies to label previously unprecedented compounds for in vivo evaluations.
Collapse
Affiliation(s)
- Vadim Bernard-Gauthier
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Thomas L Collier
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA; Advion Inc., Research and Development, Ithaca, NY 14850, USA
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
175
|
Abstract
A compelling need in the field of neurodegenerative diseases is the development and validation of biomarkers for early identification and differential diagnosis. The availability of positron emission tomography (PET) neuroimaging tools for the assessment of molecular biology and neuropathology has opened new venues in the diagnostic design and the conduction of new clinical trials. PET techniques, allowing the in vivo assessment of brain function and pathology changes, are increasingly showing great potential in supporting clinical diagnosis also in the early and even preclinical phases of dementia. This review will summarize the most recent evidence on fluorine-18 fluorodeoxyglucose-, amyloid -, tau -, and neuroinflammation - PET tools, highlighting strengths and limitations and possible new perspectives in research and clinical applications. Appropriate use of PET tools is crucial for a prompt diagnosis and target evaluation of new developed drugs aimed at slowing or preventing dementia.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Arianna Sala
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Paola Caminiti
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Nuclear Medicine Unit, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
176
|
Honer M, Gobbi L, Knust H, Kuwabara H, Muri D, Koerner M, Valentine H, Dannals RF, Wong DF, Borroni E. Preclinical Evaluation of 18F-RO6958948, 11C-RO6931643, and 11C-RO6924963 as Novel PET Radiotracers for Imaging Tau Aggregates in Alzheimer Disease. J Nucl Med 2017; 59:675-681. [PMID: 28970331 DOI: 10.2967/jnumed.117.196741] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/25/2017] [Indexed: 01/08/2023] Open
Abstract
Tau aggregates and amyloid-β (Aβ) plaques are key histopathologic features in Alzheimer disease (AD) and are considered targets for therapeutic intervention as well as biomarkers for diagnostic in vivo imaging agents. This article describes the preclinical in vitro and in vivo characterization of 3 novel compounds-RO6958948, RO6931643, and RO6924963-that bind specifically to tau aggregates and have the potential to become PET tracers for future human use. Methods: RO6958948, RO6931643, and RO6924963 were identified as high-affinity competitors at the 3H-T808 binding site on native tau aggregates in human late-stage AD brain tissue. Binding of tritiated compounds to brain tissue sections of AD patients and healthy controls was analyzed by macro- and microautoradiography and by costaining of tau aggregates and Aβ plaques on the same tissue section using specific antibodies. All 3 tracer candidates were radiolabeled with a PET nuclide and tested in vivo in tau-naïve baboons to assess brain uptake, distribution, clearance, and metabolism. Results:3H-RO6958948, 3H-RO6931643, and 3H-RO6924963 bound with high affinity and specificity to tau aggregates, clearly lacking affinity for concomitant Aβ plaques in human AD Braak V tissue sections. The specificity of all 3 radioligands for tau aggregates was supported, first, by binding patterns in AD sections comparable to the tau-specific radioligand 3H-T808; second, by very low nonspecific binding in brain tissue devoid of tau pathology, excluding significant radioligand binding to any other central nervous system target; and third, by macroscopic and microscopic colocalization and quantitative correlation of radioligand binding and tau antibody staining on the same tissue section. RO6958948, RO6931643, and RO6924963 were successfully radiolabeled with a PET nuclide at high specific activity, radiochemical purity, and yield. After intravenous administration of 18F-RO6958948, 11C-RO6931643, and 11C-RO6924963 to baboons, PET scans indicated good brain entry, rapid washout, and a favorable metabolism pattern. Conclusion:18F-RO6958948, 11C-RO6931643, and 11C-RO6924963 are promising PET tracers for visualization of tau aggregates in AD. Head-to-head comparison and validation of these tracer candidates in AD patients and healthy controls will be reported in due course.
