Skip to main content
Erschienen in: Molecular Imaging and Biology 3/2018

01.06.2018 | Brief Article

MAO-B Inhibitors Do Not Block In Vivo Flortaucipir([18F]-AV-1451) Binding

verfasst von: Allan K. Hansen, David J. Brooks, Per Borghammer

Erschienen in: Molecular Imaging and Biology | Ausgabe 3/2018

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Recent evidence suggests that the tau radiotracer [18F]THK-5351 displays high affinity for the monoamine oxidase type B (MAO-B) enzyme. Utilizing another tau-tracer, flortaucipir ([18F]AV-1451), we previously reported that non-demented Parkinson’s disease patients show off-target binding in subcortical structures, but no appreciable cortical uptake. However, 59 % of these patients were receiving MAO-B inhibitors at the time of their scan. Here, we retrospectively investigated if MAO-B inhibitors in clinical doses affect flortaucipir binding.

Procedures

We compared the standard uptake values of flortaucipir at regional and voxel levels in Parkinson’s disease patients who received MAO-B inhibitors with those who did not.

Results

Sixteen of 27 Parkinson’s disease patients received MAO-B inhibitors at the time of scan. We found no significant flortaucipir uptake differences between the groups at voxel or regional levels.

Conclusion

Use of MAO-B inhibitors at pharmaceutical levels did not significantly affect flortaucipir binding. Thus, MAO-B does not appear to be a significant binding target of flortaucipir.
Literatur
1.
Zurück zum Zitat Johnson KA, Schultz A, Betensky RA et al (2016) Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann Neurol 79:110–119PubMedCrossRef Johnson KA, Schultz A, Betensky RA et al (2016) Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann Neurol 79:110–119PubMedCrossRef
3.
Zurück zum Zitat Cho H, Choi JY, Hwang MS et al (2016) Tau PET in Alzheimer disease and mild cognitive impairment. Neurology 87:375–383PubMedCrossRef Cho H, Choi JY, Hwang MS et al (2016) Tau PET in Alzheimer disease and mild cognitive impairment. Neurology 87:375–383PubMedCrossRef
4.
Zurück zum Zitat Pontecorvo MJ, Devous MD, Navitsky M et al (2017) Relationships between flortaucipir PET tau binding and amyloid burden, clinical diagnosis, age and cognition. Brain 140:748–763PubMedPubMedCentral Pontecorvo MJ, Devous MD, Navitsky M et al (2017) Relationships between flortaucipir PET tau binding and amyloid burden, clinical diagnosis, age and cognition. Brain 140:748–763PubMedPubMedCentral
5.
Zurück zum Zitat Hansen AK, Knudsen K, Lillethorup TP et al (2016) In vivo imaging of neuromelanin in Parkinson’s disease using 18F-AV-1451 PET. Brain 139:2039–2049PubMedCrossRef Hansen AK, Knudsen K, Lillethorup TP et al (2016) In vivo imaging of neuromelanin in Parkinson’s disease using 18F-AV-1451 PET. Brain 139:2039–2049PubMedCrossRef
6.
Zurück zum Zitat Passamonti L, Vázquez Rodríguez P, Hong YT et al (2017) 18F-AV-1451 positron emission tomography in Alzheimer’s disease and progressive supranuclear palsy. Brain 140:781–791PubMedPubMedCentral Passamonti L, Vázquez Rodríguez P, Hong YT et al (2017) 18F-AV-1451 positron emission tomography in Alzheimer’s disease and progressive supranuclear palsy. Brain 140:781–791PubMedPubMedCentral
7.
