Sigma-1 and dopamine D2/D3 receptor occupancy of pridopidine in healthy volunteers and patients with Huntington disease: a [18F] fluspidine and [18F] fallypride PET study

Igor D Grachev, Philipp M Meyer, Georg A Becker, Marcus Bronzel, Doug Marsteller, Gina Pastino, Ole Voges, Laura Rabinovich, Helena Knebel, Franziska Zientek, Michael Rullmann, Bernhard Sattler, Marianne Patt, Thilo Gerhards, Maria Strauss, Andreas Kluge, Peter Brust, Juha-Matti Savola, Mark F Gordon, Michal Geva, Swen Hesse, Henryk Barthel, Michael R Hayden, Osama Sabri, Igor D Grachev, Philipp M Meyer, Georg A Becker, Marcus Bronzel, Doug Marsteller, Gina Pastino, Ole Voges, Laura Rabinovich, Helena Knebel, Franziska Zientek, Michael Rullmann, Bernhard Sattler, Marianne Patt, Thilo Gerhards, Maria Strauss, Andreas Kluge, Peter Brust, Juha-Matti Savola, Mark F Gordon, Michal Geva, Swen Hesse, Henryk Barthel, Michael R Hayden, Osama Sabri

Abstract

Purpose: Pridopidine is an investigational drug for Huntington disease (HD). Pridopidine was originally thought to act as a dopamine stabilizer. However, pridopidine shows highest affinity to the sigma-1 receptor (S1R) and enhances neuroprotection via the S1R in preclinical studies. Using [18F] fluspidine and [18F] fallypride PET, the purpose of this study was to assess in vivo target engagement/receptor occupancy of pridopidine to the S1R and dopamine D2/D3 receptor (D2/D3R) at clinical relevant doses in healthy volunteers (HVs) and as proof-of-concept in a small number of patients with HD.

Methods: Using [18F] fluspidine PET (300 MBq, 0-90 min), 11 male HVs (pridopidine 0.5 to 90 mg; six dose groups) and three male patients with HD (pridopidine 90 mg) were investigated twice, without and 2 h after single dose of pridopidine. Using [18F] fallypride PET (200 MBq, 0-210 min), four male HVs were studied without and 2 h following pridopidine administration (90 mg). Receptor occupancy was analyzed by the Lassen plot.

Results: S1R occupancy as function of pridopidine dose (or plasma concentration) in HVs could be described by a three-parameter Hill equation with a Hill coefficient larger than one. A high degree of S1R occupancy (87% to 91%) was found throughout the brain at pridopidine doses ranging from 22.5 to 90 mg. S1R occupancy was 43% at 1 mg pridopidine. In contrast, at 90 mg pridopidine, the D2/D3R occupancy was only minimal (~ 3%).

Conclusions: Our PET findings indicate that at clinically relevant single dose of 90 mg, pridopidine acts as a selective S1R ligand showing near to complete S1R occupancy with negligible occupancy of the D2/D3R. The dose S1R occupancy relationship suggests cooperative binding of pridopidine to the S1R. Our findings provide significant clarification about pridopidine's mechanism of action and support further use of the 45-mg twice-daily dose to achieve full and selective targeting of the S1R in future clinical trials of neurodegenerative disorders. Clinical Trials.gov Identifier: NCT03019289 January 12, 2017; EUDRA-CT-Nr. 2016-001757-41.

Keywords: Dopamine D2/D3 receptor occupancy; Huntington disease; PET; Pridopidine; Sigma-1 receptor occupancy; [18F]fluspidine.