Collapse
Affiliation(s)
- Michael Honer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Luca Gobbi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Henner Knust
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Hiroto Kuwabara
- PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Section of High Resolution Brain PET, PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dieter Muri
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Matthias Koerner
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Heather Valentine
- PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Section of High Resolution Brain PET, PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert F Dannals
- PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dean F Wong
- PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Section of High Resolution Brain PET, PET Center, Division of Nuclear Medicine, Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland; and.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edilio Borroni
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
177
|
Bischof GN, Endepols H, van Eimeren T, Drzezga A. Tau-imaging in neurodegeneration. Methods 2017; 130:114-123. [PMID: 28790016 DOI: 10.1016/j.ymeth.2017.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/25/2017] [Accepted: 08/04/2017] [Indexed: 10/19/2022] Open
Abstract
Pathological cerebral aggregations of proteins are suggested to play a crucial role in the development of neurodegenerative disorders. For example, aggregation of the protein ß-amyloid in form of extracellular amyloid-plaques as well as intraneuronal depositions of the protein tau in form of neurofibrillary tangles represent hallmarks of Alzheimer's disease (AD). Recently, novel tracers for in vivo molecular imaging of tau-aggregates in the brain have been introduced, complementing existing tracers for imaging amyloid-plaques. Available data on these novel tracers indicate that the subject of Tau-PET may be of considerable complexity. On the one hand this refers to the various forms of appearance of tau-pathology in different types of neurodegenerative disorders. On the other hand, a number of hurdles regarding validation of these tracers still need to be overcome with regard to comparability and standardization of the different tracers, observed off-target/non-specific binding and quantitative interpretation of the signal. These issues will have to be clarified before systematic clinical application of this exciting new methodological approach may become possible. Potential applications refer to early detection of neurodegeneration, differential diagnosis between tauopathies and non-tauopathies and specific patient selection and follow-up in therapy trials.
Collapse
Affiliation(s)
| | - Heike Endepols
- Department of Nuclear Medicine, University of Cologne, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University of Cologne, Germany; German Research Center for Neurodegenerative Diseases (DZNE), Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University of Cologne, Germany; German Research Center for Neurodegenerative Diseases (DZNE), Germany.
| |
Collapse
|
178
|
Gobbi LC, Knust H, Körner M, Honer M, Czech C, Belli S, Muri D, Edelmann MR, Hartung T, Erbsmehl I, Grall-Ulsemer S, Koblet A, Rueher M, Steiner S, Ravert HT, Mathews WB, Holt DP, Kuwabara H, Valentine H, Dannals RF, Wong DF, Borroni E. Identification of Three Novel Radiotracers for Imaging Aggregated Tau in Alzheimer’s Disease with Positron Emission Tomography. J Med Chem 2017; 60:7350-7370. [DOI: 10.1021/acs.jmedchem.7b00632] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Luca C. Gobbi
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Henner Knust
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Matthias Körner
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Michael Honer
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Christian Czech
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Sara Belli
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Dieter Muri
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Martin R. Edelmann
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Thomas Hartung
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Isabella Erbsmehl
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Sandra Grall-Ulsemer
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Andreas Koblet
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Marianne Rueher
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Sandra Steiner
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | | | | | | | | | | | | | | | - Edilio Borroni
- Pharma
Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| |
Collapse
|
179
|
Vāvere AL, Scott PJH. Clinical Applications of Small-molecule PET Radiotracers: Current Progress and Future Outlook. Semin Nucl Med 2017; 47:429-453. [PMID: 28826519 DOI: 10.1053/j.semnuclmed.2017.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Radiotracers, or radiopharmaceuticals, are bioactive molecules tagged with a radionuclide used for diagnostic imaging or radiotherapy and, when a positron-emitting radionuclide is chosen, the radiotracers are used for PET imaging. The development of novel PET radiotracers in many ways parallels the development of new pharmaceuticals, and small molecules dominate research and development pipelines in both disciplines. The 4 decades since the introduction of [18F]FDG have seen the development of many small molecule PET radiotracers. Ten have been approved by the US Food and Drug Administration as of 2016, whereas hundreds more are being evaluated clinically. These radiotracers are being used in personalized medicine and to support drug discovery programs where they are greatly improving our understanding of and ability to treat diseases across many areas of medicine including neuroscience, cardiovascular medicine, and oncology.