Zurück zum Zitat Marquié M, Normandin MD, Vanderburg CR et al (2015) Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann Neurol 78:787–800PubMedPubMedCentralCrossRef Marquié M, Normandin MD, Vanderburg CR et al (2015) Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann Neurol 78:787–800PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Ng KP, Pascoal TA, Mathotaarachchi S et al (2017) Monoamine oxidase B inhibitor, selegiline, reduces 18F-THK5351 uptake in the human brain. Alzheimers Res Ther 9:25PubMedPubMedCentralCrossRef Ng KP, Pascoal TA, Mathotaarachchi S et al (2017) Monoamine oxidase B inhibitor, selegiline, reduces 18F-THK5351 uptake in the human brain. Alzheimers Res Ther 9:25PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Carter SF, Scholl M, Almkvist O et al (2012) Evidence for Astrocytosis in prodromal Alzheimer disease provided by 11C-deuterium-L-Deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18F-FDG. J Nucl Med 53:37–46PubMedCrossRef Carter SF, Scholl M, Almkvist O et al (2012) Evidence for Astrocytosis in prodromal Alzheimer disease provided by 11C-deuterium-L-Deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18F-FDG. J Nucl Med 53:37–46PubMedCrossRef
10.
Zurück zum Zitat Tong J, Meyer JH, Furukawa Y et al (2013) Distribution of monoamine oxidase proteins in human brain: implications for brain imaging studies. J Cereb Blood Flow Metab 33:863–871PubMedPubMedCentralCrossRef Tong J, Meyer JH, Furukawa Y et al (2013) Distribution of monoamine oxidase proteins in human brain: implications for brain imaging studies. J Cereb Blood Flow Metab 33:863–871PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Hansen AK, Damholdt MF, Fedorova TD et al (2017) In vivo cortical tau in Parkinson’s disease using 18F-AV-1451 positron emission tomography. Mov Disord 32:922–927PubMedCrossRef Hansen AK, Damholdt MF, Fedorova TD et al (2017) In vivo cortical tau in Parkinson’s disease using 18F-AV-1451 positron emission tomography. Mov Disord 32:922–927PubMedCrossRef
12.
Zurück zum Zitat Xia CF, Arteaga J, Chen G et al (2013) [(18)F]T807, a novel tau positron emission tomography imaging agent for Alzheimer’s disease. Alzheimers Dement 9:666–676PubMedCrossRef Xia CF, Arteaga J, Chen G et al (2013) [(18)F]T807, a novel tau positron emission tomography imaging agent for Alzheimer’s disease. Alzheimers Dement 9:666–676PubMedCrossRef
13.
Zurück zum Zitat Vermeiren C, Mercier J, Viot D et al (2015) T807, a reported selective tau tracer, binds with nanomolar affinity to monoamine oxidase a. Alzheimers Dement 11:P283CrossRef Vermeiren C, Mercier J, Viot D et al (2015) T807, a reported selective tau tracer, binds with nanomolar affinity to monoamine oxidase a. Alzheimers Dement 11:P283CrossRef
15.
Zurück zum Zitat Marquié M, Normandin MD, Meltzer AC et al (2017) Pathological correlations of [F-18]-AV-1451 imaging in non-alzheimer tauopathies. Ann Neurol 81:117–128PubMedPubMedCentralCrossRef Marquié M, Normandin MD, Meltzer AC et al (2017) Pathological correlations of [F-18]-AV-1451 imaging in non-alzheimer tauopathies. Ann Neurol 81:117–128PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Smith R, Schöll M, Honer M et al (2017) Tau neuropathology correlates with FDG-PET, but not AV-1451-PET, in progressive supranuclear palsy. Acta Neuropathol 133:149–151PubMedCrossRef Smith R, Schöll M, Honer M et al (2017) Tau neuropathology correlates with FDG-PET, but not AV-1451-PET, in progressive supranuclear palsy. Acta Neuropathol 133:149–151PubMedCrossRef
17.
Zurück zum Zitat Nag S, Fazio P, Lehmann L et al (2016) In vivo and in vitro characterization of a novel MAO-B inhibitor Radioligand, 18F-labeled deuterated Fluorodeprenyl. J Nucl Med 57:315–320PubMedCrossRef Nag S, Fazio P, Lehmann L et al (2016) In vivo and in vitro characterization of a novel MAO-B inhibitor Radioligand, 18F-labeled deuterated Fluorodeprenyl. J Nucl Med 57:315–320PubMedCrossRef
18.
