1
|
Kallinen A, Kassiou M. Tracer development for PET imaging of proteinopathies. Nucl Med Biol 2022; 114-115:108-120. [PMID: 35487833 DOI: 10.1016/j.nucmedbio.2022.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/17/2022] [Accepted: 04/04/2022] [Indexed: 12/27/2022]
Abstract
This review outlines small molecule radiotracers developed for positron emission tomography (PET) imaging of proteinopathies, neurodegenerative diseases characterised by accumulation of malformed proteins, over the last two decades with the focus on radioligands that have progressed to clinical studies. Introduction provides a short summary of proteinopathy targets used for PET imaging, including vastly studied proteins Aβ and tau and emerging α-synuclein. In the main section, clinically relevant Aβ and tau radioligand classes and their properties are discussed, including an overview of lead compounds and radioligand candidates studied as α-synuclein imaging agents in the early discovery and preclinical development phase. Lastly, the specific challenges and future directions in proteinopathy radioligand development are summarized.
Collapse
Affiliation(s)
- Annukka Kallinen
- Garvan Institute of Medical Research, 384 Victoria St, NSW 2010, Australia.
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, NSW 2006, Australia
| |
Collapse
|
2
|
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: 26] [Impact Index Per Article: 3.7] [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
|
3
|
Valotassiou V, Malamitsi J, Papatriantafyllou J, Dardiotis E, Tsougos I, Psimadas D, Alexiou S, Hadjigeorgiou G, Georgoulias P. SPECT and PET imaging in Alzheimer’s disease. Ann Nucl Med 2018; 32:583-593. [PMID: 30128693 DOI: 10.1007/s12149-018-1292-6] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Varvara Valotassiou
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece.
| | - Julia Malamitsi
- Medical Physics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Ioannis Tsougos
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - Dimitrios Psimadas
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - Sotiria Alexiou
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - George Hadjigeorgiou
- Neurology Department, University Hospital of Larissa, Thessaly, Greece
- Department of Neurology, Medical School, University of Cyprus, Nicosia, Greece
| | - Panagiotis Georgoulias
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| |
Collapse
|
4
|
Rafique W, Kramer V, Pardo T, Smits R, Spilhaug MM, Hoepping A, Savio E, Engler H, Kuljs R, Amaral H, Riss PJ. Image-Guided Development of Heterocyclic Sulfoxides as Ligands for Tau Neurofibrillary Tangles: From First-in-Man to Second-Generation Ligands. ACS OMEGA 2018; 3:7567-7579. [PMID: 30087917 PMCID: PMC6068598 DOI: 10.1021/acsomega.8b00975] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/15/2018] [Indexed: 06/08/2023]
Abstract
Positron emission tomography (PET) imaging of misfolded protein aggregates that form in neurodegenerative processes of the brain is key to providing a robust marker for improved diagnosis and evaluation of treatments. We report the development of advanced radiotracer candidates based on the sulfoxide scaffold found in proton pump inhibitors (lansoprazole, prevacid) with inherent affinity to neurofibrillary tangles in Alzheimer's disease and related disorders (e.g., dementia with Lewy bodies and the frontotemporal degeneration syndrome). First-in-man results obtained with [18F]lansoprazole and N-methyl-[18F]lansoprazole were used to guide the design of a set of 24 novel molecules with suitable properties for neuroimaging with PET. Compounds were synthesized and characterized pharmacologically, and the binding affinity of the compounds to synthetic human tau-441 fibrils was determined. Selectivity of binding was assessed using α-synuclein and β-amyloid fibrils to address the key misfolded proteins of relevance in dementia. To complete the pharmacokinetic profiling in vitro, plasma protein binding and lipophilicity were investigated. Highly potent and selective new radiotracer candidates were identified for further study.
Collapse
Affiliation(s)
- Waqas Rafique
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands
vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Vasko Kramer
- Positronpharma
SA, Rancagua 878, 7500921 Providencia, Santiago, Chile
- Center
of Nuclear Medicine Positronmed, Julio Prado 714, 7501068 Providencia, Santiago, Chile
| | - Tania Pardo
- Departamento
de Montevideo, Uruguayan Centre of Molecular
Imaging (CUDIM), Av.