Collapse
Affiliation(s)
- Amy L Vāvere
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN
| | - Peter J H Scott
- Department of Radiology, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
180
|
Hall B, Mak E, Cervenka S, Aigbirhio FI, Rowe JB, O’Brien JT. In vivo tau PET imaging in dementia: Pathophysiology, radiotracer quantification, and a systematic review of clinical findings. Ageing Res Rev 2017; 36:50-63. [PMID: 28315409 DOI: 10.1016/j.arr.2017.03.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/15/2017] [Accepted: 03/06/2017] [Indexed: 12/14/2022]
Abstract
In addition to the deposition of β-amyloid plaques, neurofibrillary tangles composed of aggregated hyperphosphorylated tau are one of the pathological hallmarks of Alzheimer's disease and other neurodegenerative disorders. Until now, our understanding about the natural history and topography of tau deposition has only been based on post-mortem and cerebrospinal fluid studies, and evidence continues to implicate tau as a central driver of downstream neurodegenerative processes and cognitive decline. Recently, it has become possible to assess the regional distribution and severity of tau burden in vivo with the development of novel radiotracers for positron emission tomography (PET) imaging. In this article, we provide a comprehensive discussion of tau pathophysiology, its quantification with novel PET radiotracers, as well as a systematic review of tau PET imaging in normal aging and various dementia conditions: mild cognitive impairment, Alzheimer's disease, frontotemporal dementia, progressive supranuclear palsy, and Lewy body dementia. We discuss the main findings in relation to group differences, clinical-cognitive correlations of tau PET, and multi-modal relationships among tau PET and other pathological markers. Collectively, the small but growing literature of tau PET has yielded consistent anatomical patterns of tau accumulation that recapitulate post-mortem distribution of neurofibrillary tangles which correlate with cognitive functions and other markers of pathology. In general, AD is characterised by increased tracer retention in the inferior temporal lobe, extending into the frontal and parietal regions in more severe cases. It is also noted that the spatial topography of tau accumulation is markedly distinct to that of amyloid burden in aging and AD. Tau PET imaging has also revealed characteristic spatial patterns among various non-AD tauopathies, supporting its potential role for differential diagnosis. Finally, we propose novel directions for future tau research, including (a) longitudinal imaging in preclinical dementia, (b) multi-modal mapping of tau pathology onto other pathological processes such as neuroinflammation, and (c) the need for more validation studies against post-mortem samples of the same subjects.
Collapse
|
181
|
Xia C, Dickerson BC. Multimodal PET Imaging of Amyloid and Tau Pathology in Alzheimer Disease and Non-Alzheimer Disease Dementias. PET Clin 2017; 12:351-359. [PMID: 28576172 PMCID: PMC5690983 DOI: 10.1016/j.cpet.2017.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Biomarkers of the molecular pathology underpinning dementia syndromes are increasingly recognized as crucial for diagnosis and development of disease-modifying treatments. Amyloid PET imaging is an integral part of the diagnostic assessment of Alzheimer disease. Its use has also deepened understanding of the role of amyloid pathology in Lewy body disorders and aging. Tau PET imaging is an imaging biomarker that will likely play an important role in the diagnosis, monitoring, and treatment in dementias. Using tau PET imaging to examine how tau pathology relates to amyloid and other markers of neurodegeneration will serve to better understand the pathophysiologic cascade that leads to dementia.
Collapse
Affiliation(s)
- Chenjie Xia
- Department of Neurology, Jewish General Hospital, McGill University, 3755 Chemin de la Côte-Sainte-Catherine Road, Suite E-005, Montreal, QC H3T 1E2, Canada
| | - Bradford C Dickerson
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital, Harvard University, 149 13th Street, Suite 2691, Charlestown, Boston, MA 02129, USA.
| |
Collapse
|
182
|
Heurling K, Leuzy A, Jonasson M, Frick A, Zimmer ER, Nordberg A, Lubberink M. Quantitative positron emission tomography in brain research. Brain Res 2017; 1670:220-234. [PMID: 28652218 DOI: 10.1016/j.brainres.2017.06.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022]
Abstract
The application of positron emission tomography (PET) in brain research has increased substantially during the past 20years, and is still growing. PET provides a unique insight into physiological and pathological processes in vivo. In this article we introduce the fundamentals of PET, and the methods available for acquiring quantitative estimates of the parameters of interest. A short introduction to different areas of application is also given, including basic research of brain function and in neurology, psychiatry, drug receptor occupancy studies, and its application in diagnostics of neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease. Our aim is to inform the unfamiliar reader of the underlying basics and potential applications of PET, hoping to inspire the reader into considering how the technique could be of benefit for his or her own research.
