Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 75
1.
Mol Imaging Biol ; 25(6): 1115-1124, 2023 Dec.
Article En | MEDLINE | ID: mdl-37580462

PURPOSE: Noninvasive imaging of protein aggregates in the brain is critical for the early diagnosis, disease monitoring, and evaluation of the effectiveness of novel therapies for Alzheimer's disease (AD). Near-infrared fluorescence (NIRF) imaging with specific probes is a promising technique for the in vivo detection of protein deposits without radiation exposure. Comprehensive screening of fluorescent compounds identified a novel compound, THK-565, for the in vivo imaging of amyloid-ß (Aß) deposits in the mouse brain. This study assessed whether THK-565 could detect amyloid-ß deposits in vivo in the AD mouse model. PROCEDURES: The fluorescent properties of THK-565 were evaluated in the presence and absence of Aß fibrils. APP knock-in (APP-KI) mice were used as an animal model of AD. In vivo NIRF images were acquired after the intravenous administration of THK-565 and THK-265 in mice. The binding selectivity of THK-565 to Aß was evaluated using brain slices obtained from these mouse models. RESULTS: The fluorescence intensity of the THK-565 solution substantially increased by mixing with Aß fibrils. The maximum emission wavelength of the complex of THK-565 and Aß fibrils was 704 nm, which was within the optical window range. THK-565 selectively bound to amyloid deposits in brain sections of APP-KI mice After the intravenous administration of THK-565, the fluorescence signal in the head of APP-KI mice was significantly higher than that of wild-type mice and higher than that after administration of THK-265. Ex vivo analysis confirmed that the THK-565 signal corresponded to Aß immunostaining in the brain sections of these mice. CONCLUSIONS: A novel NIRF probe, THK-565, enabled the in vivo detection of Aß deposits in the brains of the AD mouse model, suggesting that NIRF imaging with THK-565 could non-invasively assess disease-specific pathology in AD.


Alzheimer Disease , Mice , Animals , Alzheimer Disease/metabolism , Plaque, Amyloid/metabolism , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Disease Models, Animal , Fluorescent Dyes/chemistry , Mice, Transgenic
2.
J Nucl Med ; 64(9): 1495-1501, 2023 09.
Article En | MEDLINE | ID: mdl-37321821

Tau PET tracers are expected to be sufficiently sensitive to track the progression of age-related tau pathology in the medial temporal cortex. The tau PET tracer N-(4-[18F]fluoro-5-methylpyridin-2-yl)-7-aminoimidazo[1,2-a]pyridine ([18F]SNFT-1) has been successfully developed by optimizing imidazo[1,2-a]pyridine derivatives. We characterized the binding properties of [18F]SNFT-1 using a head-to-head comparison with other reported 18F-labeled tau tracers. Methods: The binding affinity of SNFT-1 to tau, amyloid, and monoamine oxidase A and B was compared with that of the second-generation tau tracers MK-6240, PM-PBB3, PI-2620, RO6958948, JNJ-64326067, and flortaucipir. In vitro binding properties of 18F-labeled tau tracers were evaluated through the autoradiography of frozen human brain tissues from patients with diverse neurodegenerative disease spectra. Pharmacokinetics, metabolism, and radiation dosimetry were assessed in normal mice after intravenous administration of [18F]SNFT-1. Results: In vitro binding assays demonstrated that [18F]SNFT-1 possesses high selectivity and high affinity for tau aggregates in Alzheimer disease (AD) brains. Autoradiographic analysis of tau deposits in medial temporal brain sections from patients with AD showed a higher signal-to-background ratio for [18F]SNFT-1 than for the other tau PET tracers and no significant binding with non-AD tau, α-synuclein, transactiviation response DNA-binding protein-43, and transmembrane protein 106B aggregates in human brain sections. Furthermore, [18F]SNFT-1 did not bind significantly to various receptors, ion channels, or transporters. [18F]SNFT-1 showed a high initial brain uptake and rapid washout from the brains of normal mice without radiolabeled metabolites. Conclusion: These preclinical data suggest that [18F]SNFT-1 is a promising and selective tau radiotracer candidate that allows the quantitative monitoring of age-related accumulation of tau aggregates in the human brain.


