Skip to main content
Top
Published in: EJNMMI Research 1/2018

Open Access 01-12-2018 | Original research

First in human evaluation of [18F]PK-209, a PET ligand for the ion channel binding site of NMDA receptors

Authors: Jasper van der Aart, Sandeep S. V. Golla, Marieke van der Pluijm, Lothar A. Schwarte, Robert C. Schuit, Pieter J. Klein, Athanasios Metaxas, Albert D. Windhorst, Ronald Boellaard, Adriaan A. Lammertsma, Bart N. M. van Berckel

Published in: EJNMMI Research | Issue 1/2018

Login to get access

Abstract

Background

Efforts to develop suitable positron emission tomography (PET) tracers for the ion channel site of human N-methyl-d-aspartate (NMDA) receptors have had limited success. [18F]PK-209 is a GMOM derivative that binds to the intrachannel phencyclidine site with high affinity and selectivity. Primate PET studies have shown that the volume of distribution in the brain was reduced by administration of the NMDA receptor antagonist MK-801, consistent with substantial specific binding. The purpose of the present study was to evaluate [18F]PK-209 in 10 healthy humans by assessing test–retest reproducibility and binding specificity following intravenous S-ketamine administration (0.5 mg ∙ kg−1). Five healthy subjects underwent a test–retest protocol, and five others a baseline-ketamine protocol. In all cases dynamic, 120-min PET scans were acquired together with metabolite-corrected arterial plasma input functions. Additional input functions were tested based on within-subject and population-average parent fractions.

Results

Best fits of the brain time-activity curves were obtained using an irreversible two-tissue compartment model with additional blood volume parameter. Mean test–retest variability of the net rate of influx Ki varied between 7 and 24% depending on the input function. There were no consistent changes in [18F]PK-209 PET parameters following ketamine administration, which may be a consequence of the complex endogenous ligand processes that affect channel gating.

Conclusions

The molecular interaction between [18F]PK-209 and the binding site within the NMDA receptor ion channel is insufficiently reproducible and specific to be a reliable imaging agent for its quantification.