Collapse
Affiliation(s)
- Kerstin Heurling
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Antoine Leuzy
- Department Neurobiology, Care Sciences and Society, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - My Jonasson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Andreas Frick
- Department of Psychology, Uppsala University, Uppsala, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Eduardo R Zimmer
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Agneta Nordberg
- Department Neurobiology, Care Sciences and Society, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden; Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mark Lubberink
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
183
|
Choi Y, Ha S, Lee YS, Kim YK, Lee DS, Kim DJ. Development of tau PET Imaging Ligands and their Utility in Preclinical and Clinical Studies. Nucl Med Mol Imaging 2017; 52:24-30. [PMID: 29391909 DOI: 10.1007/s13139-017-0484-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/10/2017] [Accepted: 05/22/2017] [Indexed: 12/16/2022] Open
Abstract
The pathological features of Alzheimer's disease are senile plaques which are aggregates of β-amyloid peptides and neurofibrillary tangles in the brain. Neurofibrillary tangles are aggregates of hyperphosphorylated tau proteins, and these induce various other neurodegenerative diseases, such as progressive supranuclear palsy, corticobasal degeneration, frontotemporal lobar degeneration, frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), and chronic traumatic encephalopathy. In the case of Alzheimer's disease, the measurement of neurofibrillary tangles associated with cognitive decline is suitable for differential diagnosis, disease progression assessment, and to monitor the effects of therapeutic treatment. This review discusses considerations for the development of tau ligands for imaging and summarizes the results of the first-in-human and preclinical studies of the tau tracers that have been developed thus far. The development of tau ligands for imaging studies will be helpful for differential diagnosis and for the development of therapeutic treatments for tauopathies including Alzheimer's disease.
Collapse
Affiliation(s)
- Yoori Choi
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,2Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744 South Korea
| | - Seunggyun Ha
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Yun-Sang Lee
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Yun Kyung Kim
- 4Institute of Brain Science, Korean Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 136-791 South Korea
| | - Dong Soo Lee
- 1Department of Nuclear Medicine, College of Medicine, Seoul National University, 110-744, 28 Yongon-Dong, Jongno-Gu, Seoul, South Korea.,2Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 110-744 South Korea.,3Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, 03080, 103 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Dong Jin Kim
- 4Institute of Brain Science, Korean Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 136-791 South Korea
| |
Collapse
|
184
|
van Eimeren T, Bischof GN, Drzezga A. Is Tau Imaging More Than Just Upside-Down 18F-FDG Imaging? J Nucl Med 2017; 58:1357-1359. [DOI: 10.2967/jnumed.117.190082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/05/2017] [Indexed: 11/16/2022] Open
|
185
|
Vandenberghe R, Schaeverbeke J. Knowing your enemy: from post-mortem scene reconstruction to real-time monitoring of the spread of tau and amyloid. Brain 2017; 140:1179-1182. [PMID: 29050370 DOI: 10.1093/brain/awx065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Belgium.,Alzheimer Research Centre KU Leuven, Leuven Institute for Neuroscience and Disease, KU Leuven, Belgium.,Neurology Department, University Hospitals Leuven, Leuven, Belgium
| | - Jolien Schaeverbeke
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Belgium.,Alzheimer Research Centre KU Leuven, Leuven Institute for Neuroscience and Disease, KU Leuven, Belgium
| |
Collapse
|
186
|