Alzheimer Disease , Neurodegenerative Diseases , Humans , Mice , Animals , Neurodegenerative Diseases/metabolism , Alzheimer Disease/metabolism , Pyridines/pharmacokinetics , Brain/metabolism , tau Proteins/metabolism , Positron-Emission Tomography
3.
PLoS One ; 18(6): e0287047, 2023.
Article En | MEDLINE | ID: mdl-37315033

Astrogliosis is a crucial feature of neuroinflammation and is characterized by the significant upregulation of glial fibrillary acidic protein (GFAP) expression. Hence, visualizing GFAP in the living brain of patients with damaged central nervous system using positron emission tomography (PET) is of great importance, and it is expected to depict neuroinflammation more directly than existing neuroinflammation imaging markers. However, no PET radiotracers for GFAP are currently available. Therefore, neuroimaging with antibody-like affinity proteins could be a viable strategy for visualizing imaging targets that small molecules rarely recognize, such as GFAP, while we need to overcome the challenges of slow clearance and low brain permeability. The E9 nanobody, a small-affinity protein with high affinity and selectivity for GFAP, was utilized in this study. E9 was engineered by fusing a brain shuttle peptide that facilitates blood-brain barrier permeation via two different types of linker domains: E9-GS-ApoE (EGA) and E9-EAK-ApoE (EEA). E9, EGA and EEA were radiolabeled with fluorine-18 using cell-free protein radiosynthesis. In vitro autoradiography showed that all radiolabeled proteins exhibited a significant difference in neuroinflammation in the brain sections created from a rat model constructed by injecting lipopolysaccharide (LPS) into the unilateral striatum of wildtype rats, and an excess competitor displaced their binding. However, exploratory in vivo PET imaging and ex vivo biodistribution studies in the rat model failed to distinguish neuroinflammatory lesions within 3 h of 18F-EEA intravenous injection. This study contributes to a better understanding of the characteristics of small-affinity proteins fused with a brain shuttle peptide for further research into the use of protein molecules as PET tracers for imaging neuropathology.


Neuroinflammatory Diseases , Tomography, X-Ray Computed , Animals , Rats , Apolipoproteins E , Brain/diagnostic imaging , Glial Fibrillary Acidic Protein , Peptides , Tissue Distribution , Single-Domain Antibodies
4.
J Neuropathol Exp Neurol ; 82(5): 427-437, 2023 04 20.
Article En | MEDLINE | ID: mdl-36882045

Quantification of in vivo reactive astrogliosis, which represents neural inflammation and remodeling in the brain, is an emerging methodology for the evaluation of patients with neurodegenerative diseases. [18F]THK-5351 is a positron emission tomography (PET) tracer for monoamine oxidase B (MAO-B), a molecular marker of reactive astrogliosis. We performed in vivo [18F]THK-5351 PET in a patient who at autopsy was found to have argyrophilic grain disease (AGD) with comorbid pathology to visualize reactive astrogliosis for the first time. We aimed to validate an imaging-pathology correlation using [18F]THK-5351 PET and the autopsy brain. The patient, a 78-year-old man, was pathologically diagnosed with AGD combined with limbic-predominant age-related transactive response DNA-binding protein of 43 kDa encephalopathy and Lewy body disease without Alzheimer disease-related neuropathological changes. Reactive astrogliosis in the postmortem brain was abundant in the inferior temporal gyrus, insular gyrus, entorhinal cortex, and ambient gyrus where premortem [18F]THK-5351 signals were high. We found a proportional correlation between the amount of reactive astrogliosis in the postmortem brain and the in vivo [18F]THK-5351 standardized uptake value ratio (r = 0.8535, p = 0.0004). These results indicated that reactive astrogliosis in AGD with comorbid pathology could be identified and quantified by in vivo MAO-B imaging.


Alzheimer Disease , Neurodegenerative Diseases , Male , Humans , Aged , Gliosis/pathology , Alzheimer Disease/pathology , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/metabolism , Brain/pathology , Positron-Emission Tomography , Monoamine Oxidase/metabolism , tau Proteins/metabolism
5.
Nihon Yakurigaku Zasshi ; 158(1): 26-29, 2023.
Article Ja | MEDLINE | ID: mdl-36596483

Glial cells are non-neuronal cells that make up the central nervous system, including astrocytes, oligodendrocytes, microglia, and ependymal cells, which play an important role in brain homeostasis. However, activated microglia and reactive astrocytes cause neuroinflammation, which is closely related to neurodegeneration. Neuronal loss, gliosis, and accumulation of misfolded proteins are commonly observed in the brain of many neurodegenerative diseases at autopsy. Therefore, in vivo imaging of glial cell responses by positron emission tomography (PET) would be useful not only for understanding pathological processes, but also for differential diagnosis and evaluation of disease-modifying therapeutics targeting glial cells. The gold standard marker for reactive astrocytes is glial fibrillary acidic protein (GFAP), but no specific ligands are available. To date, there are two targets of reactive astrocytes that are under intense investigation: Monoamine oxidase-B (MAO-B) and imidazoline2 binding site (I2BS). PET radiopharmaceuticals for MAO-B and I2BS have been developed and are under clinical investigation. In this chapter, we review the MAO-B and I2BS as molecular targets for imaging reactive astrocytes and introduce the PET tracers and their clinical studies.


