Skip to main content
Top
Published in: European Journal of Nuclear Medicine and Molecular Imaging 4/2014

01-04-2014 | Original Article

Evaluation of 18F-BCPP-EF for mitochondrial complex 1 imaging in the brain of conscious monkeys using PET

Authors: Hideo Tsukada, Hiroyuki Ohba, Masakatsu Kanazawa, Takeharu Kakiuchi, Norihiro Harada

Published in: European Journal of Nuclear Medicine and Molecular Imaging | Issue 4/2014

Login to get access

Abstract

Purpose

We have reported on the development of a novel PET probe, 18F-2-tert-butyl-4-chloro-5-{6-[2-(2-fluoroethoxy)-ethoxy]-pyridin-3-ylmethoxy}-2H-pyridazin-3-one (18F-BCPP-EF), for quantitative imaging of mitochondrial complex 1 (MC-1) activity in the brain of the living rat. For clinical application in humans, translational research in the monkey was conducted.

Methods

PET measurements with 18F-BCPP-EF were performed in young and old monkeys (Macaca mulatta) in a conscious state with arterial blood sampling. The binding specificity of 18F-BCPP-EF was evaluated with rotenone, a specific MC-1 inhibitor, in young animals. The binding (total distribution volume, V T) of 18F-BCPP-EF was calculated using Logan graphical analysis, and one-tissue compartment model (1-TC) and two-tissue compartment model (2-TC) analyses using a metabolite-corrected plasma input function.

Results

F-BCPP-EF was rapidly taken up into the brain just after intravenous injection, peaked between 10 and 20 min after injection, and was then gradually eliminated. The 2-TC analysis provided a better fit than the 1-TC analysis, and the V T values from the 2-TC analysis correlated well with those from the Logan plot. With predosing with rotenone, 18F-BCPP-EF showed a higher uptake peak in the brain, followed by more rapid elimination thereafter than in the vehicle condition, resulting in significant reductions in 2-TC V T values in all regions. In old animals, the kinetics of 18F-BCPP-EF were slightly slower with lower peak levels than in young animals, resulting age-related reductions in 18F-BCPP-EF binding in all brain regions.