Conflict of interest statement

Dr. Grachev was an employee of Teva Pharmaceuticals at the time of the study and is currently an employee of Guide Pharmaceutical Consulting. Drs. Gordon, Rabinovich, and Knebel are working for Teva Pharmaceuticals. Drs. Savola, Marsteller, and Pastino were employed by Teva Pharmaceuticals at the time of the study. Drs. Kluge, Bronzel, and Voges are staff members of ABX-CRO Advanced Pharmaceutical Services. Drs. Geva and Hayden who are now employed by Prilenia Therapeutics Development were former staff members of Teva Pharmaceuticals at the time of the study. In September 2018, Teva Pharmaceutical sold and transferred its rights pertaining to pridopidine to Prilenia Therapeutics. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1
[18F] fluspidine baseline and post-drug PET of sigma-1 receptor (S1R) availability in healthy volunteers (HVs) and patients with Huntington disease (HD) and non-linear relationship between pridopidine dose (or plasma concentration) and S1R occupancy. [18F] fluspidine PET of S1R availability at baseline and post-drug in HVs and patients with HD. Almost complete S1R engagement/occupancy (VT) by pridopidine exemplified in one healthy volunteer (a) and one HD patient (c) is demonstrated within the whole brain at post-drug PET (90 mg pridopidine) as compared with baseline PET. For visualization purpose, parametric PET/MR images are shown. There is a sigmoidal curve relationship (b) between the pridopidine dose (left) or plasma concentration (Cavg2-4h; right, both on logarithmic scale) and S1R occupancy in HVs treated by a single dose of pridopidine ranging from 0.5 to 90 mg. The continuous line curve reflects fitting with a three-parameter model (Hill coefficient > 1) which was preferred, whereas the dotted line curve presents a two-parameter type model (Hill coefficient = 1)
Fig. 2
Fig. 2
One-tissue compartment model fits of 90 min [18F] fluspidine PET data at baseline and post-drug in healthy volunteers (HVs) and patients with Huntington disease (HD). One-tissue compartment model fits of 90 min [18F] fluspidine PET data at baseline (a, c; PET1) and acquired 2 h after oral administration of 90 mg pridopidine (b, d; PET2) are exemplified for the cerebellum, frontal cortex, striatum, and corpus callosum in one representative HV (a, b) and one patient with HD (c, d)
Fig. 3
Fig. 3
[18F] fallypride baseline and post-drug PET of dopamine D2/D3 receptor (D2/D3R) availability in healthy volunteers. Parametric PET/MR images of D2/D3R are demonstrated. There is no D2/D3R occupancy of pridopidine paradigmatically demonstrated in one healthy volunteer as assessed by [18F] fallypride PET at baseline and 2 h following single dose of 90 mg pridopidine
Fig. 4
Fig. 4
Simplified reference tissue model fits of 210 min [18F] fallypride PET data at baseline and post-drug in healthy volunteers (HVs). Simplified reference tissue model fits of 210 min [18F] fallypride PET data at baseline (a; PET1) and acquired 2 h after oral administration of 90 mg pridopidine (b; PET2) are exemplified for the striatum, thalamus, and midbrain using the cerebellum as reference region in one representative healthy volunteer (HV)