Gliosis , Positron-Emission Tomography , Humans , Gliosis/diagnostic imaging , Gliosis/metabolism , Gliosis/pathology , Positron-Emission Tomography/methods , Brain/diagnostic imaging , Brain/metabolism , Astrocytes/metabolism , Monoamine Oxidase/metabolism
6.
Nihon Yakurigaku Zasshi ; 157(6): 453-457, 2022.
Article Ja | MEDLINE | ID: mdl-36328560

Alzheimer's disease (AD) is one of the most common causes of dementia in the world. Neurodegeneration, gliosis, and misfolded proteins such as amyloid plaques and tau tangles are neuropathological hallmarks in AD. In vivo imaging of these neuropathological lesions would be good biomarkers to understand pathophysiology as well as surrogate markers for clinical trials. We developed THK tau radiotracers including [18F]THK-5351 and tested them in humans. Validations studies identified monoamine oxidase-B (MAO-B) as the off-target binding substrate of [18F]THK-5351. Since the elevation of MAO-B, which is highly expressed in reactive astrocytes, were observed in various neurological conditions, MAO-B would be a promising target for imaging reactive astrogliosis. In fact, [18F]THK-5351 PET studies demonstrated that high tracer uptake in site susceptible regions to occur astrogliosis in various neurological disorders. However, the lack of binding selectivity affects the interpretation of PET images. Therefore, we performed lead optimization from [18F]THK-5351 generating a selective and reversible MAO-B PET tracer, [18F]SMBT-1. These translational and reverse translational studies, from the development of PET tracers to validation of PET images, led to the generation of new biomarkers. In this review, we will introduce the development of [18F]THK-5351, identification of off-target binding substrates, imaging-autopsy validations, new tracer development ([18F]SMBT-1), and finally recent clinical studies of [18F]SMBT-1.


Alzheimer Disease , Quinolines , Humans , Positron-Emission Tomography/methods , Gliosis/metabolism , Brain/diagnostic imaging , Brain/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Monoamine Oxidase/metabolism , Biomarkers , tau Proteins/metabolism
9.
Front Neurosci ; 16: 807435, 2022.
Article En | MEDLINE | ID: mdl-35210989

Many neurodegenerative diseases are neuropathologically characterized by neuronal loss, gliosis, and the deposition of misfolded proteins such as ß-amyloid (Aß) plaques and tau tangles in Alzheimer's disease (AD). In postmortem AD brains, reactive astrocytes and activated microglia are observed surrounding Aß plaques and tau tangles. These activated glial cells secrete pro-inflammatory cytokines and reactive oxygen species, which may contribute to neurodegeneration. Therefore, in vivo imaging of glial response by positron emission tomography (PET) combined with Aß and tau PET would provide new insights to better understand the disease process, as well as aid in the differential diagnosis, and monitoring glial response disease-specific therapeutics. There are two promising targets proposed for imaging reactive astrogliosis: monoamine oxidase-B (MAO-B) and imidazoline2 binding site (I2BS), which are predominantly expressed in the mitochondrial membranes of astrocytes and are upregulated in various neurodegenerative conditions. PET tracers targeting these two MAO-B and I2BS have been evaluated in humans. [18F]THK-5351, which was originally designed to target tau aggregates in AD, showed high affinity for MAO-B and clearly visualized reactive astrocytes in progressive supranuclear palsy (PSP). However, the lack of selectivity of [18F]THK-5351 binding to both MAO-B and tau, severely limits its clinical utility as a biomarker. Recently, [18F]SMBT-1 was developed as a selective and reversible MAO-B PET tracer via compound optimization of [18F]THK-5351. In this review, we summarize the strategy underlying molecular imaging of reactive astrogliosis and clinical studies using MAO-B and I2BS PET tracers.