Dr. Américo Ricaldoni 2010, 11600 Montevideo, Uruguay
| | - René Smits
- Advanced
Biochemical Compounds GmbH, Heinrich-Glaeser-Strasse 10-14, D-01454 Radeberg, Germany
| | - Mona M. Spilhaug
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands
vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Alexander Hoepping
- Advanced
Biochemical Compounds GmbH, Heinrich-Glaeser-Strasse 10-14, D-01454 Radeberg, Germany
| | - Eduardo Savio
- Departamento
de Montevideo, Uruguayan Centre of Molecular
Imaging (CUDIM), Av.
Dr. Américo Ricaldoni 2010, 11600 Montevideo, Uruguay
| | - Henry Engler
- Departamento
de Montevideo, Uruguayan Centre of Molecular
Imaging (CUDIM), Av.
Dr. Américo Ricaldoni 2010, 11600 Montevideo, Uruguay
| | - Rodrigo Kuljs
- Zdrav
Mozak Clinical Neuroscience Center, Julio Prado 714, 7501068 Providencia, Santiago, Chile
| | - Horacio Amaral
- Positronpharma
SA, Rancagua 878, 7500921 Providencia, Santiago, Chile
- Center
of Nuclear Medicine Positronmed, Julio Prado 714, 7501068 Providencia, Santiago, Chile
| | - Patrick J. Riss
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands
vei 26, Kjemibygningen, 0371 Oslo, Norway
- Klinik
for Kirurgi og Nevrofag, Oslo Universitets
Sykehus HF—Rikshospitalet, Postboks
4950 Nydalen, 0424 Oslo, Norway
- Norsk
Medisinsk Syklotronsenter AS, Gaustad,
Postboks 4950 Nydalen, 0424 Oslo, Norway
| |
Collapse
|
5
|
Barrio JR. The Irony of PET Tau Probe Specificity. J Nucl Med 2017; 59:115-116. [DOI: 10.2967/jnumed.117.198960] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 11/16/2022] Open
|
6
|
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: 99] [Impact Index Per Article: 12.4] [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
|
7
|
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: 1.1] [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
|
8
|
Insight into the Molecular Imaging of Alzheimer's Disease. Int J Biomed Imaging 2016; 2016:7462014. [PMID: 26880871 PMCID: PMC4736963 DOI: 10.1155/2016/7462014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/16/2015] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease is a complex neurodegenerative disease affecting millions of individuals worldwide. Earlier it was diagnosed only via clinical assessments and confirmed by postmortem brain histopathology. The development of validated biomarkers for Alzheimer's disease has given impetus to improve diagnostics and accelerate the development of new therapies. Functional imaging like positron emission tomography (PET), single photon emission computed tomography (SPECT), functional magnetic resonance imaging (fMRI), and proton magnetic resonance spectroscopy provides a means of detecting and characterising the regional changes in brain blood flow, metabolism, and receptor binding sites that are associated with Alzheimer's disease. Multimodal neuroimaging techniques have indicated changes in brain structure and metabolic activity, and an array of neurochemical variations that are associated with neurodegenerative diseases. Radiotracer-based PET and SPECT potentially provide sensitive, accurate methods for the early detection of disease. This paper presents a review of neuroimaging modalities like PET, SPECT, and selected imaging biomarkers/tracers used for the early diagnosis of AD. Neuroimaging with such biomarkers and tracers could achieve a much higher diagnostic accuracy for AD and related disorders in the future.
Collapse
|
9
|
Pike VW. Considerations in the Development of Reversibly Binding PET Radioligands for Brain Imaging. Curr Med Chem 2016; 23:1818-69. [PMID: 27087244 PMCID: PMC5579844 DOI: 10.2174/0929867323666160418114826] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/04/2016] [Accepted: 04/15/2016] [Indexed: 12/17/2022]
Abstract
The development of reversibly binding radioligands for imaging brain proteins in vivo, such as enzymes, neurotransmitter transporters, receptors and ion channels, with positron emission tomography (PET) is keenly sought for biomedical studies of neuropsychiatric disorders and for drug discovery and development, but is recognized as being highly challenging at the medicinal chemistry level. This article aims to compile and discuss the main considerations to be taken into account by chemists embarking on programs of radioligand development for PET imaging of brain protein targets.