References

    1. Caron NS, Dorsey ER, Hayden MR. Therapeutic approaches to Huntington disease: from the bench to the clinic. Nat Rev Drug Discov. 2018;17:729–750. doi: 10.1038/nrd.2018.133.
    1. Mancuso R, Navarro X. Amyotrophic lateral sclerosis: current perspectives from basic research to the clinic. Prog Neurobiol. 2015;133:1–26. doi: 10.1016/j.pneurobio.2015.07.004.
    1. Dyhring T, Nielsen EØ, Sonesson C, Pettersson F, Karlsson J, Svensson P, et al. The dopaminergic stabilizers pridopidine (ACR16) and (-)-OSU6162 display dopamine D(2) receptor antagonism and fast receptor dissociation properties. Eur J Pharmacol. 2010;628:19–26. doi: 10.1016/j.ejphar.2009.11.025.
    1. Waters S, Tedroff J, Ponten H, Klamer D, Sonesson C, Waters N. Pridopidine: overview of pharmacology and rationale for its use in Huntington's disease. J Huntingtons Dis. 2018;7:1–16. doi: 10.3233/JHD-170267.
    1. Rung JP, Rung E, Helgeson L, Johansson AM, Svensson K, Carlsson A, et al. Effects of (-)-OSU6162 and ACR16 on motor activity in rats, indicating a unique mechanism of dopaminergic stabilization. J Neural Transm (Vienna) 2018;115:899–908. doi: 10.1007/s00702-008-0038-3.
    1. Johnston TH, Geva M, Steiner L, Orbach A, Papapetropoulos S, Savola JM, et al. Pridopidine, a clinic-ready compound, reduces 3,4-dihydroxyphenylalanine-induced dyskinesia in Parkinsonian macaques. Mov Disord. 2019;34:708–716. doi: 10.1002/mds.27565.
    1. Sahlholm K, Århem P, Fuxe K, Marcellino D. The dopamine stabilizers ACR16 and (−)-OSU6162 display nanomolar affinities at the σ-1 receptor. Mol Psychiatry. 2013;18:12–14. doi: 10.1038/mp.2012.3.
    1. Sahlholm K, Sijbesma JW, Maas B, Kwizera C, Marcellino D, Ramakrishnan NK, et al. Pridopidine selectively occupies sigma-1 rather than dopamine D2 receptors at behaviorally active doses. Psychopharmacology. 2015;232:3443–3453. doi: 10.1007/s00213-015-3997-8.
    1. Su TP, Hayashi T, Maurice T, Buch S, Ruoho AE. The sigma-1 receptor chaperone as an inter-organelle signaling modulator. Trends Pharmacol Sci. 2010;31:557–566. doi: 10.1016/j.tips.2010.08.007.
    1. Ruscher K, Wieloch T. The involvement of the sigma-1 receptor in neurodegeneration and neurorestoration. J Pharmacol Sci. 2015;127:30–35. doi: 10.1016/j.jphs.2014.11.011.
    1. Al-Saif A, Al-Mohanna F, Bohlega SA. Mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann Neurol. 2011;70:913–919. doi: 10.1002/ana.22534.
    1. Gregianin E, Pallafacchina G, Zanin S, Crippa V, Rusmini P, Poletti A, et al. Loss-of-function mutations in the SIGMAR1 gene cause distal hereditary motor neuropathy by impairing ER-mitochondria tethering and Ca2+ signalling. Hum Mol Genet. 2016;25:3741–3753. doi: 10.1093/hmg/ddw220.
    1. Squitieri F, Di Pardo A, Favellato M, Amico E, Maglione V, Frati L. Pridopidine, a dopamine stabilizer, improves motor performance and shows neuroprotective effects in Huntington disease R6/2 mouse model. J Cell Mol Med. 2015;19:2540–2548. doi: 10.1111/jcmm.12604.
    1. Geva M, Kusko R, Soares H, Fowler KD, Birnberg T, Barash S, et al. Pridopidine activates neuroprotective pathways impaired in Huntington disease. Hum Mol Genet. 2016;25:3975–3987. doi: 10.1093/hmg/ddw238.
    1. Francardo V, Geva M, Bez F, Denis Q, Steiner L, Hayden MR, et al. Pridopidine induces functional neurorestoration via the sigma-1 receptor in a mouse model of Parkinson's disease. Neurotherapeutics. 2019;16:465–479. doi: 10.1007/s13311-018-00699-9.
    1. Ionescu A, Gradus T, Altman T, Maimon R, Saraf Avraham N, et al. Targeting the sigma-1 receptor via pridopidine ameliorates central features of ALS pathology in a SOD1G93A model. Cell Death Dis. 2019;10:210. doi: 10.1038/s41419-019-1451-2.
    1. Ryskamp D, Wu J, Geva M, Kusko R, Grossman I, Hayden M, et al. The sigma-1 receptor mediates the beneficial effects of pridopidine in a mouse model of Huntington disease. Neurobiol Dis. 2017;97:46–59. doi: 10.1016/j.nbd.2016.10.006.
    1. Reilmann R, McGarry A, Grachev ID, Savola JM, Borowsky B, Eyal E, et al. European Huntington's disease network; Huntington study group investigators. Safety and efficacy of pridopidine in patients with Huntington's disease (PRIDE-HD): a phase 2, randomised, placebo-controlled, multicentre, dose-ranging study. Lancet Neurol. 2019;18:165–176. doi: 10.1016/S1474-4422(18)30391-0.
    1. Fischer S, Wiese C, Maestrup EG, Hiller A, Deuther-Conrad W, Scheunemann M, et al. Molecular imaging of σ receptors: synthesis and evaluation of the potent σ1 selective radioligand [18F]fluspidine. Eur J Nucl Med Mol Imaging. 2011;38:540–551. doi: 10.1007/s00259-010-1658-z.
    1. Brust P, Deuther-Conrad W, Becker G, Patt M, Donat CK, Stittsworth S, et al. Distinctive in vivo kinetics of the new sigma-1 receptor ligands (R)-(+)- and (S)-(-)-18F-Fluspidine in porcine brain. J Nucl Med. 2014;55:1730–1736. doi: 10.2967/jnumed.114.137562.