10.
ACS Chem Neurosci ; 13(3): 322-329, 2022 02 02.
Article En | MEDLINE | ID: mdl-35049267

(S)-(2-Methylpyrid-5-yl)-6-[(3-[18F]fluoro-2-hydroxy)propoxy]quinoline ([18F]SMBT-1) was recently developed as a novel class of selective and reversible monoamine oxidase-B (MAO-B) tracers for in vivo imaging of reactive astrogliosis via positron emission tomography. To investigate the effect of the chirality of [18F]SMBT-1 on tracer performance, we synthesized (S)-[18F]6 ([18F]SMBT-1) and (R)-[18F]6 and compared their binding properties, pharmacokinetics, and metabolism. (S)-6 showed higher binding affinity to MAO-B and lower binding affinity to MAO-A than (R)-6, demonstrating a higher selectivity ratio (MAO-B/MAO-A). A pharmacokinetic study in mice demonstrated that both (S)-[18F]6 and (R)-[18F]6 showed sufficient initial brain uptake. However, (S)-[18F]6 was cleared significantly faster from the body. An abundant sulfoconjugate metabolite M2 was observed in plasma for (S)-[18F]6 but not for (R)-[18F]6. In vitro sulfation assays confirmed that (S)-6 was more reactive than (R)-6, consistent with the in vivo findings. Mefenamic acid, a selective sulfotransferase 1A1 (SULT1A1) inhibitor, strongly inhibited the in vitro sulfation of (S)-6 by mouse liver fractions, human liver cytosol fractions, and human recombinant SULT1A1 enzyme. Genetic polymorphisms of SULT1A1 did not affect the sulfation of (S)-6 in vitro. In conclusion, (S)-[18F]6 had a more favorable binding affinity and binding selectivity for MAO-B than (R)-[18F]6. Additionally, (S)-[18F]6 also possessed better pharmacological and metabolic properties than (R)-[18F]6. These results suggest that (S)-[18F]6 ([18F]SMBT-1) is a promising candidate for application in the imaging of MAO-B in vivo.


Monoamine Oxidase , Positron-Emission Tomography , Animals , Brain , Gliosis , Mice , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemistry , Positron-Emission Tomography/methods
11.
J Nucl Med ; 63(10): 1560-1569, 2022 10.
Article En | MEDLINE | ID: mdl-35086892

A neuroinflammatory reaction in Alzheimer disease (AD) brains involves reactive astrocytes that overexpress monoamine oxidase-B (MAO-B). 18F-(S)-(2-methylpyrid-5-yl)-6-[(3-fluoro-2-hydroxy)propoxy]quinoline (18F-SMBT-1) is a novel 18F PET tracer highly selective for MAO-B. We characterized the clinical performance of 18F-SMBT-1 PET across the AD continuum as a potential surrogate marker of reactive astrogliosis. Methods: We assessed 18F-SMBT-1 PET regional binding in 77 volunteers (76 ± 5.5 y old; 41 women, 36 men) across the AD continuum: 57 who were cognitively normal (CN) (44 amyloid-ß [Aß]-negative [Aß-] and 13 Aß-positive [Aß+]), 12 who had mild cognitive impairment (9 Aß- and 3 Aß+), and 8 who had AD dementia (6 Aß+ and 2 Aß-). All participants also underwent Aß and tau PET imaging, 3-T MRI, and neuropsychologic evaluation. Tau imaging results were expressed in SUV ratios using the cerebellar cortex as a reference region, whereas Aß burden was expressed in centiloids. 18F-SMBT-1 outcomes were expressed as SUV ratio using the subcortical white matter as a reference region. Results: 18F-SMBT-1 yielded high-contrast images at steady state (60-80 min after injection). When compared with the Aß- CN group, there were no significant differences in 18F-SMBT-1 binding in the group with Aß- mild cognitive impairment. Conversely, 18F-SMBT-1 binding was significantly higher in several cortical regions in the Aß+ AD group but also was significantly lower in the mesial temporal lobe and basal ganglia. Most importantly, 18F-SMBT-1 binding was significantly higher in the same regions in the Aß+ CN group as in the Aß- CN group. When all clinical groups were considered together, 18F-SMBT-1 correlated strongly with Aß burden and much less with tau burden. Although in most cortical regions 18F-SMBT-1 did not correlate with brain volumetrics, regions known for high MAO-B concentrations presented a direct association with hippocampal and gray matter volumes, whereas the occipital lobe was directly associated with white matter hyperintensity. 18F-SMBT-1 binding was inversely correlated with Mini Mental State Examination and the Australian Imaging Biomarkers and Lifestyle's Preclinical Alzheimer Cognitive Composite in some neocortical regions such as the frontal cortex, lateral temporal lobe, and supramarginal gyrus. Conclusion: Cross-sectional human PET studies with 18F-SMBT-1 showed that Aß+ AD patients, but most importantly, Aß+ CN individuals, had significantly higher regional 18F-SMBT-1 binding than Aß- CN individuals. Moreover, in several regions in the brain, 18F-SMBT-1 retention was highly associated with Aß load. These findings suggest that increased 18F-SMBT-1 binding is detectable at the preclinical stages of Aß accumulation, providing strong support for its use as a surrogate marker of astrogliosis in the AD continuum.