Trial registration

EudraCT 2014-001735-36. Registered 28 April 2014
Literature
1.
go back to reference Paoletti P, Bellone C, Zhou Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci. 2013;14:383–400.CrossRefPubMed Paoletti P, Bellone C, Zhou Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci. 2013;14:383–400.CrossRefPubMed
2.
go back to reference Javitt DC, Zukin SR. Biexponential kinetics of [3H]MK-801 binding: evidence for access to closed and open N-methyl-D-aspartate receptor channels. Mol Pharmacol. 1989;35:387–93.PubMed Javitt DC, Zukin SR. Biexponential kinetics of [3H]MK-801 binding: evidence for access to closed and open N-methyl-D-aspartate receptor channels. Mol Pharmacol. 1989;35:387–93.PubMed
3.
5.
go back to reference Danysz W, Parsons CG. Alzheimer’s disease, β-amyloid, glutamate, NMDA receptors and memantine--searching for the connections. Br J Pharmacol. 2012;167:324–52.CrossRefPubMedPubMedCentral Danysz W, Parsons CG. Alzheimer’s disease, β-amyloid, glutamate, NMDA receptors and memantine--searching for the connections. Br J Pharmacol. 2012;167:324–52.CrossRefPubMedPubMedCentral
6.
go back to reference Ametamey SM, Bruehlmeier M, Kneifel S, et al. PET studies of 18F-memantine in healthy volunteers. Nucl Med Biol. 2002;29:227–31.CrossRefPubMed Ametamey SM, Bruehlmeier M, Kneifel S, et al. PET studies of 18F-memantine in healthy volunteers. Nucl Med Biol. 2002;29:227–31.CrossRefPubMed
7.
go back to reference Asselin M, Hammers A, Turton D, Osman S, Koepp M, Brooks D. Initial kinetic analyses of the in vivo binding of the putative NMDA receptor ligand [C-11]CNS 5161 in humans. Neuroimage. 2004;22:T137. Asselin M, Hammers A, Turton D, Osman S, Koepp M, Brooks D. Initial kinetic analyses of the in vivo binding of the putative NMDA receptor ligand [C-11]CNS 5161 in humans. Neuroimage. 2004;22:T137.
8.
go back to reference Hartvig P, Valtysson J, Lindner KJ, et al. Central nervous system effects of subdissociative doses of (S)-ketamine are related to plasma and brain concentrations measured with positron emission tomography in healthy volunteers. Clin Pharmacol Ther. 1995;58:165–73.CrossRefPubMed Hartvig P, Valtysson J, Lindner KJ, et al. Central nervous system effects of subdissociative doses of (S)-ketamine are related to plasma and brain concentrations measured with positron emission tomography in healthy volunteers. Clin Pharmacol Ther. 1995;58:165–73.CrossRefPubMed
9.
go back to reference Klein PJ, Chomet M, Metaxas A, et al. Radiolabeling and evaluation of novel amine guanidine derivatives as potential positron emission tomography tracers for the ion channel of the N -methyl-d-aspartate receptor. Eur J Med Chem. 2016;118:143–60.CrossRefPubMed Klein PJ, Chomet M, Metaxas A, et al. Radiolabeling and evaluation of novel amine guanidine derivatives as potential positron emission tomography tracers for the ion channel of the N -methyl-d-aspartate receptor. Eur J Med Chem. 2016;118:143–60.CrossRefPubMed
10.
go back to reference van der Doef TF, Golla SS, Klein PJ, et al. Quantification of the novel N-methyl-d-aspartate receptor ligand [11C]GMOM in man. J Cereb Blood Flow Metab. 2016;36:1111–21. van der Doef TF, Golla SS, Klein PJ, et al. Quantification of the novel N-methyl-d-aspartate receptor ligand [11C]GMOM in man. J Cereb Blood Flow Metab. 2016;36:1111–21.
11.
go back to reference Waterhouse RN, Slifstein M, Dumont F, et al. In vivo evaluation of [11C]N-(2-chloro-5-thiomethylphenyl)-N′-(3-methoxy-phenyl)-N′-methylguanidine ([11C]GMOM) as a potential PET radiotracer for the PCP/NMDA receptor. Nucl Med Biol. 2004;31:939–48.CrossRefPubMed Waterhouse RN, Slifstein M, Dumont F, et al. In vivo evaluation of [11C]N-(2-chloro-5-thiomethylphenyl)-N′-(3-methoxy-phenyl)-N′-methylguanidine ([11C]GMOM) as a potential PET radiotracer for the PCP/NMDA receptor. Nucl Med Biol. 2004;31:939–48.CrossRefPubMed
12.
go back to reference van der Aart J, van der Doef TF, Horstman P, et al. Human dosimetry of the N-methyl-d-aspartate receptor ligand 11 C-GMOM. J Nucl Med. 2017;58:1330–3.CrossRefPubMed van der Aart J, van der Doef TF, Horstman P, et al. Human dosimetry of the N-methyl-d-aspartate receptor ligand 11 C-GMOM. J Nucl Med. 2017;58:1330–3.CrossRefPubMed
13.