Koga S, Ono M, Sahara N, Higuchi M, Dickson DW. Fluorescence and autoradiographic evaluation of tau PET ligand PBB3 to α-synuclein pathology. Mov Disord 2017; 32:884-892. [PMID: 28440890 DOI: 10.1002/mds.27013] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/06/2017] [Accepted: 03/17/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The tau PET ligand 2-((1E,3E)-4-(6-([11 C]methylamino)pyridin-3-yl)buta-1,3-dienyl)benzo[d]thiazol-6-ol ([11 C]PBB3) binds to a wide range of tau pathology; however, binding property of PBB3 to non-tau inclusions remains unknown. To clarify whether [11 C]PBB3 binds to α-synuclein pathology, reactivity of PBB3 was assessed by in vitro fluorescence and autoradiographic labeling of brain sections from α-synucleinopathies patients. METHOD Of 10 pure Lewy body disease and 120 multiple system atrophy (MSA) cases in the Mayo Clinic brain bank, we selected 3 Lewy body disease and 4 MSA cases with a range of α-synuclein severity based on the quantitative analysis of α-synuclein burden. PBB3 fluorescence labeling, double or single immunostaining for α-synuclein and phospho-tau, Prussian blue staining, and in vitro autoradiography with [11 C]PBB3 were performed for these selected samples. RESULTS PBB3 fluorescence labeled various α-synuclein lesions including Lewy bodies, Lewy neurites, spheroids, glial cytoplasmic inclusions, and neuronal cytoplasmic inclusions. Meanwhile, autoradiographic labeling with [11 C]PBB3 at 10 nM demonstrated no significant binding in Lewy body disease cases. In contrast, significant autoradiographic binding of [11 C]PBB3 to the striatopallidal fibers was found in 2 MSA cases, which had high densities of glial cytoplasmic inclusions without tau or iron deposits in this region. CONCLUSIONS Given that the maximum concentration of [11 C]PBB3 in human PET scans is approximately 10 nM, the present data imply that α-synuclein pathology in Lewy body disease is undetectable by [11 C]PBB3-PET, whereas those in a subset of MSA cases with high densities of glial cytoplasmic inclusions could be captured by this radioligand. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Maiko Ono
- National Institutes for Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan.,Tohoku University Graduate School of Medicine, Department of Molecular Neuroimaging, Sendai, Japan
| | - Naruhiko Sahara
- National Institutes for Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Makoto Higuchi
- National Institutes for Quantum and Radiological Science and Technology, National Institute of Radiological Sciences, Chiba, Japan
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
187
|
Collier TL, Yokell DL, Livni E, Rice PA, Celen S, Serdons K, Neelamegam R, Bormans G, Harris D, Walji A, Hostetler ED, Bennacef I, Vasdev N. cGMP production of the radiopharmaceutical [ 18 F]MK-6240 for PET imaging of human neurofibrillary tangles. J Labelled Comp Radiopharm 2017; 60:263-269. [PMID: 28185305 DOI: 10.1002/jlcr.3496] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 01/15/2023]
Abstract
Fluorine-18-labelled 6-(fluoro)-3-(1H-pyrrolo[2,3-c]pyridin-1-yl)isoquinolin-5-amine ([18 F]MK-6240) is a novel potent and selective positron emission tomography (PET) radiopharmaceutical for detecting human neurofibrillary tangles, which are made up of aggregated tau protein. Herein, we report the fully automated 2-step radiosynthesis of [18 F]MK-6240 using a commercially available radiosynthesis module, GE Healthcare TRACERlab FXFN . Nucleophilic fluorination of the 5-diBoc-6-nitro precursor with potassium cryptand [18 F]fluoride (K[18 F]/K222 ) was performed by conventional heating, followed by acid deprotection and semipreparative high-performance liquid chromatography under isocratic conditions. The isolated product was diluted with formulation solution and sterile filtered under Current Good Manufacturing Practices, and quality control procedures were established to validate this radiopharmaceutical for human use. At the end of synthesis, 6.3 to 9.3 GBq (170-250 mCi) of [18 F]MK-6240 was formulated and ready for injection, in an uncorrected radiochemical yield of 7.5% ± 1.9% (relative to starting [18 F]fluoride) with a specific activity of 222 ± 67 GBq/μmol (6.0 ± 1.8 Ci/μmol) at the end of synthesis (90 minutes; n = 3). [18 F]MK-6240 was successfully validated for human PET studies meeting all Food and Drug Administration and United States Pharmacopeia requirements for a PET radiopharmaceutical. The present method can be easily adopted for use with other radiofluorination modules for widespread clinical research use.