    1. Baum E, Cai Z, Bois F, Holden D, Lin SF, Lara-Jaime T, et al. PET imaging evaluation of four σ1 radiotracers in nonhuman primates. J Nucl Med. 2017;58:982–988. doi: 10.2967/jnumed.116.188052.
    1. Kranz M, Sattler B, Wüst N, Deuther-Conrad W, Patt M, Meyer PM, et al. Evaluation of the enantiomer specific biokinetics and radiation doses of [(18)F]fluspidine-a new tracer in clinical translation for imaging of σ1 receptors. Molecules. 2016;21:pii: E1164. doi: 10.3390/molecules21091164.
    1. Becker GA, Meyer PM, Patt M, Hesse S, Luthardt J, Patt T, et al. Kinetic modeling of the new sigma-1 receptor ligand (-)-[18F] Fluspidine in the human brain. Nuklearmedizin. 2018;57:02(A7). 10.1055/s-008-39473.
    1. Mukherjee J, Christian BT, Dunigan KA, Shi B, Narayanan TK, Satter M, et al. Brain imaging of 18F-fallypride in normal volunteers: blood analysis, distribution, test-retest studies, and preliminary assessment of sensitivity to aging effects on dopamine D-2/D-3 receptors. Synapse. 2002;46:170–188. doi: 10.1002/syn.10128.
    1. Gründer G, Fellows C, Janouschek H, Veselinovic T, Boy C, Bröcheler A, et al. Brain and plasma pharmacokinetics of aripiprazole in patients with schizophrenia: an [18F] fallypride PET study. Am J Psychiatry. 2008;165:988–995. doi: 10.1176/appi.ajp.2008.07101574.
    1. Huntington Study Group Unified Huntington's disease rating scale: reliability and consistency. Mov Disord. 1996;11:136–142. doi: 10.1002/mds.870110204.
    1. Shannon KM. Pridopidine for the treatment of Huntington's disease. Expert Opin Investig Drugs. 2016;25:485–492. doi: 10.1517/13543784.2016.1153627.
    1. Maisonial-Besset A, Funke U, Wenzel B, Fischer S, Holl K, Wünsch B, et al. Automation of the radiosynthesis and purification procedures for [18F] Fluspidine preparation, a new radiotracer for clinical investigations in PET imaging of σ(1) receptors in brain. Appl Radiat Isot. 2014;84:1–7. doi: 10.1016/j.apradiso.2013.10.015.
    1. Piel M, Schmitt U, Bausbacher N, Buchholz HG, Gründer G, Hiemke C, et al. Evaluation of P-glycoprotein (abcb1a/b) modulation of [(18)F] fallypride in microPET imaging studies. Neuropharmacology. 2014;84:152–158. doi: 10.1016/j.neuropharm.2013.04.062.
    1. Lancaster JL, Woldorff MG, Parsons LM, Liotti M, Freitas CS, Rainey L, et al. Automated Talairach atlas labels for functional brain mapping. Hum Brain Mapp. 2000;10:120–131. doi: 10.1002/1097-0193(200007)10:3<120::AID-HBM30>;2-8.
    1. Tzourio-Mazoyer N, Landeau B, Papathanassiou D, Crivello F, Etard O, Delcroix N, et al. Automated anatomical labeling of activations in SPM using a macroscopic anatomical parcellation of the MNI MRI single-subject brain. Neuroimage. 2002;15:273–289. doi: 10.1006/nimg.2001.0978.
    1. Patt M, Becker GA, Grossmann U, Habermann B, Schildan A, Wilke S, et al. Evaluation of metabolism, plasma protein binding and other biological parameters after administration of (-)-[(18)F] Flubatine in humans. Nucl Med Biol. 2014;41:489–494. doi: 10.1016/j.nucmedbio.2014.03.018.
    1. Stober T, Wussow W, Schimrigk K. Bicaudate diameter - the most specific and simple CT parameter in the diagnosis of Huntington's disease. Neuroradiology. 1984;26:25–28. doi: 10.1007/BF00328198.
    1. 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;10:740–747. doi: 10.1038/jcbfm.1990.127.
    1. Lammertsma AA, Hume SP. Simplified reference tissue model for PET receptor studies. Neuroimage. 1996;4:153–158. doi: 10.1006/nimg.1996.0066.
    1. Cunningham VJ, Rabiner EA, Slifstein M, Laruelle M, Gunn RN. Measuring drug occupancy in the absence of a reference region: the Lassen plot re-visited. J Cereb Blood Flow Metab. 2010;30:46–50. doi: 10.1038/jcbfm.2009.190.
    1. Kirby S, Brain P, Jones B. Fitting E (max) models to clinical trial dose-response data. Pharm Stat. 2011;10:143–149. doi: 10.1002/pst.432.
    1. Helldén A, Panagiotidis G, Johansson P, Waters N, Waters S, Tedroff J, et al. The dopaminergic stabilizer pridopidine is to a major extent N-depropylated by CYP2D6 in humans. Eur J Clin Pharmacol. 2012;68:1281–1286. doi: 10.1007/s00228-012-1248-z.
    1. Eddings CR, Arbez N, Akimov S, Geva M, Hayden MR, Ross CA. Pridopidine protects neurons from mutant-huntingtin toxicity via the sigma-1 receptor. Neurobiol Dis. 2019;129:118–129. doi: 10.1016/j.nbd.2019.05.009.
    1. Garcia-Miralles M, Geva M, Tan JY, Yusof NABM, Cha Y, Kusko R, et al. Early pridopidine treatment improves behavioral and transcriptional deficits in YAC128 Huntington disease mice. JCI Insight. 2017;2(23):pii: 95665. doi: 10.1172/jci.insight.95665.
    1. Chu UB, Ruoho AE. Biochemical pharmacology of the sigma-1 receptor. Mol Pharmacol. 2016;89:142–153. doi: 10.1124/mol.115.101170.
    1. Schmidt HR, Zheng S, Gurpinar E, Koehl A, Manglik A, Kruse AC. Crystal structure of the human σ1 receptor. Nature. 2016;532:527–530. doi: 10.1038/nature17391.

Source: PubMed

3
Tilaa