Conclusion

The present study demonstrated that 18F-BCPP-EF may be a potential PET probe for quantitative imaging MC-1 activity in the living brain using PET.
Literature
1.
go back to reference Meisenzahl EM, Schmitt GJ, Scheuerecker J, Möller HJ. The role of dopamine for the pathophysiology of schizophrenia. Int Rev Psychiatry. 2007;19:337–45.PubMedCrossRef Meisenzahl EM, Schmitt GJ, Scheuerecker J, Möller HJ. The role of dopamine for the pathophysiology of schizophrenia. Int Rev Psychiatry. 2007;19:337–45.PubMedCrossRef
2.
go back to reference Takahashi H, Higuchi M, Suhara T. The role of extrastriatal dopamine D2 receptors in schizophrenia. Biol Psychiatry. 2006;59:919–28.PubMedCrossRef Takahashi H, Higuchi M, Suhara T. The role of extrastriatal dopamine D2 receptors in schizophrenia. Biol Psychiatry. 2006;59:919–28.PubMedCrossRef
3.
go back to reference Brooke DJ. Imaging approaches to Parkinson disease. J Nucl Med. 2010;51:596–609.CrossRef Brooke DJ. Imaging approaches to Parkinson disease. J Nucl Med. 2010;51:596–609.CrossRef
4.
go back to reference Maetzler W, Liepelt I, Berg D. Progression of Parkinson’s disease in the clinical phase: potential markers. Lancet Neurol. 2009;8:1158–71.PubMedCrossRef Maetzler W, Liepelt I, Berg D. Progression of Parkinson’s disease in the clinical phase: potential markers. Lancet Neurol. 2009;8:1158–71.PubMedCrossRef
5.
go back to reference Kadir A, Nordberg A. Target-specific PET probes for neurodegenerative disorders related to dementia. J Nucl Med. 2010;51:1418–30.PubMedCrossRef Kadir A, Nordberg A. Target-specific PET probes for neurodegenerative disorders related to dementia. J Nucl Med. 2010;51:1418–30.PubMedCrossRef
6.
go back to reference Neyer JH. Neuroimaging markers of cellular function in major depressive disorder: implications for therapeutics, personalized medicine, and prevention. Clin Pharmacol Ther. 2012;91:201–14.CrossRef Neyer JH. Neuroimaging markers of cellular function in major depressive disorder: implications for therapeutics, personalized medicine, and prevention. Clin Pharmacol Ther. 2012;91:201–14.CrossRef
7.
go back to reference Schroeter M, Dennin MA, Walberer M, Backes H, Neumaier B, Fink GR, et al. Neuroinflammation extends brain tissue at risk to vital peri-infarct tissue: a double tracer [11C]PK11195- and [18F]FDG-PET study. J Cereb Blood Flow Metab. 2009;29:1216–25. Schroeter M, Dennin MA, Walberer M, Backes H, Neumaier B, Fink GR, et al. Neuroinflammation extends brain tissue at risk to vital peri-infarct tissue: a double tracer [11C]PK11195- and [18F]FDG-PET study. J Cereb Blood Flow Metab. 2009;29:1216–25.
8.
go back to reference Fukumoto D, Hosoya T, Nishiyama S, Harada N, Iwata H, Yamamoto S, et al. Multiparametric assessment of acute and sub-acute ischemic neuronal damage: a small animal PET study with rat photochemically induced thrombosis (PIT) model. Synapse. 2011;65:207–14.PubMedCrossRef Fukumoto D, Hosoya T, Nishiyama S, Harada N, Iwata H, Yamamoto S, et al. Multiparametric assessment of acute and sub-acute ischemic neuronal damage: a small animal PET study with rat photochemically induced thrombosis (PIT) model. Synapse. 2011;65:207–14.PubMedCrossRef
9.
go back to reference Winkeler A, Boisgard R, Martin A, Tavitian B. Radioisotopic imaging of neuroinflammation. J Nucl Med. 2010;51:1–4.PubMedCrossRef Winkeler A, Boisgard R, Martin A, Tavitian B. Radioisotopic imaging of neuroinflammation. J Nucl Med. 2010;51:1–4.PubMedCrossRef
10.
11.
go back to reference Purohit A, Radeke H, Azure M, Hanson K, Benetti R, Su F, et al. Synthesis and biological evaluation of pyridazinone analogues as potential cardiac positron emission tomography tracers. J Med Chem. 2008;51:2954–70.PubMedCrossRef Purohit A, Radeke H, Azure M, Hanson K, Benetti R, Su F, et al. Synthesis and biological evaluation of pyridazinone analogues as potential cardiac positron emission tomography tracers. J Med Chem. 2008;51:2954–70.PubMedCrossRef
