Skip to main content
Top
Published in: Alzheimer's Research & Therapy 1/2017

Open Access 01-12-2017 | Research

Monoamine oxidase B inhibitor, selegiline, reduces 18F-THK5351 uptake in the human brain

Authors: Kok Pin Ng, Tharick A. Pascoal, Sulantha Mathotaarachchi, Joseph Therriault, Min Su Kang, Monica Shin, Marie-Christine Guiot, Qi Guo, Ryuichi Harada, Robert A. Comley, Gassan Massarweh, Jean-Paul Soucy, Nobuyuki Okamura, Serge Gauthier, Pedro Rosa-Neto

Published in: Alzheimer's Research & Therapy | Issue 1/2017

Login to get access

Abstract

Background

18F-THK5351 is a quinoline-derived tau imaging agent with high affinity to paired helical filaments (PHF). However, high levels of 18F-THK5351 retention in brain regions thought to contain negligible concentrations of PHF raise questions about the interpretation of the positron emission tomography (PET) signals, particularly given previously described interactions between quinolone derivatives and monoamine oxidase B (MAO-B). Here, we tested the effects of MAO-B inhibition on 18F-THK5351 brain uptake using PET and autoradiography.

Methods

Eight participants (five mild cognitive impairment, two Alzheimer’s disease, and one progressive supranuclear palsy) had baseline 18F-AZD4694 and 18F-THK5351 scans in order to quantify brain amyloid and PHF load, respectively. A second 18F-THK5351 scan was conducted 1 week later, 1 h after a 10-mg oral dose of selegiline. Three out of eight patients also had a third 18F-THK5351 scan 9–28 days after the selegiline administration. The primary outcome measure was standardized uptake value (SUV), calculated using tissue radioactivity concentration from 50 to 70 min after 18F-THK5351 injection, normalizing for body weight and injected radioactivity. The SUV ratio (SUVR) was determined using the cerebellar cortex as the reference region. 18F-THK5351 competition autoradiography studies in postmortem tissue were conducted using 150 and 500 nM selegiline.

Results

At baseline, 18F-THK5351 SUVs were highest in the basal ganglia (0.64 ± 0.11) and thalamus (0.62 ± 0.14). In the post-selegiline scans, the regional SUVs were reduced on average by 36.7% to 51.8%, with the greatest reduction noted in the thalamus (51.8%) and basal ganglia (51.4%). MAO-B inhibition also reduced 18F-THK5351 SUVs in the cerebellar cortex (41.6%). The SUVs remained reduced in the three patients imaged at 9–28 days. Tissue autoradiography confirmed the effects of MAO-B inhibition on 18F-THK5351 uptake.