Collapse
Affiliation(s)
- Thomas Lee Collier
- Division of Nuclear Medicine and Molecular Imaging, Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA.,Advion Inc., Ithaca, NY, USA
| | - Daniel L Yokell
- Division of Nuclear Medicine and Molecular Imaging, Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Eli Livni
- Division of Nuclear Medicine and Molecular Imaging, Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Peter A Rice
- Division of Nuclear Medicine and Molecular Imaging, Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Sofie Celen
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg O&N 2, KU Leuven, Leuven, Belgium
| | - Kim Serdons
- Department of Nuclear Medicine, UZ Leuven, Leuven, Belgium
| | - Ramesh Neelamegam
- Division of Nuclear Medicine and Molecular Imaging, Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Guy Bormans
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg O&N 2, KU Leuven, Leuven, Belgium
| | - Dawn Harris
- Merck Research Laboratories, Merck & Co., West Point, PA, USA
| | - Abbas Walji
- Merck Research Laboratories, Merck & Co., West Point, PA, USA
| | | | - Idriss Bennacef
- Merck Research Laboratories, Merck & Co., West Point, PA, USA
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Gordon Center for Medical Imaging, Massachusetts General Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
188
|
|
189
|
Saint-Aubert L, Lemoine L, Chiotis K, Leuzy A, Rodriguez-Vieitez E, Nordberg A. Tau PET imaging: present and future directions. Mol Neurodegener 2017; 12:19. [PMID: 28219440 PMCID: PMC5319037 DOI: 10.1186/s13024-017-0162-3] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/15/2017] [Indexed: 12/15/2022] Open
Abstract
Abnormal aggregation of tau in the brain is a major contributing factor in various neurodegenerative diseases. The role of tau phosphorylation in the pathophysiology of tauopathies remains unclear. Consequently, it is important to be able to accurately and specifically target tau deposits in vivo in the brains of patients. The advances of molecular imaging in the recent years have now led to the recent development of promising tau-specific tracers for positron emission tomography (PET), such as THK5317, THK5351, AV-1451, and PBB3. These tracers are now available for clinical assessment in patients with various tauopathies, including Alzheimer's disease, as well as in healthy subjects. Exploring the patterns of tau deposition in vivo for different pathologies will allow discrimination between neurodegenerative diseases, including different tauopathies, and monitoring of disease progression. The variety and complexity of the different types of tau deposits in the different diseases, however, has resulted in quite a challenge for the development of tau PET tracers. Extensive work remains in order to fully characterize the binding properties of the tau PET tracers, and to assess their usefulness as an early biomarker of the underlying pathology. In this review, we summarize recent findings on the most promising tau PET tracers to date, discuss what has been learnt from these findings, and offer some suggestions for the next steps that need to be achieved in a near future.
Collapse
Affiliation(s)
- Laure Saint-Aubert
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Laetitia Lemoine
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Konstantinos Chiotis
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Antoine Leuzy
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Elena Rodriguez-Vieitez
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden
| | - Agneta Nordberg
- Department NVS, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Novum 5th floor, 141 57, Huddinge, Sweden. .,Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
190
|
Hane FT, Robinson M, Lee BY, Bai O, Leonenko Z, Albert MS. Recent Progress in Alzheimer's Disease Research, Part 3: Diagnosis and Treatment. J Alzheimers Dis 2017; 57:645-665. [PMID: 28269772 PMCID: PMC5389048 DOI: 10.3233/jad-160907] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2016] [Indexed: 12/12/2022]
Abstract
The field of Alzheimer's disease (AD) research has grown exponentially over the past few decades, especially since the isolation and identification of amyloid-β from postmortem examination of the brains of AD patients. Recently, the Journal of Alzheimer's Disease (JAD) put forth approximately 300 research reports which were deemed to be the most influential research reports in the field of AD since 2010. JAD readers were asked to vote on these most influential reports. In this 3-part review, we review the results of the 300 most influential AD research reports to provide JAD readers with a readily accessible, yet comprehensive review of the state of contemporary research. Notably, this multi-part review identifies the "hottest" fields of AD research providing guidance for both senior investigators as well as investigators new to the field on what is the most pressing fields within AD research. Part 1 of this review covers pathogenesis, both on a molecular and macro scale. Part 2 review genetics and epidemiology, and part 3 covers diagnosis and treatment. This part of the review, diagnosis and treatment, reviews the latest diagnostic criteria, biomarkers, imaging, and treatments in AD.