Alzheimer Disease , Quinolines , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Australia , Biomarkers , Cross-Sectional Studies , Female , Gliosis , Humans , Inflammation , Male , Monoamine Oxidase
12.
J Nucl Med ; 63(10): 1551-1559, 2022 10.
Article En | MEDLINE | ID: mdl-35086898

Reactive gliosis, characterized by reactive astrocytes and activated microglia, contributes greatly to neurodegeneration throughout the course of Alzheimer disease (AD). Reactive astrocytes overexpress monoamine oxidase B (MAO-B). We characterized the clinical performance of 18F-(S)-(2-methylpyrid-5-yl)-6-[(3-fluoro-2-hydroxy)propoxy]quinoline (18F-SMBT-1), a novel MAO-B PET tracer as a potential surrogate marker of reactive astrogliosis. Methods: Seventy-seven participants-53 who were elderly and cognitively normal, 7 with mild cognitive impairment, 7 with AD, and 10 who were young and cognitively normal-were recruited for the different aspects of the study. Older participants underwent 3-dimensional magnetization-prepared rapid gradient-echo MRI and amyloid-ß, tau, and 18F-SMBT-1 PET. To ascertain 18F-SMBT-1 selectivity to MAO-B, 9 participants underwent 2 18F-SMBT-1 scans, before and after receiving 5 mg of selegiline twice daily for 5 d. To compare selectivity, 18F-THK5351 studies were also conducted before and after selegiline. Amyloid-ß burden was expressed in centiloids. 18F-SMBT-1 outcomes were expressed as SUV, as well as tissue ratios and binding parameters using the subcortical white matter as a reference region. Results: 18F-SMBT-1 showed robust entry into the brain and reversible binding kinetics, with high tracer retention in basal ganglia, intermediate retention in cortical regions, and the lowest retention in cerebellum and white matter, which tightly follows the known regional brain distribution of MAO-B (R 2 = 0.84). More than 85% of 18F-SMBT-1 signal was blocked by selegiline across the brain, and in contrast to 18F-THK5351, no residual cortical activity was observed after the selegiline regimen, indicating high selectivity for MAO-B and low nonspecific binding. 18F-SMBT-1 also captured the known MAO-B increases with age, with an annual rate of change (∼2.6%/y) similar to the in vitro rates of change (∼1.9%/y). Quantitative and semiquantitative measures of 18F-SMBT-1 binding were strongly associated (R 2 > 0.94), suggesting that a simplified tissue-ratio approach could be used to generate outcome measures. Conclusion: 18F-SMBT-1 is a highly selective MAO-B tracer, with low nonspecific binding, high entry into the brain, and reversible kinetics. Moreover, 18F-SMBT-1 brain distribution matches the reported in vitro distribution and captures the known MAO-B increases with age, suggesting that 18F-SMBT-1 can potentially be used as a surrogate marker of reactive astrogliosis. Further validation of these findings with 18F-SMBT-1 will require examination of a much larger series, including participants with mild cognitive impairment and AD.


Alzheimer Disease , Quinolines , Aged , Alzheimer Disease/metabolism , Aminopyridines , Amyloid beta-Peptides , Gliosis , Humans , Monoamine Oxidase/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/metabolism , Selegiline
13.
Front Aging Neurosci ; 13: 761010, 2021.
Article En | MEDLINE | ID: mdl-34912209

Introduction: We aimed to determine whether in vivo tau deposits and monoamine oxidase B (MAO-B) detection using 18F-THK5351 positron emission tomography (PET) can assist in the differential distribution in patients with corticobasal syndrome (CBS), progressive supranuclear palsy (PSP), and Alzheimer's disease (AD) and whether 18F-THK5351 retention of lesion sites in CBS and PSP can correlate with clinical parameters. Methods: 18F-THK5351 PET was performed in 35 participants, including 7, 9, and 10 patients with CBS, PSP, and AD, respectively, and 9 age-matched normal controls. In CBS and PSP, cognitive and motor functions were assessed using the Montreal Cognitive Assessment, Addenbrooke's Cognitive Examination-Revised, and Frontal Assessment Battery, Unified Parkinson's Disease Rating Scale Motor Score, and PSP Rating Scale. Results: 18F-THK5351 retention was observed in sites susceptible to disease-related pathologies in CBS, PSP, and AD. 18F-THK5351 uptake in the precentral gyrus clearly differentiated patients with CBS from those with PSP and AD. Furthermore, 18F-THK5351 uptake in the inferior temporal gyrus clearly differentiated patients with AD from those with CBS and PSP. Regional 18F-THK5351 retention was associated with the cognitive function in CBS and PSP. Conclusion: Measurement of the tau deposits and MAO-B density in the brain using 18F-THK5351 may be helpful for the differential diagnosis of tauopathies and for understanding disease stages.