Conclusions

These results indicate that the interpretation of 18F-THK5351 PET images, with respect to tau, is confounded by the high MAO-B availability across the entire brain. In addition, the heterogeneous MAO-B availability across the cortex may limit the interpretation of 18F-THK5351 scans using reference region methods.
Literature
1.
go back to reference Villemagne VL, Fodero-Tavoletti MT, Masters CL, Rowe CC. Tau imaging: early progress and future directions. Lancet Neurol. 2015;14(1):114–24.CrossRefPubMed Villemagne VL, Fodero-Tavoletti MT, Masters CL, Rowe CC. Tau imaging: early progress and future directions. Lancet Neurol. 2015;14(1):114–24.CrossRefPubMed
2.
go back to reference Johnson KA, Schultz A, Betensky RA, Becker JA, Sepulcre J, Rentz D, et al. Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann Neurol. 2016;79:110–9.CrossRefPubMed Johnson KA, Schultz A, Betensky RA, Becker JA, Sepulcre J, Rentz D, et al. Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann Neurol. 2016;79:110–9.CrossRefPubMed
3.
go back to reference Harada R, Okamura N, Furumoto S, Furukawa K, Ishiki A, Tomita N, et al. 18F-THK5351: a novel PET radiotracer for imaging neurofibrillary pathology in Alzheimer’s disease. J Nucl Med. 2015;57:1–43. Harada R, Okamura N, Furumoto S, Furukawa K, Ishiki A, Tomita N, et al. 18F-THK5351: a novel PET radiotracer for imaging neurofibrillary pathology in Alzheimer’s disease. J Nucl Med. 2015;57:1–43.
4.
go back to reference Fowler JS, MacGregor RR, Wolf AP, Arnett CD, Dewey SL, Schlyer D, et al. Mapping human brain monoamine oxidase A and B with 11C-labeled suicide inactivators and PET. Science. 1987;235:481–5.CrossRefPubMed Fowler JS, MacGregor RR, Wolf AP, Arnett CD, Dewey SL, Schlyer D, et al. Mapping human brain monoamine oxidase A and B with 11C-labeled suicide inactivators and PET. Science. 1987;235:481–5.CrossRefPubMed
5.
go back to reference Tong J, Meyer JH, Furukawa Y, Boileau I, Chang L-J, Wilson AA, et al. Distribution of monoamine oxidase proteins in human brain: implications for brain imaging studies. J Cereb Blood Flow Metab. 2013;33:863–71.CrossRefPubMedPubMedCentral Tong J, Meyer JH, Furukawa Y, Boileau I, Chang L-J, Wilson AA, et al. Distribution of monoamine oxidase proteins in human brain: implications for brain imaging studies. J Cereb Blood Flow Metab. 2013;33:863–71.CrossRefPubMedPubMedCentral
6.
go back to reference Lee MK, Hwang BY, Lee SA, Oh GJ, Choi WH, Hong SS, et al. 1-methyl-2-undecyl-4(1H)-quinolone as an irreversible and selective inhibitor of type B monoamine oxidase. Chem Pharm Bull (Tokyo). 2003;51:409–11.CrossRef Lee MK, Hwang BY, Lee SA, Oh GJ, Choi WH, Hong SS, et al. 1-methyl-2-undecyl-4(1H)-quinolone as an irreversible and selective inhibitor of type B monoamine oxidase. Chem Pharm Bull (Tokyo). 2003;51:409–11.CrossRef
7.
go back to reference Mahmood I. Clinical pharmacokinetics and pharmacodynamics of selegiline. An update. Clin Pharmacokinet. 1997;33:91–102.CrossRefPubMed Mahmood I. Clinical pharmacokinetics and pharmacodynamics of selegiline. An update. Clin Pharmacokinet. 1997;33:91–102.CrossRefPubMed
8.
go back to reference Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12:189–98.CrossRefPubMed Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12:189–98.CrossRefPubMed
9.
go back to reference Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, Collin I, et al. The Montreal Cognitive Assessment, MoCA: A Brief Screening Tool For Mild Cognitive Impairment. J. Am. Geriatr. Soc. Blackwell Science Inc; 2005;53:695–9. Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, Collin I, et al. The Montreal Cognitive Assessment, MoCA: A Brief Screening Tool For Mild Cognitive Impairment. J. Am. Geriatr. Soc. Blackwell Science Inc; 2005;53:695–9.  
10.
11.
go back to reference McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: Report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 2011;77:333.CrossRef McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: Report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 2011;77:333.CrossRef
12.