12.
go back to reference Yalamanchili P, Wexler E, Hayes M, Yu M, Bozek J, Kagan M, et al. Mechanism of uptake and retention of F-18 BMS-747158-02 in cardiomyocytes: a novel PET myocardial imaging agent. J Nucl Cardiol. 2007;14:782–8.PubMedCrossRef Yalamanchili P, Wexler E, Hayes M, Yu M, Bozek J, Kagan M, et al. Mechanism of uptake and retention of F-18 BMS-747158-02 in cardiomyocytes: a novel PET myocardial imaging agent. J Nucl Cardiol. 2007;14:782–8.PubMedCrossRef
13.
go back to reference Huisman MC, Higuchi T, Reder S, Nekolla SG, Poethko T, Wester HJ, et al. Initial characterization of an 18F-labeled myocardial perfusion tracer. J Nucl Med. 2008;49:630–6. Huisman MC, Higuchi T, Reder S, Nekolla SG, Poethko T, Wester HJ, et al. Initial characterization of an 18F-labeled myocardial perfusion tracer. J Nucl Med. 2008;49:630–6.
14.
go back to reference Fukumoto D, Nishiyama S, Harada N, Yamamoto S, Tsukada H. Detection of ischemic neuronal damage with [18F]BMS-747158-02, a mitochondrial complex-1 PET ligand: small animal PET study in rat brain. Synapse. 2012;66:909–17. Fukumoto D, Nishiyama S, Harada N, Yamamoto S, Tsukada H. Detection of ischemic neuronal damage with [18F]BMS-747158-02, a mitochondrial complex-1 PET ligand: small animal PET study in rat brain. Synapse. 2012;66:909–17.
15.
go back to reference Harada N, Nishiyama S, Kanazawa M, Tsukada H. Development of novel PET probes, [18F]BCPP-EF, [18F]BCPP-BF, and [11C]BCPP-EM for mitochondrial complex 1 imaging in the living brain. J Label Compd Radiopharm. 2013;56:553–61. Harada N, Nishiyama S, Kanazawa M, Tsukada H. Development of novel PET probes, [18F]BCPP-EF, [18F]BCPP-BF, and [11C]BCPP-EM for mitochondrial complex 1 imaging in the living brain. J Label Compd Radiopharm. 2013;56:553–61.
16.
go back to reference Noda A, Takamatsu H, Minoshima S, Tsukada H, Nishimura S. Determination of kinetic rate constants for FDG and partition coefficient of water in conscious macaque and alterations in aging or anesthesia examined on parametric images with an anatomic standardization technique. J Cereb Blood Flow Metab. 2003;23:1441–7.PubMedCrossRef Noda A, Takamatsu H, Minoshima S, Tsukada H, Nishimura S. Determination of kinetic rate constants for FDG and partition coefficient of water in conscious macaque and alterations in aging or anesthesia examined on parametric images with an anatomic standardization technique. J Cereb Blood Flow Metab. 2003;23:1441–7.PubMedCrossRef
17.
go back to reference Tsukada H, Harada N, Nishiyama S, Ohba H, Kakiuchi T. Cholinergic neuronal modulation alters dopamine D2 receptor availability in vivo by regulating receptor affinity induced by facilitated synaptic dopamine turnover? Positron emission tomography studies with microdialysis in the conscious monkey brain. J Neurosci. 2000;20:7067–73.PubMed Tsukada H, Harada N, Nishiyama S, Ohba H, Kakiuchi T. Cholinergic neuronal modulation alters dopamine D2 receptor availability in vivo by regulating receptor affinity induced by facilitated synaptic dopamine turnover? Positron emission tomography studies with microdialysis in the conscious monkey brain. J Neurosci. 2000;20:7067–73.PubMed
18.
go back to reference Watanabe M, Okada H, Shimizu K, Omura T, Yoshikawa E, Kosugi T, et al. A high resolution animal PET scanner using compact PS-PMT detectors. IEEE Trans Nucl Sci. 1997;44:1277–82.CrossRef Watanabe M, Okada H, Shimizu K, Omura T, Yoshikawa E, Kosugi T, et al. A high resolution animal PET scanner using compact PS-PMT detectors. IEEE Trans Nucl Sci. 1997;44:1277–82.CrossRef
19.
go back to reference Jones EG, Stone JM, Karten HJ. High-resolution digital brain atlases: a Hubble telescope for the brain. Ann N Y Acad Sci. 2011;1225(S1):E147–59.PubMedCrossRef Jones EG, Stone JM, Karten HJ. High-resolution digital brain atlases: a Hubble telescope for the brain. Ann N Y Acad Sci. 2011;1225(S1):E147–59.PubMedCrossRef