14.
Sci Rep ; 11(1): 2588, 2021 01 28.
Article En | MEDLINE | ID: mdl-33510301

Brain positron emission tomography (PET) imaging with radiolabelled proteins is an emerging concept that potentially enables visualization of unique molecular targets in the brain. However, the pharmacokinetics and protein radiolabelling methods remain challenging. Here, we report the performance of an engineered, blood-brain barrier (BBB)-permeable affibody molecule that exhibits rapid clearance from the brain, which was radiolabelled using a unique fluorine-18 labelling method, a cell-free protein radiosynthesis (CFPRS) system. AS69, a small (14 kDa) dimeric affibody molecule that binds to the monomeric and oligomeric states of α-synuclein, was newly designed for brain delivery with an apolipoprotein E (ApoE)-derived brain shuttle peptide as AS69-ApoE (22 kDa). The radiolabelled products 18F-AS69 and 18F-AS69-ApoE were successfully synthesised using the CFPRS system. Notably, 18F-AS69-ApoE showed higher BBB permeability than 18F-AS69 in an ex vivo study at 10 and 30 min post injection and was partially cleared from the brain at 120 min post injection. These results suggest that small, a brain shuttle peptide-fused fluorine-18 labelled protein binders can potentially be utilised for brain molecular imaging.


Apolipoproteins E/metabolism , Fluorine Radioisotopes/analysis , Peptides/chemistry , Recombinant Fusion Proteins/chemical synthesis , Animals , Apolipoproteins E/genetics , Humans , Male , Pharmacokinetics , Positron-Emission Tomography , Recombinant Fusion Proteins/chemistry
15.
J Nucl Med ; 62(2): 253-258, 2021 02.
Article En | MEDLINE | ID: mdl-32646880

Reactive astrocytes play a key role in the pathogenesis of various neurodegenerative diseases. Monoamine oxidase-B (MAO-B) is one of the promising targets for the imaging of astrogliosis in the human brain. A novel selective and reversible MAO-B tracer, (S)-(2-methylpyrid-5-yl)-6-[(3-18F-fluoro-2-hydroxy)propoxy]quinoline (18F-SMBT-1), was successfully developed via lead optimization from the first-generation tau PET tracer 18F-THK-5351. Methods: SMBT-1 was radiolabeled with 18F using the corresponding precursor. The binding affinity of radiolabeled compounds to MAO-B was assessed using saturation and competitive binding assays. The binding selectivity of 18F-SMBT-1 to MAO-B was evaluated by autoradiography of frozen human brain tissues. The pharmacokinetics and metabolism were assessed in normal mice after intravenous administration of 18F-SMBT-1. A 14-d toxicity study after the intravenous administration of 18F-SMBT-1 was performed using rats and mice. Results: In vitro binding assays demonstrated a high binding affinity of 18F-SMBT-1 to MAO-B (dissociation constant, 3.7 nM). In contrast, it showed low binding affinity to MAO-A and protein aggregates such as amyloid-ß and tau fibrils. Autoradiographic analysis showed higher amounts of 18F-SMBT-1 binding in the Alzheimer disease brain sections than in the control brain sections. 18F-SMBT-1 binding was completely displaced with the reversible MAO-B inhibitor lazabemide, demonstrating the high selectivity of 18F-SMBT-1 for MAO-B. Furthermore, 18F-SMBT-1 showed a high uptake by brain, rapid washout, and no radiolabeled metabolites in the brain of normal mice. 18F-SMBT-1 showed no significant binding to various receptors, ion channels, or transporters, and no toxic effects related to its administration were observed in mice and rats. Conclusion:18F-SMBT-1 is a promising and selective MAO-B PET tracer candidate, which would be useful for quantitative monitoring of astrogliosis in the human brain.