go back to reference Litvan I, Agid Y, Calne D, Campbell G, Dubois B, Duvoisin RC, et al. Clinical research criteria for the diagnosis of progressive supranuclear palsy (Steele-Richardson-Olszewski syndrome): report of the NINDS-SPSP international workshop. Neurology. 1996;47:1–9.CrossRefPubMed Litvan I, Agid Y, Calne D, Campbell G, Dubois B, Duvoisin RC, et al. Clinical research criteria for the diagnosis of progressive supranuclear palsy (Steele-Richardson-Olszewski syndrome): report of the NINDS-SPSP international workshop. Neurology. 1996;47:1–9.CrossRefPubMed
13.
go back to reference Cselényi Z, Jönhagen ME, Forsberg A, Halldin C, Julin P, Schou M, et al. Clinical validation of 18F-AZD4694, an amyloid-β-specific PET radioligand. J Nucl Med. 2012;53:415–24.CrossRefPubMed Cselényi Z, Jönhagen ME, Forsberg A, Halldin C, Julin P, Schou M, et al. Clinical validation of 18F-AZD4694, an amyloid-β-specific PET radioligand. J Nucl Med. 2012;53:415–24.CrossRefPubMed
14.
go back to reference Ad-Dab’bagh Y, Lyttelton O, Muehlboeck JS, Lepage C, Einarson D, Mok K, et al. The CIVET image-processing environment: a fully automated comprehensive pipeline for anatomical neuroimaging research. Proc. 12th Annu. Meet. Organ. Hum. Brain Mapp. 2006. p. 2266. Ad-Dab’bagh Y, Lyttelton O, Muehlboeck JS, Lepage C, Einarson D, Mok K, et al. The CIVET image-processing environment: a fully automated comprehensive pipeline for anatomical neuroimaging research. Proc. 12th Annu. Meet. Organ. Hum. Brain Mapp. 2006. p. 2266.
15.
go back to reference Zijdenbos AP, Forghani R, Evans AC. Automatic “pipeline” analysis of 3-D MRI data for clinical trials: application to multiple sclerosis. IEEE Trans Med Imaging. 2002;21:1280–91.CrossRefPubMed Zijdenbos AP, Forghani R, Evans AC. Automatic “pipeline” analysis of 3-D MRI data for clinical trials: application to multiple sclerosis. IEEE Trans Med Imaging. 2002;21:1280–91.CrossRefPubMed
16.
go back to reference Pascoal TA, Mathotaarachchi S, Mohades S, Benedet AL, Chung C-O, Shin M, et al. Amyloid-β and hyperphosphorylated tau synergy drives metabolic decline in preclinical Alzheimer’s disease. Mol. Psychiatry. Nature Publishing Group; 2017;22:306–11. Pascoal TA, Mathotaarachchi S, Mohades S, Benedet AL, Chung C-O, Shin M, et al. Amyloid-β and hyperphosphorylated tau synergy drives metabolic decline in preclinical Alzheimer’s disease. Mol. Psychiatry. Nature Publishing Group; 2017;22:306–11.
17.
go back to reference Parent MJ, Bedard M-A, Aliaga A, Minuzzi L, Mechawar N, Soucy J-P, et al. Cholinergic depletion in Alzheimer’s disease shown by [18F]FEOBV autoradiography. Int J Mol Imaging. 2013;2013:1–6.CrossRef Parent MJ, Bedard M-A, Aliaga A, Minuzzi L, Mechawar N, Soucy J-P, et al. Cholinergic depletion in Alzheimer’s disease shown by [18F]FEOBV autoradiography. Int J Mol Imaging. 2013;2013:1–6.CrossRef
18.
go back to reference Levitt P, Pintar JE, Breakefield XO. Immunocytochemical demonstration of monoamine oxidase B in brain astrocytes and serotonergic neurons. Proc Natl Acad Sci U S A. 1982;79:6385–9.CrossRefPubMedPubMedCentral Levitt P, Pintar JE, Breakefield XO. Immunocytochemical demonstration of monoamine oxidase B in brain astrocytes and serotonergic neurons. Proc Natl Acad Sci U S A. 1982;79:6385–9.CrossRefPubMedPubMedCentral
19.
go back to reference Saura J, Andrés N, Andrade C, Ojuel J, Eriksson K, Mahy N, et al. Biphasic and region-specific MAO-B response to aging in normal human brain. Neurobiol Aging. 1980;18:497–507.CrossRef Saura J, Andrés N, Andrade C, Ojuel J, Eriksson K, Mahy N, et al. Biphasic and region-specific MAO-B response to aging in normal human brain. Neurobiol Aging. 1980;18:497–507.CrossRef
20.
go back to reference Fowler JS, Volkow ND, Wang GJ, Logan J, Pappas N, Shea C, et al. Age-related increases in brain monoamine oxidase B in living healthy human subjects. Neurobiol Aging. 1997;18:431–5.CrossRefPubMed Fowler JS, Volkow ND, Wang GJ, Logan J, Pappas N, Shea C, et al. Age-related increases in brain monoamine oxidase B in living healthy human subjects. Neurobiol Aging. 1997;18:431–5.CrossRefPubMed
21.