Collapse
Affiliation(s)
- Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Rm. B3C346A, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
10
|
Okamura N, Harada R, Furumoto S, Arai H, Yanai K, Kudo Y. Tau PET imaging in Alzheimer's disease. Curr Neurol Neurosci Rep 2015; 14:500. [PMID: 25239654 DOI: 10.1007/s11910-014-0500-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In several neurodegenerative diseases that are collectively called tauopathies, progressive accumulation of tau in the brain is closely associated with neurodegeneration and cognitive impairment. Noninvasive detection of tau protein deposits in the brain would be useful to diagnose tauopathies as well as to track and predict disease progression. Recently, several tau PET tracers including T807, THK-5117, and PBB3 have been developed and succeeded in imaging neurofibrillary pathology in vivo. For use of tau PET as a biomarker of tau pathology in Alzheimer's disease, PET tracers should have high affinity to PHF-tau and high selectivity for tau over amyloid-β and other protein deposits. PET tau imaging enables the longitudinal assessment of the spatial pattern of tau deposition and its relation to amyloid-β pathology and neurodegeneration. This technology could also be applied to the pharmacological assessment of anti-tau therapy, thereby allowing preventive interventions.
Collapse
Affiliation(s)
- Nobuyuki Okamura
- Department of Pharmacology, Tohoku University School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Sendai, 9808575, Japan,
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
The military conflicts of the last decade have highlighted the growing problem of traumatic brain injury in combatants returning from the battlefield. The considerable evidence pointing at the accumulation of tau aggregates and its recognition as a risk factor in neurodegenerative conditions such as Alzheimer's disease have led to a major effort to develop selective tau ligands that would allow research into the physiopathologic underpinnings of traumatic brain injury and chronic traumatic encephalopathy in military personnel and the civilian population. These tracers will allow new insights into tau pathology in the human brain, facilitating research into causes, diagnosis, and treatment of traumatic encephalopathy and major neurodegenerative dementias, such as Alzheimer's disease and some variants of frontotemporal lobar degeneration, in which tau plays a role. The field of selective tau imaging has to overcome several obstacles, some of them associated with the idiosyncrasies of tau aggregation and others related to radiotracer design. A worldwide effort has focused on the development of imaging agents that will allow selective tau imaging in vivo. Recent progress in the development of these tracers is enabling the noninvasive assessment of the extent of tau pathology in the brain, eventually allowing the quantification of changes in tau pathology over time and its relation to cognitive performance, brain volumetrics, and other biomarkers, as well as assessment of efficacy and patient recruitment for antitau therapeutic trials.
Collapse
Affiliation(s)
- Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Melbourne, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
| | - Nobuyuki Okamura
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
12
|
Villemagne VL, Fodero-Tavoletti MT, Masters CL, Rowe CC. Tau imaging: early progress and future directions. Lancet Neurol 2015; 14:114-24. [PMID: 25496902 DOI: 10.1016/s1474-4422(14)70252-2] [Citation(s) in RCA: 365] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Use of selective in-vivo tau imaging will enable improved understanding of tau aggregation in the brain, facilitating research into causes, diagnosis, and treatment of major tauopathies such as Alzheimer's disease, progressive supranuclear palsy, corticobasal syndrome, chronic traumatic encephalopathy, and some variants of frontotemporal lobar degeneration. Neuropathological studies of Alzheimer's disease show a strong association between tau deposits, decreased cognitive function, and neurodegenerative changes. Selective tau imaging will allow the in-vivo exploration of such associations and measure the global and regional changes in tau deposits over time. Such imaging studies will comprise non-invasive assessment of the spatial and temporal pattern of tau deposition over time, providing insight into the role tau plays in ageing and helping to establish the relation between cognition, genotype, neurodegeneration, and other biomarkers. Once validated, selective tau imaging might be useful as a diagnostic, prognostic, and progression biomarker, and a surrogate marker for the monitoring of efficacy and patient recruitment for anti-tau therapeutic trials.