Collapse
Affiliation(s)
- Francis T. Hane
- Department of Chemistry, Lakehead University, Thunder Bay, ON, Canada
- Thunder Bay Regional Research Institute, Thunder Bay, ON, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Morgan Robinson
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Brenda Y. Lee
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Owen Bai
- Thunder Bay Regional Research Institute, Thunder Bay, ON, Canada
| | - Zoya Leonenko
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
| | - Mitchell S. Albert
- Department of Chemistry, Lakehead University, Thunder Bay, ON, Canada
- Thunder Bay Regional Research Institute, Thunder Bay, ON, Canada
- Northern Ontario School of Medicine, Thunder Bay, ON, Canada
| |
Collapse
|
191
|
Neelamegam R, Yokell DL, Rice PA, Furumoto S, Kudo Y, Okamura N, El Fakhri G. A report of the automated radiosynthesis of the tau positron emission tomography radiopharmaceutical, [ 18 F]-THK-5351. J Labelled Comp Radiopharm 2016; 60:140-146. [PMID: 27859483 DOI: 10.1002/jlcr.3482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 11/08/2022]
Abstract
The radiotracer, [18 F]-THK-5351, is a highly selective and high-binding affinity PET imaging agent for aggregates of hyper-phosphorylated tau protein. Our report is a simplified 1-pot, 2-step radiosynthesis of [18 F]-THK-5351. This report is broadly applicable for routine clinical production and multi-center trials on account of favorable half-life of flourine-18 and the use of a commercially available radiosynthesis module, the GE TRACERlab™ FXFN . First, the O-THP protected tosyl precursor underwent nucleophilic fluorinating reaction with potassium cryptand fluoride ([18 F] fluoride (K[18 F]/K222 )) in Dimethyl sulfoxide at 110°C for 10 minutes followed by O-THP removal by using diluted hydrochloric acid (HCl) at same temperature. [18 F]-THK-5351 was purified via semi-preparative high-performance liquid chromatography and formulated by using 10% EtOH, United States Pharmacopeia (USP) in 0.9% sodium chloride for injection, USP and an uncorrected radiochemical yield of 21 ± 3.5%, with a specific activity of 153.11 ± 25.9 GBq/μmol (4138 ± 700 mCi/μmol) at the end of synthesis (63 minutes; n = 3).
Collapse
Affiliation(s)
- Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Daniel L Yokell
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Peter A Rice
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Shozo Furumoto
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yukitsuka Kudo
- Division of Neuro-imaging, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA.,Department of Radiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
192
|
Betthauser TJ, Lao PJ, Murali D, Barnhart TE, Furumoto S, Okamura N, Stone CK, Johnson SC, Christian BT. In Vivo Comparison of Tau Radioligands 18F-THK-5351 and 18F-THK-5317. J Nucl Med 2016; 58:996-1002. [PMID: 27856627 DOI: 10.2967/jnumed.116.182980] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/25/2016] [Indexed: 11/16/2022] Open
Abstract
This study compared the in vivo imaging characteristics of tau PET ligands 18F-THK-5351 and 18F-THK-5317 in the context of Alzheimer disease (AD). Additionally, reference tissue distribution volume ratio (DVR) estimation methods and SUV ratio (SUVR) timing windows were evaluated to determine the optimal strategy for specific binding quantification. Methods: Twenty-eight subjects (mean age ± SD, 71 ± 7 y) underwent either dynamic 90-min 18F-THK-5317 or 18F-THK-5351 PET scans. Bland-Altman plots were used to compare the simplified reference tissue method, multilinear reference tissue method (MRTM2), and Logan reference tissue DVR estimates and to assess temporal stability of SUVR windows using cerebellar gray matter as a reference region. In vivo kinetics and DVR estimates were directly compared for 10 subjects who underwent both 18F-THK-5317 and 18F-THK-5351 PET scans. Results: THK-5351 exhibited faster cerebellar gray matter clearance, faster cortical white matter clearance, and higher DVR estimates in AD tau-associated regions of interest than THK-5317. The MRTM2 method produced the most reliable DVR estimates for both tracers, particularly when scan duration was shortened to 60 min. SUVR stability was observed 50-70 min after injection for both tracers. Parametric images revealed differences between MRTM2, Logan, and SUVR binding in white matter regions for THK-5317. Conclusion: THK-5317 and THK-5351 show promise for in vivo detection of AD tau. THK-5351 has more favorable pharmacokinetics and imaging characteristics than THK-5317.
Collapse
Affiliation(s)
- Tobey J Betthauser
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin .,Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Patrick J Lao
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin.,Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Dhanabalan Murali
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Shozo Furumoto
- Division of Radiopharmaceutical Neuroimaging, Tohoku University, Sendai, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Charles K Stone
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Sterling C Johnson
- Geriatric Research Education and Clinical Center, William S. Middleton Veterans Hospital, Madison, Wisconsin; and.,Wisconsin Alzheimer's Institute, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Bradley T Christian
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin.,Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|