Fluorine Radioisotopes/chemistry , Monoamine Oxidase/metabolism , Positron-Emission Tomography/methods , Animals , Brain/diagnostic imaging , Brain/metabolism , Humans , Mice , Radioactive Tracers , Tissue Distribution
16.
Nucl Med Biol ; 93: 11-18, 2021 02.
Article En | MEDLINE | ID: mdl-33221641

INTRODUCTION: [18F]THK-5351 was originally developed as a positron emission tomography (PET) imaging tracer for the detection of accumulated tau proteins, the pathological hallmark of Alzheimer's disease (AD). However, clinical studies of [18F]THK-5351 revealed the existence of off-target binding to monoamine oxidase-B (MAO-B). To overcome this off-target binding, in this work, we synthesized and evaluated 2-pyrrolopyridinylquinoline (PPQ) derivatives as selective tau PET imaging tracers. METHODS: The core structure of PPQ derivatives was synthesized mainly using the Buchwald-Hartwig amination coupling reaction. All derivatives were evaluated for binding affinity towards tau and MAO-B by in vitro competitive binding assay. Radiosynthesis of PPQ derivatives was performed by 18F-radiolabeling of their tosylate precursors with activated [18F]KF/Kryptofix222 complex in dimethylsulfoxide by heating at 110 °C for 10 min. The biological properties of these [18F]PPQ derivatives were characterized by in vitro autoradiography of postmortem AD brain sections and by assay of ex vivo biodistribution in mice. RESULTS: The PPQ derivatives were synthesized, with yields of 49-84%. In vitro competitive binding assay revealed that two novel PPQ derivatives-PPQ8 and PPQ9-demonstrated high binding affinity for tau (IC50 = 4.9 and 6.9 nM, respectively). The radiosynthesis of [18F]PPQ8 and [18F]PPQ9 yielded 1.4% and 50.1% isolated non-decay corrected radiochemical yield, respectively, with >99% radiochemical purity. The molar radioactivities of [18F]PPQ8 and [18F]PPQ9 were 16.9 and 64.8 GBq/µmol, respectively. The in vitro and ex vivo biological characterization of [18F]PPQ8 and [18F]PPQ9 revealed that these tracers were selective for tau in AD brain sections without off-target binding, and they furthermore demonstrated brain uptake in normal mice. CONCLUSIONS: 18F-labeled PPQ derivatives improved binding affinity and selectivity for tau aggregates in AD. Further structural optimization to improve pharmacokinetics for potent tau PET imaging tracers is required.


Alzheimer Disease/diagnostic imaging , Positron-Emission Tomography/methods , Quinolines/chemistry , Quinolines/chemical synthesis , tau Proteins/metabolism , Animals , Chemistry Techniques, Synthetic , Mice , Quinolines/pharmacokinetics , Tissue Distribution
17.
Appl Radiat Isot ; 166: 109361, 2020 Dec.
Article En | MEDLINE | ID: mdl-32877862

When applied to a radiosynthesis, a microscale approach can help to save precursor and improve yields. Thus, a 5-10 µL microscale method based on a concentration procedure was developed and applied to the radiosynthesis of [18F]FET and [18F]fallypride. In spite of using an amount of precursor ca. 100 times smaller, radiochemical yields were comparable or even higher than those reported in literature. Because of the very low reaction volumes, the possible effects of concentrated dose of activity and carrier fluoride were also investigated.

18.
Nihon Yakurigaku Zasshi ; 155(3): 159-163, 2020.
Article Ja | MEDLINE | ID: mdl-32378635

Positron emission tomography (PET) is a molecular imaging technique that visualizes pathophysiology in the body using radiotracers at tracer doses (~µg). PET would provide the information regarding not only pharmacokinetics of radiolabeled compounds, but also target engagements, patient selection, and biomarkers. Previously small molecules are widely used as radiotracers, but recently biopharmaceuticals are launched, providing a novel type radiotracer. In general, antibodies are radiolabeled by long physiological half-lives radionuclides such as Cu-64 and Zr-89 because of their slow pharmacokinetics. However, shorter half-lives radiolabeled tracers (C-11 and F-18) might be suitable on the point view of radiation. Now small protein ligands such as affibodies (~7 kDa) are developed as a radiotracer. We are trying to develop a novel approach to label proteins for PET imaging, which are based on radiolabeled amino acids and cell-free protein synthesis system. In this review, we introduced the topics of protein-based PET tracers.


Biological Products , Positron-Emission Tomography , Proteins/chemistry , Radioisotopes/chemistry , Copper Radioisotopes , Humans , Zirconium
19.
J Pharmacol Exp Ther ; 373(3): 361-369, 2020 06.
Article En | MEDLINE | ID: mdl-32217770