go back to reference Gulyás B, Pavlova E, Kása P, Gulya K, Bakota L, Várszegi S, et al. Activated MAO-B in the brain of Alzheimer patients, demonstrated by [11C]-l-deprenyl using whole hemisphere autoradiography. Neurochem Int. 2011;58:60–8.CrossRefPubMed Gulyás B, Pavlova E, Kása P, Gulya K, Bakota L, Várszegi S, et al. Activated MAO-B in the brain of Alzheimer patients, demonstrated by [11C]-l-deprenyl using whole hemisphere autoradiography. Neurochem Int. 2011;58:60–8.CrossRefPubMed
22.
go back to reference Schöll M, Carter SF, Westman E, Rodriguez-Vieitez E, Almkvist O, Thordardottir S, et al. Early astrocytosis in autosomal dominant Alzheimer’s disease measured in vivo by multi-tracer positron emission tomography. Sci Rep. 2015;5:16404.CrossRefPubMedPubMedCentral Schöll M, Carter SF, Westman E, Rodriguez-Vieitez E, Almkvist O, Thordardottir S, et al. Early astrocytosis in autosomal dominant Alzheimer’s disease measured in vivo by multi-tracer positron emission tomography. Sci Rep. 2015;5:16404.CrossRefPubMedPubMedCentral
23.
go back to reference Carter SF, Scholl M, Almkvist O, Wall A, Engler H, Langstrom B, et al. 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. 2012;53:37–46.CrossRefPubMed Carter SF, Scholl M, Almkvist O, Wall A, Engler H, Langstrom B, et al. 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. 2012;53:37–46.CrossRefPubMed
24.
go back to reference Foley P, Gerlach M, Youdim MB, Riederer P, Johnston JP, Kochersperger LM, et al. MAO-B inhibitors: multiple roles in the therapy of neurodegenerative disorders? Parkinsonism Relat Disord. 2000;6:25–47.CrossRefPubMed Foley P, Gerlach M, Youdim MB, Riederer P, Johnston JP, Kochersperger LM, et al. MAO-B inhibitors: multiple roles in the therapy of neurodegenerative disorders? Parkinsonism Relat Disord. 2000;6:25–47.CrossRefPubMed
25.
go back to reference Takano A, Halldin C, Farde L. Neuroimaging in psychiatric drug development and radioligand development for new targets. PET SPECT Psychiatry. Berlin, Heidelberg: Springer Berlin Heidelberg; 2014. p. 3–14. Takano A, Halldin C, Farde L. Neuroimaging in psychiatric drug development and radioligand development for new targets. PET SPECT Psychiatry. Berlin, Heidelberg: Springer Berlin Heidelberg; 2014. p. 3–14.
26.
go back to reference Logan J, Fowler JS, Volkow ND, Wang GJ, MacGregor RR, Shea C, et al. Reproducibility of repeated measures of deuterium substituted [11C]L-deprenyl ([11C]L-deprenyl-D2) binding in the human brain. Nucl Med Biol. 2000;27:43–9.CrossRefPubMed Logan J, Fowler JS, Volkow ND, Wang GJ, MacGregor RR, Shea C, et al. Reproducibility of repeated measures of deuterium substituted [11C]L-deprenyl ([11C]L-deprenyl-D2) binding in the human brain. Nucl Med Biol. 2000;27:43–9.CrossRefPubMed
27.
go back to reference Freedman NMT, Mishani E, Krausz Y, Weininger J, Lester H, Blaugrund E, et al. In vivo measurement of brain monoamine oxidase B occupancy by rasagiline, using (11)C-l-deprenyl and PET. J Nucl Med. 2005;46:1618–24.PubMed Freedman NMT, Mishani E, Krausz Y, Weininger J, Lester H, Blaugrund E, et al. In vivo measurement of brain monoamine oxidase B occupancy by rasagiline, using (11)C-l-deprenyl and PET. J Nucl Med. 2005;46:1618–24.PubMed
28.
go back to reference Fowler JS, Volkow ND, Wang G-J, Pappas N, Logan J, MacGregor R, et al. Inhibition of monoamine oxidase B in the brains of smokers. Nature. 1996;379:733–6.CrossRefPubMed Fowler JS, Volkow ND, Wang G-J, Pappas N, Logan J, MacGregor R, et al. Inhibition of monoamine oxidase B in the brains of smokers. Nature. 1996;379:733–6.CrossRefPubMed
29.
go back to reference Vina D, Serra S, Lamela M, Delogu G. Herbal natural products as a source of monoamine oxidase inhibitors: a review. Curr Top Med Chem. 2012;12:2131–44.CrossRefPubMed Vina D, Serra S, Lamela M, Delogu G. Herbal natural products as a source of monoamine oxidase inhibitors: a review. Curr Top Med Chem. 2012;12:2131–44.CrossRefPubMed
30.
go back to reference Fowler JS, Volkow ND, Logan J, Wang G-J, Mac Gregor RR, Schlyer D, et al. Slow recovery of human brain MAO B after L-deprenyl (Selegeline) withdrawal. Synapse. 1994;18:86–93.CrossRefPubMed Fowler JS, Volkow ND, Logan J, Wang G-J, Mac Gregor RR, Schlyer D, et al. Slow recovery of human brain MAO B after L-deprenyl (Selegeline) withdrawal. Synapse. 1994;18:86–93.CrossRefPubMed