20.
go back to reference Logan J, Fowler JS, Volkow ND, Wolf AP, Dewey SL, Schlyer DJ, et al. Graphical analysis of reversible radioligand binding from time-activity measurements applied to [N-11C-methyl]-(-)-cocaine PET studies in human subjects. J Cereb Blood Flow Metab. 1990;10:740–7. Logan J, Fowler JS, Volkow ND, Wolf AP, Dewey SL, Schlyer DJ, et al. Graphical analysis of reversible radioligand binding from time-activity measurements applied to [N-11C-methyl]-(-)-cocaine PET studies in human subjects. J Cereb Blood Flow Metab. 1990;10:740–7.
21.
go back to reference Huang SH, Barrio J, Phelps M. Neuroreceptor assay with positron emission tomography; equilibrium versus dynamic approach. J Cereb Blood Flow Metab. 1986;6:515–21.PubMedCrossRef Huang SH, Barrio J, Phelps M. Neuroreceptor assay with positron emission tomography; equilibrium versus dynamic approach. J Cereb Blood Flow Metab. 1986;6:515–21.PubMedCrossRef
22.
go back to reference Mintun MA, Raichle ME, Kilbourn MR, Wooten GF, Welch MJ. A quantitative model for the in vivo assessment of drug binding sites with positron emission tomography. Ann Neurol. 1984;15:217–27.PubMedCrossRef Mintun MA, Raichle ME, Kilbourn MR, Wooten GF, Welch MJ. A quantitative model for the in vivo assessment of drug binding sites with positron emission tomography. Ann Neurol. 1984;15:217–27.PubMedCrossRef
23.
go back to reference Phelps ME. PET molecular imaging and its biological applications. New York: Springer; 2004. p. 125–216. Phelps ME. PET molecular imaging and its biological applications. New York: Springer; 2004. p. 125–216.
24.
go back to reference Akaike H. A new look at the statistical model identification. IEEE Trans Autom Control. 1974;19:716–23.CrossRef Akaike H. A new look at the statistical model identification. IEEE Trans Autom Control. 1974;19:716–23.CrossRef
25.
go back to reference Waterhouse RN. Determination of lipophilicity and its use as a predictor of blood–brain barrier penetration of molecular imaging agents. Mol Imaging Biol. 2003;5:376–89.PubMedCrossRef Waterhouse RN. Determination of lipophilicity and its use as a predictor of blood–brain barrier penetration of molecular imaging agents. Mol Imaging Biol. 2003;5:376–89.PubMedCrossRef
26.
go back to reference Carson RE, Kiesewetter DO, Jagoda E, Der MG, Herscovich P, Eckelman WC. Muscarinic cholinergic receptor measurements with [18F]FP-TZTP: control and competition studies. J Cereb Blood Flow Metab. 1998;18:1130–42. Carson RE, Kiesewetter DO, Jagoda E, Der MG, Herscovich P, Eckelman WC. Muscarinic cholinergic receptor measurements with [18F]FP-TZTP: control and competition studies. J Cereb Blood Flow Metab. 1998;18:1130–42.
27.
go back to reference Nishiyama S, Tsukada H, Sato K, Kakiuchi T, Ohba H, Harada N, et al. Evaluation of PET ligands (+)N-[11C]ethyl-3-piperidyl benzilate and (+)N-[11C]propyl-3-piperidyl benzilate for muscarinic cholinergic receptors: a PET study with microdialysis in comparison with (+)N-[11C]methyl-3-piperidyl benzilate in the conscious monkey brain. Synapse. 2001;40:159–69. Nishiyama S, Tsukada H, Sato K, Kakiuchi T, Ohba H, Harada N, et al. Evaluation of PET ligands (+)N-[11C]ethyl-3-piperidyl benzilate and (+)N-[11C]propyl-3-piperidyl benzilate for muscarinic cholinergic receptors: a PET study with microdialysis in comparison with (+)N-[11C]methyl-3-piperidyl benzilate in the conscious monkey brain. Synapse. 2001;40:159–69.
28.
go back to reference Koeppe RA, Frey KA, Mulholland GK, Kilbourn MR, Buck A, Lee KS, et al. [11C]Tropanyl benzilate binding to muscarinic cholinergic receptors: methodology and kinetic modeling alterations. J Cereb Blood Flow Metab. 1994;14:85–99.PubMedCrossRef Koeppe RA, Frey KA, Mulholland GK, Kilbourn MR, Buck A, Lee KS, et al. [11C]Tropanyl benzilate binding to muscarinic cholinergic receptors: methodology and kinetic modeling alterations. J Cereb Blood Flow Metab. 1994;14:85–99.PubMedCrossRef