The orally available and novel small molecule ONO-7579 (N-{2-[4-(2-amino-5-chloropyridin-3-yl)phenoxy]pyrimidin-5-yl}-N'-[2-(methanesulfonyl)-5-(trifluoromethyl)phenyl]urea) is a highly potent and selective pan-tropomyosin receptor kinase (TRK) inhibitor. The objective of the present study was to characterize the pharmacokinetic (PK), pharmacodynamic (PD), and antitumor efficacy relationships of ONO-7579 in mice xenografted with a human colorectal cancer cell line, KM12 (harboring the tropomyosin 3 (TPM3) -neurotrophic tyrosine receptor kinase 1 fusion gene), via a PK/PD modeling approach. Plasma and tumor concentrations of ONO-7579, tumor levels of phosphorylated TPM3-TRKA (pTRKA), and tumor volumes in the murine model were measured with a single or multiple dose of ONO-7579 (0.06-0.60 mg/kg) administered once daily. The PK/PD/efficacy models were developed in a sequential manner. Changes in plasma concentrations of ONO-7579 were described with an oral one-compartment model. Tumor concentrations of ONO-7579 were higher than plasma concentrations, and changes in ONO-7579 tumor concentrations were described with an additional tumor compartment that had no influence on plasma concentrations. pTRKA in tumors was described with a direct Emax model, and the tumor ONO-7579 concentration causing 50% of the maximum effect was estimated to be 17.6 ng/g. In addition, a pTRKA-driven tumor growth inhibition model indicated that ONO-7579 started to sharply increase the antitumor effect at pTRKA inhibition rates >60% and required >91.5% to reduce tumors. In conclusion, the developed PK/PD/efficacy models revealed a "switch-like" relationship between pTRKA inhibition rate and antitumor effect in a murine KM12 xenograft model, demonstrating that pTRKA in tumors could serve as an effective biomarker for scheduling the dose regimen in early-stage clinical studies. SIGNIFICANCE STATEMENT: In recent years, clinical development of TRK inhibitors in patients with neurotrophic tyrosine receptor kinase fusion-positive solid tumors has been accelerated. This research found that phosphorylated TRKA was a useful biomarker for explaining the antitumor efficacy of TRK inhibitors using a pharmacokinetic/pharmacodynamic modeling approach in xenograft mice. This finding suggests a rational dosing regimen in early-stage clinical studies for ONO-7579 (N-{2-[4-(2-amino-5-chloropyridin-3-yl)phenoxy]pyrimidin-5-yl}-N'-[2-(methanesulfonyl)-5-(trifluoromethyl)phenyl]urea), a novel pan-TRK inhibitor.


Organic Chemicals/pharmacology , Organic Chemicals/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Disease Models, Animal , Female , Heterografts/drug effects , Heterografts/metabolism , Humans , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Xenograft Model Antitumor Assays/methods
20.
Pharmacol Res Perspect ; 7(4): e00499, 2019 08.
Article En | MEDLINE | ID: mdl-31338198

Some histamine H1 receptor (H1R) antagonists induce adverse sedative reactions caused by blockade of histamine transmission in the brain. Desloratadine is a second-generation antihistamine for treatment of allergic disorders. Its binding to brain H1Rs, which is the basis of sedative property of antihistamines, has not been examined previously in the human brain by positron emission tomography (PET). We examined brain H1R binding potential ratio (BPR), H1R occupancy (H1RO), and subjective sleepiness after oral desloratadine administration in comparison to loratadine. Eight healthy male volunteers underwent PET imaging with [11C]-doxepin, a PET tracer for H1Rs, after a single oral administration of desloratadine (5 mg), loratadine (10 mg), or placebo in a double-blind crossover study. BPR and H1RO in the cerebral cortex were calculated, and plasma concentrations of loratadine and desloratadine were measured. Subjective sleepiness was quantified by the Line Analogue Rating Scale (LARS) and the Stanford Sleepiness Scale (SSS). BPR was significantly lower after loratadine administration than after placebo (0.504 ± 0.074 vs 0.584 ± 0.059 [mean ± SD], P < 0.05), but BPR after desloratadine administration was not significantly different from BPR after placebo (0.546 ± 0.084 vs 0.584 ± 0.059, P = 0.250). The plasma concentration of loratadine was negatively correlated with BPR in subjects receiving loratadine, but that of desloratadine was not correlated with BPR. Brain H1ROs after desloratadine and loratadine administration were 6.47 ± 10.5% and 13.8 ± 7.00%, respectively (P = 0.103). Subjective sleepiness did not significantly differ among subjects receiving the two antihistamines and placebo. At therapeutic doses, desloratadine did not bind significantly to brain H1Rs and did not induce any significant sedation.


Histamine H1 Antagonists, Non-Sedating/administration & dosage , Loratadine/analogs & derivatives , Loratadine/administration & dosage , Receptors, Histamine H1/metabolism , Adult , Brain/diagnostic imaging , Brain/metabolism , Cross-Over Studies , Double-Blind Method , Healthy Volunteers , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Humans , Loratadine/pharmacokinetics , Male , Positron-Emission Tomography/methods , Sleepiness , Young Adult
...