31.
32.
go back to reference Ishiki A, Harada R, Okamura N, Tomita N, Rowe CC, Villemagne VL, et al. Tau imaging with [ 18 F]THK-5351 in progressive supranuclear palsy. Eur. J. Neurol. 2017;24:130–6. Ishiki A, Harada R, Okamura N, Tomita N, Rowe CC, Villemagne VL, et al. Tau imaging with [ 18 F]THK-5351 in progressive supranuclear palsy. Eur. J. Neurol. 2017;24:130–6.
33.
go back to reference Bench CJ, Price GW, Lammertsma AA, Cremer JC, Luthra SK, Turton D, et al. Measurement of human cerebral monoamine oxidase type B (MAO-B) activity with positron emission tomography (PET): a dose ranging study with the reversible inhibitor Ro 19-6327. Eur J Clin Pharmacol. 1991;40:169–73.CrossRefPubMed Bench CJ, Price GW, Lammertsma AA, Cremer JC, Luthra SK, Turton D, et al. Measurement of human cerebral monoamine oxidase type B (MAO-B) activity with positron emission tomography (PET): a dose ranging study with the reversible inhibitor Ro 19-6327. Eur J Clin Pharmacol. 1991;40:169–73.CrossRefPubMed
34.
go back to reference Lammertsma AA, Bench CJ, Hume SP, Osman S, Gunn K, Brooks DJ, et al. Comparison of methods for analysis of clinical [11C]raclopride studies. J Cereb Blood Flow Metab. 1996;16:42–52.CrossRefPubMed Lammertsma AA, Bench CJ, Hume SP, Osman S, Gunn K, Brooks DJ, et al. Comparison of methods for analysis of clinical [11C]raclopride studies. J Cereb Blood Flow Metab. 1996;16:42–52.CrossRefPubMed
35.
go back to reference Ekelund J, Slifstein M, Narendran R, Guillin O, Belani H, Guo N-N, et al. In vivo DA D1 receptor selectivity of NNC 112 and SCH 23390. Mol Imaging Biol. 2007;9:117–25.CrossRefPubMed Ekelund J, Slifstein M, Narendran R, Guillin O, Belani H, Guo N-N, et al. In vivo DA D1 receptor selectivity of NNC 112 and SCH 23390. Mol Imaging Biol. 2007;9:117–25.CrossRefPubMed
36.
go back to reference Kiss B, Horti F, Bobok A. In vitro and in vivo comparison of [3H](+)-PHNO and [3H]raclopride binding to rat striatum and lobes 9 and 10 of the cerebellum: a method to distinguish dopamine D3 from D2 receptor sites. Synapse. 2011;65:467–78.CrossRefPubMed Kiss B, Horti F, Bobok A. In vitro and in vivo comparison of [3H](+)-PHNO and [3H]raclopride binding to rat striatum and lobes 9 and 10 of the cerebellum: a method to distinguish dopamine D3 from D2 receptor sites. Synapse. 2011;65:467–78.CrossRefPubMed
37.
go back to reference Marquié M, Normandin MD, Vanderburg CR, Costantino IM, Bien EA, Rycyna LG, et al. Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann Neurol. 2015;78:787–800.CrossRefPubMedPubMedCentral Marquié M, Normandin MD, Vanderburg CR, Costantino IM, Bien EA, Rycyna LG, et al. Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann Neurol. 2015;78:787–800.CrossRefPubMedPubMedCentral
38.
go back to reference Braak H, Braak E. Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiol Aging. 1995;16:271–8.CrossRefPubMed Braak H, Braak E. Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiol Aging. 1995;16:271–8.CrossRefPubMed
39.
go back to reference Thomas T, McLendon C, Thomas G. L-deprenyl: nitric oxide production and dilation of cerebral blood vessels. Neuroreport. 1998;9:2595–600.CrossRefPubMed Thomas T, McLendon C, Thomas G. L-deprenyl: nitric oxide production and dilation of cerebral blood vessels. Neuroreport. 1998;9:2595–600.CrossRefPubMed
Metadata
Title
Monoamine oxidase B inhibitor, selegiline, reduces 18F-THK5351 uptake in the human brain
Authors
Kok Pin Ng
Tharick A. Pascoal
Sulantha Mathotaarachchi
Joseph Therriault
Min Su Kang
Monica Shin
Marie-Christine Guiot
Qi Guo
Ryuichi Harada
Robert A. Comley
Gassan Massarweh
Jean-Paul Soucy
Nobuyuki Okamura
Serge Gauthier
Pedro Rosa-Neto
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Alzheimer's Research & Therapy / Issue 1/2017
Electronic ISSN: 1758-9193
DOI
https://doi.org/10.1186/s13195-017-0253-y

Other articles of this Issue 1/2017

Alzheimer's Research & Therapy 1/2017 Go to the issue