Zurück zum Zitat Lockhart SN, Baker SL, Okamura N et al (2016) Dynamic PET measures of tau accumulation in cognitively normal older adults and Alzheimer’s disease patients measured using [18F] THK-5351. PLoS One 11:1–16CrossRef Lockhart SN, Baker SL, Okamura N et al (2016) Dynamic PET measures of tau accumulation in cognitively normal older adults and Alzheimer’s disease patients measured using [18F] THK-5351. PLoS One 11:1–16CrossRef
19.
Zurück zum Zitat Ikonomovic MD, Abrahamson EE, Price JC et al (2016) [F-18]AV-1451 positron emission tomography retention in choroid plexus: more than “off-target” binding. Ann Neurol 80:307–308PubMedPubMedCentralCrossRef Ikonomovic MD, Abrahamson EE, Price JC et al (2016) [F-18]AV-1451 positron emission tomography retention in choroid plexus: more than “off-target” binding. Ann Neurol 80:307–308PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Cho H, Choi JY, Hwang MS et al (2017) Subcortical 18F-AV-1451 binding patterns in progressive supranuclear palsy. Mov Disord 32:134–140PubMedCrossRef Cho H, Choi JY, Hwang MS et al (2017) Subcortical 18F-AV-1451 binding patterns in progressive supranuclear palsy. Mov Disord 32:134–140PubMedCrossRef
21.
Zurück zum Zitat Whitwell JL, Lowe VJ, Tosakulwong N et al (2017) [18F]AV-1451 tau positron emission tomography in progressive supranuclear palsy. Mov Disord 32:124–133PubMedCrossRef Whitwell JL, Lowe VJ, Tosakulwong N et al (2017) [18F]AV-1451 tau positron emission tomography in progressive supranuclear palsy. Mov Disord 32:124–133PubMedCrossRef
22.
Zurück zum Zitat Smith R, Schain M, Nilsson C et al (2017) Increased basal ganglia binding of (18) F-AV-1451 in patients with progressive supranuclear palsy. Mov Disord 32:108–114PubMedCrossRef Smith R, Schain M, Nilsson C et al (2017) Increased basal ganglia binding of (18) F-AV-1451 in patients with progressive supranuclear palsy. Mov Disord 32:108–114PubMedCrossRef
23.
Zurück zum Zitat Cho H, Choi JY, Lee SH et al (2017) 18F-AV-1451 binds to putamen in multiple system atrophy. Mov Disord 32:171–173PubMedCrossRef Cho H, Choi JY, Lee SH et al (2017) 18F-AV-1451 binds to putamen in multiple system atrophy. Mov Disord 32:171–173PubMedCrossRef
24.
Zurück zum Zitat Shcherbinin S, Schwarz AJ, Joshi A et al (2016) Kinetics of the tau PET tracer 18F-AV-1451 (T807) in subjects with normal cognitive function, mild cognitive impairment, and Alzheimer disease. J Nucl Med 57:1535–1542PubMedCrossRef Shcherbinin S, Schwarz AJ, Joshi A et al (2016) Kinetics of the tau PET tracer 18F-AV-1451 (T807) in subjects with normal cognitive function, mild cognitive impairment, and Alzheimer disease. J Nucl Med 57:1535–1542PubMedCrossRef
25.
Zurück zum Zitat Zecca L, Fariello R, Riederer P et al (2002) The absolute concentration of nigral neuromelanin, assayed by a new sensitive method, increases throughout the life and is dramatically decreased in Parkinson’s disease. FEBS Lett 510:216–220PubMedCrossRef Zecca L, Fariello R, Riederer P et al (2002) The absolute concentration of nigral neuromelanin, assayed by a new sensitive method, increases throughout the life and is dramatically decreased in Parkinson’s disease. FEBS Lett 510:216–220PubMedCrossRef
26.
Zurück zum Zitat Zecca L, Bellei C, Costi P et al (2008) New melanic pigments in the human brain that accumulate in aging and block environmental toxic metals. Proc Natl Acad Sci U S A 105:17567–17572PubMedPubMedCentralCrossRef Zecca L, Bellei C, Costi P et al (2008) New melanic pigments in the human brain that accumulate in aging and block environmental toxic metals. Proc Natl Acad Sci U S A 105:17567–17572PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Fowler JS, Logan J, Wang G-J, Volkow ND (2003) Monoamine oxidase and cigarette smoking. Neurotoxicology 24:75–82PubMedCrossRef Fowler JS, Logan J, Wang G-J, Volkow ND (2003) Monoamine oxidase and cigarette smoking. Neurotoxicology 24:75–82PubMedCrossRef