Collapse
Affiliation(s)
- Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, VIC, Australia; Department of Medicine, The University of Melbourne, Austin Health, VIC, Australia; The Florey Institute, The University of Melbourne, Victoria, Australia.
| | - Michelle T Fodero-Tavoletti
- Department of Nuclear Medicine and Centre for PET, Austin Health, VIC, Australia; The Florey Institute, The University of Melbourne, Victoria, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Victoria, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, VIC, Australia; Department of Medicine, The University of Melbourne, Austin Health, VIC, Australia
| |
Collapse
|
13
|
Fawaz M, Brooks AF, Rodnick ME, Carpenter GM, Shao X, Desmond TJ, Sherman P, Quesada CA, Hockley BG, Kilbourn MR, Albin RL, Frey KA, Scott PJH. High affinity radiopharmaceuticals based upon lansoprazole for PET imaging of aggregated tau in Alzheimer's disease and progressive supranuclear palsy: synthesis, preclinical evaluation, and lead selection. ACS Chem Neurosci 2014; 5:718-30. [PMID: 24896980 PMCID: PMC4140593 DOI: 10.1021/cn500103u] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 06/04/2014] [Indexed: 01/05/2023] Open
Abstract
Abnormally aggregated tau is the hallmark pathology of tauopathy neurodegenerative disorders and is a target for development of both diagnostic tools and therapeutic strategies across the tauopathy disease spectrum. Development of carbon-11- or fluorine-18-labeled radiotracers with appropriate affinity and specificity for tau would allow noninvasive quantification of tau burden using positron emission tomography (PET) imaging. We have synthesized [(18)F]lansoprazole, [(11)C]N-methyl lansoprazole, and [(18)F]N-methyl lansoprazole and identified them as high affinity radiotracers for tau with low to subnanomolar binding affinities. Herein, we report radiosyntheses and extensive preclinical evaluation with the aim of selecting a lead radiotracer for translation into human PET imaging trials. We demonstrate that [(18)F]N-methyl lansoprazole, on account of the favorable half-life of fluorine-18 and its rapid brain entry in nonhuman primates, favorable kinetics, low white matter binding, and selectivity for binding to tau over amyloid, is the lead compound for progression into clinical trials.
Collapse
Affiliation(s)
- Maria
V. Fawaz
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Allen F. Brooks
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Melissa E. Rodnick
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Garrett M. Carpenter
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Xia Shao
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Timothy J. Desmond
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Phillip Sherman
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Carole A. Quesada
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Brian G. Hockley
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Michael R. Kilbourn
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Roger L. Albin
- Geriatrics
Research, Education and Clinical Center, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan 48105, United States
- Department
of Neurology, The University of Michigan
Medical School, Ann Arbor, Michigan 48109, United States
- Michigan Alzheimer
Disease Center and The Interdepartmental Program in
Medicinal Chemistry, The University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kirk A. Frey
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Peter J. H. Scott
- Division
of Nuclear Medicine, Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Michigan Alzheimer
Disease Center and The Interdepartmental Program in
Medicinal Chemistry, The University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
14
|
Herholz K. The role of PET quantification in neurological imaging: FDG and amyloid imaging in dementia. Clin Transl Imaging 2014. [DOI: 10.1007/s40336-014-0073-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
15
|
Boländer A, Kieser D, Scholz C, Heyny-von Haußen R, Mall G, Goetschy V, Czech C, Schmidt B. Synthesis of Methoxy-X04 Derivatives and Their Evaluation in Alzheimer's Disease Pathology. NEURODEGENER DIS 2013; 13:209-13. [DOI: 10.1159/000351436] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 04/18/2013] [Indexed: 11/19/2022] Open
|
16
|
Cisek K, Jensen JR, Honson NS, Schafer KN, Cooper GL, Kuret J. Ligand electronic properties modulate tau filament binding site density. Biophys Chem 2012; 170:25-33. [PMID: 23072817 DOI: 10.1016/j.bpc.2012.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/05/2012] [Accepted: 09/06/2012] [Indexed: 10/27/2022]
Abstract
Small molecules that bind tau-bearing neurofibrillary lesions are being sought for premortem diagnosis, staging, and treatment of Alzheimer's disease and other tauopathic neurodegenerative diseases. The utility of these agents will depend on both their binding affinity and binding site density (B(max)). Previously we identified polarizability as a descriptor of protein aggregate binding affinity. To examine its contribution to binding site density, we investigated the ability of two closely related benzothiazole derivatives ((E)-2-[[4-(dimethylamino)phenyl]azo]-6-methoxybenzothiazole) and ((E)-2-[2-[4-(dimethylamino)phenyl]ethenyl]-6-methoxybenzothiazole) that differed in polarizability to displace probes of high (Thioflavin S) and low (radiolabeled (E,E)-1-iodo-2,5-bis(3-hydroxycarbonyl-4-methoxy)styrylbenzene; IMSB) density sites. Consistent with their site densities, Thioflavin S completely displaced radiolabeled IMSB, but IMSB was incapable of displacing Thioflavin S. Although both benzothiazoles displaced the low B(max) IMSB probe, only the highly polarizable analog displaced near saturating concentrations of the Thioflavin S probe. Quantum calculations showed that high polarizability reflected extensive pi-electron delocalization fostered by the presence of electron donating and accepting groups. These data suggest that electron delocalization promotes ligand binding at a subset of sites on tau aggregates that are present at high density, and that optimizing this aspect of ligand structure can yield tau-directed agents with superior diagnostic and therapeutic performance.