29.
go back to reference Farde L, Eriksson L, Blomqvist G, Halldin C. Kinetic analysis of central [11C]raclopride binding to D2-dopamine receptors studied by PET—a comparison to the equilibrium analysis. J Cereb Blood Flow Metab. 1989;9:696–708. Farde L, Eriksson L, Blomqvist G, Halldin C. Kinetic analysis of central [11C]raclopride binding to D2-dopamine receptors studied by PET—a comparison to the equilibrium analysis. J Cereb Blood Flow Metab. 1989;9:696–708.
30.
go back to reference Logan J, Volkow ND, Fowler JS, Wang G-J, Dewey SL, MacGregor R, et al. Effects of blood flow on [11C]raclopride binding in the brain: model simulations and kinetic analysis of PET data. J Cereb Blood Flow Metab. 1994;14:995–1010. Logan J, Volkow ND, Fowler JS, Wang G-J, Dewey SL, MacGregor R, et al. Effects of blood flow on [11C]raclopride binding in the brain: model simulations and kinetic analysis of PET data. J Cereb Blood Flow Metab. 1994;14:995–1010.
32.
go back to reference Cadenas E, Boveris A, Ragan CI, Stoppani AOM. Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria. Arch Biochem Biophys. 1977;180:248–57.PubMedCrossRef Cadenas E, Boveris A, Ragan CI, Stoppani AOM. Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria. Arch Biochem Biophys. 1977;180:248–57.PubMedCrossRef
33.
go back to reference Ojaimi J, Masters CL, Opeskin K, McKelvie P, Byrne E. Mitochondrial respiratory chain activity in the human brain as a function of age. Mech Ageing Dev. 1999;111:39–47.PubMedCrossRef Ojaimi J, Masters CL, Opeskin K, McKelvie P, Byrne E. Mitochondrial respiratory chain activity in the human brain as a function of age. Mech Ageing Dev. 1999;111:39–47.PubMedCrossRef
34.
go back to reference Navarro A, Boveris A. The mitochondrial energy transduction system and the aging process. Am J Physiol Cell Physiol. 2007;292:C670–86.PubMedCrossRef Navarro A, Boveris A. The mitochondrial energy transduction system and the aging process. Am J Physiol Cell Physiol. 2007;292:C670–86.PubMedCrossRef
35.
go back to reference Raha S, Robinson BH. Mitochondria, oxygen free radicals, disease and ageing. Trends Biochem Sci. 2000;25:502–8.PubMedCrossRef Raha S, Robinson BH. Mitochondria, oxygen free radicals, disease and ageing. Trends Biochem Sci. 2000;25:502–8.PubMedCrossRef
36.
go back to reference Manczak M, Jung Y, Park BS, Partovi D, Reddy PH. Time-course of mitochondrial gene expressions in mice brains: implications for mitochondrial dysfunction, oxidative damage, and cytochrome c in aging. J Neurochem. 2005;92:494–504.PubMedCrossRef Manczak M, Jung Y, Park BS, Partovi D, Reddy PH. Time-course of mitochondrial gene expressions in mice brains: implications for mitochondrial dysfunction, oxidative damage, and cytochrome c in aging. J Neurochem. 2005;92:494–504.PubMedCrossRef
37.
go back to reference Benzi G, Pastoris O, Marzatico F, Villa RF, Dagani F, Curti D. The mitochondrial electron transfer alteration as a factor involved in the brain aging. Neurobiol Age. 1992;13:361–8.CrossRef Benzi G, Pastoris O, Marzatico F, Villa RF, Dagani F, Curti D. The mitochondrial electron transfer alteration as a factor involved in the brain aging. Neurobiol Age. 1992;13:361–8.CrossRef
Metadata
Title
Evaluation of 18F-BCPP-EF for mitochondrial complex 1 imaging in the brain of conscious monkeys using PET
Authors
Hideo Tsukada
Hiroyuki Ohba
Masakatsu Kanazawa
Takeharu Kakiuchi
Norihiro Harada
Publication date
01-04-2014
Publisher
Springer Berlin Heidelberg
Published in
European Journal of Nuclear Medicine and Molecular Imaging / Issue 4/2014
Print ISSN: 1619-7070
Electronic ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-013-2628-z

Other articles of this Issue 4/2014

European Journal of Nuclear Medicine and Molecular Imaging 4/2014 Go to the issue