28.
Zurück zum Zitat Nakamura S, Kawamata T, Akiguchi I et al (1990) Expression of monoamine oxidase B activity in astrocytes of senile plaques. Acta Neuropathol 80:419–425PubMedCrossRef Nakamura S, Kawamata T, Akiguchi I et al (1990) Expression of monoamine oxidase B activity in astrocytes of senile plaques. Acta Neuropathol 80:419–425PubMedCrossRef
29.
30.
Zurück zum Zitat Fowler JS, MacGregor RR, Wolf AP et al (1987) Mapping human brain monoamine oxidase A and B with 11C-labeled suicide inactivators and PET. Science 235:481–485PubMedCrossRef Fowler JS, MacGregor RR, Wolf AP et al (1987) Mapping human brain monoamine oxidase A and B with 11C-labeled suicide inactivators and PET. Science 235:481–485PubMedCrossRef
31.
Zurück zum Zitat Jensen SB, Olsen AK, Pedersen K, Cumming P (2006) Effect of monoamine oxidase inhibition on amphetamine-evoked changes in dopamine receptor availability in the living pig: a dual tracer PET study with [11C]harmine and [11C]raclopride. Synapse 59:427–434PubMedCrossRef Jensen SB, Olsen AK, Pedersen K, Cumming P (2006) Effect of monoamine oxidase inhibition on amphetamine-evoked changes in dopamine receptor availability in the living pig: a dual tracer PET study with [11C]harmine and [11C]raclopride. Synapse 59:427–434PubMedCrossRef
32.
Zurück zum Zitat Ginovart N, Meyer JH, Boovariwala A et al (2006) Positron emission tomography quantification of [11C]-harmine binding to monoamine oxidase-A in the human brain. J Cereb Blood Flow Metab 26:330–344PubMedCrossRef Ginovart N, Meyer JH, Boovariwala A et al (2006) Positron emission tomography quantification of [11C]-harmine binding to monoamine oxidase-A in the human brain. J Cereb Blood Flow Metab 26:330–344PubMedCrossRef
33.
Zurück zum Zitat Fowler JS, Volkow ND, Logan J et al (1994) Slow recovery of human brain MAO B after L-deprenyl (Selegeline) withdrawal. Synapse 18:86–93PubMedCrossRef Fowler JS, Volkow ND, Logan J et al (1994) Slow recovery of human brain MAO B after L-deprenyl (Selegeline) withdrawal. Synapse 18:86–93PubMedCrossRef
34.
Zurück zum Zitat Arnett CD, Fowler JS, MacGregor RR et al (1987) Turnover of brain monoamine oxidase measured in vivo by positron emission tomography using L-[11C]deprenyl. J Neurochem 49:522–527PubMedCrossRef Arnett CD, Fowler JS, MacGregor RR et al (1987) Turnover of brain monoamine oxidase measured in vivo by positron emission tomography using L-[11C]deprenyl. J Neurochem 49:522–527PubMedCrossRef
35.
Zurück zum Zitat Lemoine L, Saint-Aubert L, Nennesmo I et al (2017) Cortical laminar tau deposits and activated astrocytes in Alzheimer’s disease visualised by 3H-THK5117 and 3H-deprenyl autoradiography. Sci Rep 7:45496PubMedPubMedCentralCrossRef Lemoine L, Saint-Aubert L, Nennesmo I et al (2017) Cortical laminar tau deposits and activated astrocytes in Alzheimer’s disease visualised by 3H-THK5117 and 3H-deprenyl autoradiography. Sci Rep 7:45496PubMedPubMedCentralCrossRef
Metadaten
Titel
MAO-B Inhibitors Do Not Block In Vivo Flortaucipir([18F]-AV-1451) Binding
verfasst von
Allan K. Hansen
David J. Brooks
Per Borghammer
Publikationsdatum
01.06.2018
Verlag
Springer International Publishing
Erschienen in
Molecular Imaging and Biology / Ausgabe 3/2018
Print ISSN: 1536-1632
Elektronische ISSN: 1860-2002
DOI
https://doi.org/10.1007/s11307-017-1143-1

Weitere Artikel der Ausgabe 3/2018

Molecular Imaging and Biology 3/2018 Zur Ausgabe

Update Radiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.