Collapse
Affiliation(s)
- Katryna Cisek
- Department of Molecular and Cellular Biochemistry, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
17
|
Boländer A, Kieser D, Voss C, Bauer S, Schön C, Burgold S, Bittner T, Hölzer J, Heyny-von Haußen R, Mall G, Goetschy V, Czech C, Knust H, Berger R, Herms J, Hilger I, Schmidt B. Bis(arylvinyl)pyrazines, -pyrimidines, and -pyridazines as imaging agents for tau fibrils and β-amyloid plaques in Alzheimer's disease models. J Med Chem 2012; 55:9170-80. [PMID: 22913544 DOI: 10.1021/jm300653b] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The in vivo diagnosis of Alzheimer's disease (AD) is of high socioeconomic interest and remains a demanding field of research. The biopathological hallmarks of the disease are extracellular plaques consisting of aggregated β-amyloid peptides (Aβ) and tau protein derived intracellular tangles. Here we report the synthesis and evaluation of fluorescent pyrazine, pyrimidine,and pyridazine derivatives in vitro and in vivo aiming at a tau-based diagnosis of AD. The probes were pre-evaluated on human brain tissue by fluorescence microscopy and were found to label all known disease-related alterations at high contrast and specificity. To quantify the binding affinity, a new thiazine red displacement assay was developed and selected candidates were toxicologically profiled. The application in transgenic mouse models demonstrated bioavailability and brain permeability for one compound. In the course of histological testing, we discovered an AD-related deposition of tau aggregates in the Bowman's glands of the olfactory epithelium, which holds potential for an endoscopic diagnosis of AD in the olfactory system.
Collapse
Affiliation(s)
- Alexander Boländer
- Clemens Schoepf-Institute of Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Petersenstrasse 22, 64287 Darmstadt, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Villemagne VL, Furumoto S, Fodero-Tavoletti M, Harada R, Mulligan RS, Kudo Y, Masters CL, Yanai K, Rowe CC, Okamura N. The challenges of tau imaging. FUTURE NEUROLOGY 2012. [DOI: 10.2217/fnl.12.34] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In vivo imaging of tau pathology will provide new insights into tau deposition in the human brain, thus facilitating research into causes, diagnosis and treatment of major dementias, such as Alzheimer’s disease, or some variants of frontotemporal lobar degeneration, in which tau plays a role. Tau imaging poses several challenges, some related to the singularities of tau aggregation, and others related to radiotracer design. Several groups around the world are working on the development of imaging agents that will allow the in vivo assessment of tau deposition in aging and in neurodegeneration. Development of a tau imaging tracer will enable researchers to noninvasively examine the degree and extent of tau pathology in the brain, quantify changes in tau deposition over time, evaluate its relation to cognition and assess the efficacy of anti-tau therapy.
Collapse
Affiliation(s)
- Victor L Villemagne
- Department of Nuclear Medicine & Centre for PET, Austin Health, 145 Studley Road, Heidelberg, VIC 3084, Melbourne, Australia
| | - Shozo Furumoto
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| | | | - Ryuichi Harada
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| | - Rachel S Mulligan
- Department of Nuclear Medicine & Centre for PET, Austin Health, 145 Studley Road, Heidelberg, VIC 3084, Melbourne, Australia
| | - Yukitsuka Kudo
- Innovation of New Biomedical Engineering Center, Tohoku University, Sendai, Japan
| | | | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| | - Chistopher C Rowe
- Department of Nuclear Medicine & Centre for PET, Austin Health, 145 Studley Road, Heidelberg, VIC 3084, Melbourne, Australia
| | - Nobuyuki Okamura
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|