go back to reference Klein PJ, Christiaans JAM, Metaxas A, et al. Synthesis, structure activity relationship, radiolabeling and preclinical evaluation of high affinity ligands for the ion channel of the N-methyl-d-aspartate receptor as potential imaging probes for positron emission tomography. Bioorg Med Chem. 2015;23:1189–206.CrossRefPubMed Klein PJ, Christiaans JAM, Metaxas A, et al. Synthesis, structure activity relationship, radiolabeling and preclinical evaluation of high affinity ligands for the ion channel of the N-methyl-d-aspartate receptor as potential imaging probes for positron emission tomography. Bioorg Med Chem. 2015;23:1189–206.CrossRefPubMed
14.
go back to reference Klein PJ, Schuit RC, Metaxas A, et al. Synthesis, radiolabeling and preclinical evaluation of a [11C]GMOM derivative as PET radiotracer for the ion channel of the N-methyl-d-aspartate receptor. Nucl Med Biol. 2017;51:25–32.CrossRefPubMed Klein PJ, Schuit RC, Metaxas A, et al. Synthesis, radiolabeling and preclinical evaluation of a [11C]GMOM derivative as PET radiotracer for the ion channel of the N-methyl-d-aspartate receptor. Nucl Med Biol. 2017;51:25–32.CrossRefPubMed
15.
go back to reference Golla SSV, Klein PJ, Bakker J, et al. Preclinical evaluation of [18F]PK-209, a new PET ligand for imaging the ion-channel site of NMDA receptors. Nucl Med Biol. 2015;42:205–12.CrossRefPubMed Golla SSV, Klein PJ, Bakker J, et al. Preclinical evaluation of [18F]PK-209, a new PET ligand for imaging the ion-channel site of NMDA receptors. Nucl Med Biol. 2015;42:205–12.CrossRefPubMed
16.
go back to reference Surti S, Kuhn A, Werner ME, et al. Performance of Philips Gemini TF PET/CT scanner with special consideration for its time-of-flight imaging capabilities. J Nucl Med. 2007;48:471–80.PubMed Surti S, Kuhn A, Werner ME, et al. Performance of Philips Gemini TF PET/CT scanner with special consideration for its time-of-flight imaging capabilities. J Nucl Med. 2007;48:471–80.PubMed
17.
go back to reference Vollmar S, Michel C, Treffert JT, et al. HeinzelCluster: accelerated reconstruction for FORE and OSEM3D. Phys Med Biol. 2002;47:307.CrossRef Vollmar S, Michel C, Treffert JT, et al. HeinzelCluster: accelerated reconstruction for FORE and OSEM3D. Phys Med Biol. 2002;47:307.CrossRef
18.
go back to reference Zhang Y, Brady M, Smith S. Segmentation of brain MR images through a hidden Markov random field model and the expectation-maximization algorithm. IEEE Trans Med Imaging. 2001;20:45–57.CrossRefPubMed Zhang Y, Brady M, Smith S. Segmentation of brain MR images through a hidden Markov random field model and the expectation-maximization algorithm. IEEE Trans Med Imaging. 2001;20:45–57.CrossRefPubMed
19.
go back to reference Hammers A, Allom R, Koepp MJ, et al. Three-dimensional maximum probability atlas of the human brain, with particular reference to the temporal lobe. Hum Brain Mapp. 2003;19:224–47.CrossRefPubMed Hammers A, Allom R, Koepp MJ, et al. Three-dimensional maximum probability atlas of the human brain, with particular reference to the temporal lobe. Hum Brain Mapp. 2003;19:224–47.CrossRefPubMed
20.
go back to reference Akaike H. Data analysis by statistical models. No To Hatatsu. 1992;24:127–33. Akaike H. Data analysis by statistical models. No To Hatatsu. 1992;24:127–33.
22.
go back to reference Kumlien E, Hartvig P, Valind S, et al. NMDA-receptor activity visualized with (S)-[N-methyl-11C]ketamine and positron emission tomography in patients with medial temporal lobe epilepsy. Epilepsia. 1999;40:30–7.CrossRefPubMed Kumlien E, Hartvig P, Valind S, et al. NMDA-receptor activity visualized with (S)-[N-methyl-11C]ketamine and positron emission tomography in patients with medial temporal lobe epilepsy. Epilepsia. 1999;40:30–7.CrossRefPubMed
23.
go back to reference Schiffer WK, Pareto-Onghena D, Wu H, et al. In vivo evaluation of [11C]CNS-5161 as a use-dependent ligand for the NMDA glutamate receptor channel. J Cereb Blood Flow Metab. 2005;25:S595.CrossRef Schiffer WK, Pareto-Onghena D, Wu H, et al. In vivo evaluation of [11C]CNS-5161 as a use-dependent ligand for the NMDA glutamate receptor channel. J Cereb Blood Flow Metab. 2005;25:S595.CrossRef
24.
go back to reference McGinnity CJ, Hammers A, Riaño Barros DA, et al. Initial evaluation of 18F-GE-179, a putative PET tracer for activated N-methyl d-aspartate receptors. J Nucl Med. 2014;55:423–30.CrossRefPubMed McGinnity CJ, Hammers A, Riaño Barros DA, et al. Initial evaluation of 18F-GE-179, a putative PET tracer for activated N-methyl d-aspartate receptors. J Nucl Med. 2014;55:423–30.CrossRefPubMed
25.
go back to reference DeLorenzo C, Gallezot JD, Gardus J, et al. In vivo variation in same-day estimates of metabotropic glutamate receptor subtype 5 binding using [11C]ABP688 and [18F]FPEB. J Cereb Blood Flow Metab. 2017;37:2716–27.CrossRefPubMed DeLorenzo C, Gallezot JD, Gardus J, et al. In vivo variation in same-day estimates of metabotropic glutamate receptor subtype 5 binding using [11C]ABP688 and [18F]FPEB. J Cereb Blood Flow Metab. 2017;37:2716–27.CrossRefPubMed
26.
go back to reference Matheson GJ, Schain M, Almeida R, et al. Diurnal and seasonal variation of the brain serotonin system in healthy male subjects. Neuroimage. 2015;112:225–31.CrossRefPubMed Matheson GJ, Schain M, Almeida R, et al. Diurnal and seasonal variation of the brain serotonin system in healthy male subjects. Neuroimage. 2015;112:225–31.CrossRefPubMed
27.
go back to reference Zeiler FA, Sader N, Gillman LM, et al. The cerebrovascular response to ketamine: a systematic review of the animal and human literature. J Neurosurg Anesthesiol. 2016;28:123–40.CrossRefPubMed Zeiler FA, Sader N, Gillman LM, et al. The cerebrovascular response to ketamine: a systematic review of the animal and human literature. J Neurosurg Anesthesiol. 2016;28:123–40.CrossRefPubMed
28.
go back to reference Schoenberger M, Schroeder FA, Placzek MS, et al. In vivo [ 18 F]GE-179 brain signal does not show NMDA-specific modulation with drug challenges in rodents and nonhuman primates. ACS Chem Neurosci. 2018;9:298–305.CrossRefPubMed Schoenberger M, Schroeder FA, Placzek MS, et al. In vivo [ 18 F]GE-179 brain signal does not show NMDA-specific modulation with drug challenges in rodents and nonhuman primates. ACS Chem Neurosci. 2018;9:298–305.CrossRefPubMed
29.
go back to reference Stone JM, Erlandsson K, Arstad E, et al. Relationship between ketamine-induced psychotic symptoms and NMDA receptor occupancy—a [123I]CNS-1261 SPET study. Psychopharmacology. 2008;197:401–8.CrossRefPubMed Stone JM, Erlandsson K, Arstad E, et al. Relationship between ketamine-induced psychotic symptoms and NMDA receptor occupancy—a [123I]CNS-1261 SPET study. Psychopharmacology. 2008;197:401–8.CrossRefPubMed
30.
go back to reference Fernandes A, Wojcik T, Baireddy P, et al. Inhibition of in vivo [3H]MK-801 binding by NMDA receptor open channel blockers and GluN2B antagonists in rats and mice. Eur J Pharmacol. 2015;766:1–8.CrossRefPubMed Fernandes A, Wojcik T, Baireddy P, et al. Inhibition of in vivo [3H]MK-801 binding by NMDA receptor open channel blockers and GluN2B antagonists in rats and mice. Eur J Pharmacol. 2015;766:1–8.CrossRefPubMed
32.
go back to reference Nikolaev MV, Magazanik LG, Tikhonov DB. Influence of external magnesium ions on the NMDA receptor channel block by different types of organic cations. Neuropharmacology. 2012;62:2078–85.CrossRefPubMed Nikolaev MV, Magazanik LG, Tikhonov DB. Influence of external magnesium ions on the NMDA receptor channel block by different types of organic cations. Neuropharmacology. 2012;62:2078–85.CrossRefPubMed
33.
go back to reference Johnson JW, Glasgow NG, Povysheva NV. Recent insights into the mode of action of memantine and ketamine. Curr Opin Pharmacol. 2015;20:54–63.CrossRefPubMed Johnson JW, Glasgow NG, Povysheva NV. Recent insights into the mode of action of memantine and ketamine. Curr Opin Pharmacol. 2015;20:54–63.CrossRefPubMed
35.
go back to reference Starmer CF, Packer DL, Grant AO. Ligand binding to transiently accessible sites: mechanisms for varying apparent binding rates. J Theor Biol. 1987;124:335–41.CrossRefPubMed Starmer CF, Packer DL, Grant AO. Ligand binding to transiently accessible sites: mechanisms for varying apparent binding rates. J Theor Biol. 1987;124:335–41.CrossRefPubMed
Metadata
Title
First in human evaluation of [18F]PK-209, a PET ligand for the ion channel binding site of NMDA receptors
Authors
Jasper van der Aart
Sandeep S. V. Golla
Marieke van der Pluijm
Lothar A. Schwarte
Robert C. Schuit
Pieter J. Klein
Athanasios Metaxas
Albert D. Windhorst
Ronald Boellaard
Adriaan A. Lammertsma
Bart N. M. van Berckel
Publication date
01-12-2018
Publisher
Springer Berlin Heidelberg
Published in
EJNMMI Research / Issue 1/2018
Electronic ISSN: 2191-219X
DOI
https://doi.org/10.1186/s13550-018-0424-2

Other articles of this Issue 1/2018

EJNMMI Research 1/2018 Go to the issue